Synthetic lethal screen using RNA interference

Abstract
The invention provides a method for identifying one or more genes in a cell of a cell type which interact with, e.g., modulate the effect of, an agent, e.g., a drug. For example, an identified gene may confer resistance or sensitivity to a drug, i.e., reduces or enhances the effect of the drug. The invention also provides STK6 and TPX2 as a gene that exhibits synthetic lethal interactions with KSP encoding a kinesin-like motor protein, and methods and compositions for treatment of diseases, e.g., cancers, by modulating the expression of STK6 or TPX2 gene and/or the activity of STK6 or TPX2 gene product. The invention also provides genes involved in cellular response to DNA damage, and their therapeutic uses.
Description
1. FIELD OF THE INVENTION

The present invention relates to methods and compositions for carrying out interaction screening, e.g., lethal/synthetic lethal screening, using RNA interference. The invention also relates to genes exhibiting synthetic lethal interactions with KSP, a kinesin-like motor protein, and their therapeutic uses. The invention also relates to genes involved in cellular response to DNA damage, and their therapeutic uses.


2. BACKGROUND OF THE INVENTION

RNA interference (RNAi) is a potent method to suppress gene expression in mammalian cells, and has generated much excitement in the scientific community (Couzin, 2002, Science 298: 2296-2297; McManus et al., 2002, Nat. Rev. Genet. 3, 737-747; Hannon, G. J., 2002, Nature 418, 244-251; Paddison et al., 2002, Cancer Cell 2, 17-23). RNA interference is conserved throughout evolution, from C. elegans to humans, and is believed to function in protecting cells from invasion by RNA viruses. When a cell is infected by a dsRNA virus, the dsRNA is recognized and targeted for cleavage by an RNaseIII-type enzyme termed Dicer. The Dicer enzyme “dices” the RNA into short duplexes of 21 nt, termed siRNAs or short-interfering RNAs, composed of 19 nt of perfectly paired ribonucleotides with two unpaired nucleotides on the 3′ end of each strand. These short duplexes associate with a multiprotein complex termed RISC, and direct this complex to mRNA transcripts with sequence similarity to the siRNA. As a result, nucleases present in the RISC complex cleave the mRNA transcript, thereby abolishing expression of the gene product. In the case of viral infection, this mechanism would result in destruction of viral transcripts, thus preventing viral synthesis. Since the siRNAs are double-stranded, either strand has the potential to associate with RISC and direct silencing of transcripts with sequence similarity.


Specific gene silencing promises the potential to harness human genome data to elucidate gene function, identify drug targets, and develop more specific therapeutics. Many of these applications assume a high degree of specificity of siRNAs for their intended targets. Cross-hybridization with transcripts containing partial identity to the siRNA sequence may elicit phenotypes reflecting silencing of unintended transcripts in addition to the target gene. This could confound the identification of the gene implicated in the phenotype. Numerous reports in the literature purport the exquisite specificity of siRNAs, suggesting a requirement for near-perfect identity with the siRNA sequence (Elbashir et al., 2001. EMBO J. 20:6877-6888; Tuschl et al., 1999, Genes Dev. 13:3191-3197; Hutvagner et al., Sciencexpress 297:2056-2060). One recent report suggests that perfect sequence complementarity is required for siRNA-targeted transcript cleavage, while partial complementarity will lead to tranlational repression without transcript degradation, in the manner of microRNAs (Hutvagner et al., Sciencexpress 297:2056-2060).


The biological function of small regulatory RNAs, including siRNAs and mRNAs is not well understood. One prevailing question regards the mechanism by which the distinct silencing pathways of these two classes of regulatory RNA are determined. mRNAs are regulatory RNAs expressed from the genome, and are processed from precursor stem-loop structures to produce single-stranded nucleic acids that bind to sequences in the 3′ UTR of the target mRNA (Lee et al., 1993, Cell 75:843-854; Reinhart et al., 2000, Nature 403:901-906; Lee et al., 2001, Science 294:862-864; Lau et al., 2001, Science 294:858-862; Hutvagner et al., 2001, Science 293:834-838). mRNAs bind to transcript sequences with only partial complementarity (Zeng et al., 2002, Molec. Cell 9:1327-1333) and repress translation without affecting steady-state RNA levels (Lee et al., 1993, Cell 75:843-854; Wightman et al., 1993, Cell 75:855-862). Both mRNAs and siRNAs are processed by Dicer and associate with components of the RNA-induced silencing complex (Hutvagner et al., 2001, Science 293:834-838; Grishok et al., 2001, Cell 106: 23-34; Ketting et al., 2001, Genes Dev. 15:2654-2659; Williams et al., 2002, Proc. Natl. Acad. Sci. USA 99:6889-6894; Hammond et al., 2001, Science 293:1146-1150; Mourlatos et al., 2002, Genes Dev. 16:720-728). A recent report (Hutvagner et al., 2002, Sciencexpress 297:2056-2060) hypothesizes that gene regulation through the mRNA pathway versus the siRNA pathway is determined solely by the degree of complementarity to the target transcript. It is speculated that siRNAs with only partial identity to the mRNA target will function in translational repression, similar to an mRNA, rather than triggering RNA degradation.


It has also been shown that siRNA and shRNA can be used to silence genes in vivo. The ability to utilize siRNA and shRNA for gene silencing in vivo has the potential to enable selection and development of siRNAs for therapeutic use. A recent report highlights the potential therapeutic application of siRNAs. Fas-mediated apoptosis is implicated in a broad spectrum of liver diseases, where lives could be saved by inhibiting apoptotic death of hepatocytes. Song (Song et al. 2003, Nat. Medicine 9, 347-351) injected mice intravenously with siRNA targeted to the Fas receptor. The Fas gene was silenced in mouse hepatocytes at the mRNA and protein levels, prevented apoptosis, and protected the mice from hepatitis-induced liver damage. Thus, silencing Fas expression holds therapeutic promise to prevent liver injury by protecting hepatocytes from cytotoxicity. As another example, injected mice intraperitoneally with siRNA targeting TNF-a. Lipopolysaccharide-induced TNF-a gene expression was inhibited, and these mice were protected from sepsis. Collectively, these results suggest that siRNAs can function in vivo, and may hold potential as therapeutic drugs (Sorensen et al., 2003, J. Mol. Biol. 327, 761-766).


Martinez et al. reported that RNA interference can be used to selectively target oncogenic mutations (Martinez et al., 2002, Proc. Natl. Acad. Sci. USA 99:14849-14854). In this report, an siRNA that targets the region of the R248W mutant of p53 containing the point mutation was shown to silence the expression of the mutant p53 but not the wild-type p53.


Wilda et al. reported that an siRNA targeting the M-BCR/ABL fusion mRNA can be used to deplete the M-BCR/ABL mRNA and the M-BRC/ABL oncoprotein in leukemic cells (Wilda et al., 2002, Oncogene 21:5716-5724). However, the report also showed that applying the siRNA in combination with Imatinib, a small-molecule ABL kinase tyrosine inhibitor, to leukemic cells did not further increase in the induction of apoptosis.


U.S. Pat. No. 6,506,559 discloses a RNA interference process for inhibiting expression of a target gene in a cell. The process comprises introducing partially or fully doubled-stranded RNA having a sequence in the duplex region that is identical to a sequence in the target gene into the cell or into the extracellular environment. RNA sequences with insertions, deletions, and single point mutations relative to the target sequence are also found as effective for expression inhibition.


U.S. Patent Application Publication No. U.S. 2002/0086356 discloses RNA interference in a Drosophila in vitro system using RNA segments 21-23 nucleotides (nt) in length. The patent application publication teaches that when these 21-23 nt fragments are purified and added back to Drosophila extracts, they mediate sequence-specific RNA interference in the absence of long dsRNA. The patent application publication also teaches that chemically synthesized oligonucleotides of the same or similar nature can also be used to target specific mRNAs for degradation in mammalian cells.


PCT publication WO 02/44321 discloses that double-stranded RNA (dsRNA) 19-23 nt in length induces sequence-specific post-transcriptional gene silencing in a Drosophila in vitro system. The PCT publication teaches that short interfering RNAs (siRNAs) generated by an RNase III-like processing reaction from long dsRNA or chemically synthesized siRNA duplexes with overhanging 3′ ends mediate efficient target RNA cleavage in the lysate, and the cleavage site is located near the center of the region spanned by the guiding siRNA. The PCT publication also provides evidence that the direction of dsRNA processing determines whether sense or antisense target RNA can be cleaved by the produced siRNP complex.


U.S. Patent Application Publication No. U.S. 2002/016216 discloses a method for attenuating expression of a target gene in cultured cells by introducing double stranded RNA (dsRNA) that comprises a nucleotide sequence that hybridizes under stringent conditions to a nucleotide sequence of the target gene into the cells in an amount sufficient to attenuate expression of the target gene.


PCT publication WO 03/006477 discloses engineered RNA precursors that when expressed in a cell are processed by the cell to produce targeted small interfering RNAs (siRNAs) that selectively silence targeted genes (by cleaving specific mRNAs) using the cell's own RNA interference (RNAi) pathway. The PCT publication teaches that by introducing nucleic acid molecules that encode these engineered RNA precursors into cells in vivo with appropriate regulatory sequences, expression of the engineered RNA precursors can be selectively controlled both temporally and spatially, i.e., at particular times and/or in particular tissues, organs, or cells.


Discussion or citation of a reference herein shall not be construed as an admission that such reference is prior art to the present invention.


3. SUMMARY OF THE INVENTION

The invention provides methods and compositions for identifying interactions, e.g., lethal/synthetic lethal interactions, between a gene or its product and an agent, e.g., a drug, and/or another gene or its product, using RNA interference. The invention also provides methods and compositions for treating cancer utilizing the synthetic lethal interaction between STK6 kinase or TPX2 and kinesin-like motor protein KSP inhibitors. The invention also provides genes involved in cellular response to DNA damage, and their therapeutic uses.


In one aspect, the invention provides a method for identifying a gene whose product modulates the effect of an agent on a cell of a cell type. The method comprises (a) contacting a plurality of groups of one or more cells of said cell type with said agent, wherein each said group of one or more cells comprises one or more different small interfering RNAs (siRNAs) from among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different genes in cells of said cell type; (b) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes; and (c) identifying a gene as said gene whose product modulates the effect of said agent on a cell of said cell type if the effect of said agent on said group of one or more cells comprising said one or more different siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes. In one embodiment, each said group of cells comprising one or more of said plurality of siRNAs is obtained by transfection with said one or more siRNAs prior to said step of contacting. In one embodiment, the contacting step (a) is carried out separately for each said groups of one or more cells.


In a specific embodiment, the invention provides a method for identifying a gene whose product modulates the effect of an agent on a cell of a cell type, said method comprising (a) transfacting each of a plurality of groups of one or more cells of said cell type with a composition comprising one or more different small interfering RNAs (siRNAs) from among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different genes in cells of said cell type; (b) contacting each of said plurality of groups of one or more cells with said agent; (c) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which is not transfected with an siRNA targeting any one of said different genes; and (d) identifying a gene as said gene whose product modulates the effect of said agent on a cell of said cell type if the effect of said agent on said group of one or more cells comprising said one or more different siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes.


The effect of said agent on each said group of one or more cells comprising said one or more different siRNAs can be enhanced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes. Alternatively, the effect of said agent on said group of one or more cells comprising said one or more different siRNAs can be reduced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes.


Preferably, the agent acts on a gene other than any one of said different genes targeted by said plurality of siRNAs, or a protein encoded thereof. Preferably, the plurality of siRNAs comprises at least k different siRNAs targeting at least one of said different genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10. More preferably, the plurality of siRNAs comprises at least k different siRNAs targeting each of at least 2 different genes of said different genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10. Still more preferably, the plurality of siRNAs comprises at least k different siRNAs targeting each of said different genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.


Preferably, the one or more different siRNAs for at least one, at least two, or each of of the plurality of different genes comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting a same target gene. In a preferred embodiment, the total siRNA concentration of the one or more siRNAs is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the one or more siRNAs is an optimal concentration for silencing the intended target gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the one or more siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the one or more siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the one or more siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more siRNAs. In another preferred embodiment, none of the siRNAs in the one or more siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more siRNAs. In a preferred embodiment, the composition of the one or more siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the one or more siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA in the one or more siRNAs has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA in the one or more siRNAs has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA in the one or more siRNAs has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the target gene.


In one embodiment, said cell type is a cancer cell type. In another embodiment, said effect is growth inhibitory effect. In a specific embodiment, said agent is a KSP inhibitor. In preferred embodiments, said different genes comprises at least 5, at least 10, at least 100, or at least 1,000 different genes. In one embodiment, said different genes are different endogenous genes.


In another aspect, the invention provides a method for identifying a gene which interacts with a primary target gene in a cell of a cell type. The method comprises (a) contacting a plurality of groups of one or more cells of said cell type with an agent, wherein said agent modulates the expression of said primary target gene and/or the activity of a protein encoded by said primary target gene, and wherein each said group of cells comprises one or more different siRNAs among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different secondary genes in said cell; (b) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes; and (c) identifying a gene as said gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said group of one or more cells comprising one or more siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes. In one embodiment, each said group of cells comprising one or more of said plurality of siRNAs is obtained by transfection with said one or more siRNA prior to said step of contacting.


In a specific embodiment, the invention provides method for identifying a gene which interacts with a primary target gene in a cell of a cell type, said method comprising (a) transfacting each of a plurality of groups of one or more cells of said cell type with a composition comprising one or more different small interfering RNAs (siRNAs) from among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different genes in cells of said cell type; (b) contacting said plurality of groups of one or more cells of said cell type with an agent, wherein said agent modulates the expression of said primary target gene and/or the activity of a protein encoded by said primary target gene; (c) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes; and (d) identifying a gene as said gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said group of one or more cells comprising one or more siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes.


In one embodiment, said agent comprises an siRNA targeting and silencing said primary target gene. In another embodiment, said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said primary target gene. In a preferred embodiment, each of said different siRNAs targeting said primary target gene. In a preferred embodiment, the total siRNA concentration of said different siRNAs is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of said different siRNAs is an optimal concentration for silencing the primary target gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the different siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the different siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the different siRNAs. In another preferred embodiment, none of the siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the different siRNAs. In a preferred embodiment, the composition of the different siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the different siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA has a concentration that causes less than 30%, 20%, 0.10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while all of the siRNAs together causes at least 80% or 90% of silencing of the target gene. In still another embodiment, said agent comprises an inhibitor of a protein encoded by said primary target gene.


The effect of said agent on said group of one or more cells can be enhanced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes. Alternatively, the effect of said agent on said group of one or more cells can be reduced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes.


Preferably, the plurality of siRNAs comprises at least k different siRNAs targeting at least one of said different secondary genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10. More preferably, the plurality of siRNAs comprises at least k different siRNAs targeting each of at least 2 different genes of said different secondary genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10. Still more preferably, the plurality of siRNAs comprises at least k different siRNAs targeting each of said different secondary genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.


Preferably, the one or more different siRNAs for at least one, at least two, or each of of the plurality of different genes comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting a same target gene. In a preferred embodiment, the total siRNA concentration of the one or more siRNAs targeting a same gene is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the one or more siRNAs is an optimal concentration for silencing the intended target gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the one or more siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the one or more siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the one or more siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more siRNAs. In another preferred embodiment, none of the siRNAs in the one or more siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more siRNAs. In a preferred embodiment, the composition of the one or more siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the one or more siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA in the one or more siRNAs has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA in the one or more siRNAs has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA in the one or more siRNAs has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the target gene.


In one embodiment, each said group of one or more cells is obtained by transfection with said one or more different siRNAs prior to said step of contacting. In another embodiment, the primary target is KSP. In preferred embodiments, said different secondary genes comprises at least 5, at least 10, at least 100, at least 1,000, at least 5,000 different genes. In one embodiment, said different secondary genes are different endogenous genes. In one embodiment, said cell type is a cancer cell type.


In still another aspect, the invention provides a method for treating a mammal having a cancer, comprising administering to said mammal a therapeutically sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, wherein said mammal is subject to a therapy comprising administering to said mammal a therapeutically sufficient amount of a KSP inhibitor. The invention also provides a method for treating a mammal having a cancer, comprising administering to said mammal i) a therapeutically sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, and ii) a therapeutically sufficient amount of a KSP inhibitor. In one embodiment, said agent reduces the expression of said STK6 or TPX2 gene in cells of said cancer. In a preferred embodiment, said agent comprises an siRNA targeting said STK6 or TPX2 gene. In another embodiment, the mammal is a human, and the siRNA can be selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6. In another preferred embodiment, said agent comprises an siRNA targeting said TPX2 gene. In another embodiment, mammal is a human, and the siRNA can be selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.


In another embodiment, the invention provides a method for treating a mammal having a cancer, comprising administering to said mammal i) a therapeutically sufficient amount of a first agent, said first agent regulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, and ii) a therapeutically sufficient amount of a second agent, said second agent regulating the expression of a KSP gene and/or activity of a protein encoded by said KSP gene. In a preferred embodiment, the first agent is an siRNA targeting said STK6 or TPX2 gene, and said second agent comprises an siRNA targeting said KSP gene. In another preferred embodiment, said mammal is a human, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6. In another preferred embodiment, said agent comprises an siRNA targeting said TPX2 gene. In another embodiment, mammal is a human, and the siRNA can be selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.


In still another embodiment, the invention provides a method for evaluating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said method comprising determining an expression level of a STK6 or TPX2 gene in said cell, wherein said expression level above a predetermined threshold level indicates that said cell is resistant to the growth inhibitory effect of said KSP inhibitor. In a preferred embodiment, the expression level of said STK6 or TPX2 gene is determined by a method comprising measuring the expression level of said STK6 or TPX2 gene using one or more polynucleotide probes, each of said one or more polynucleotide probes comprising a nucleotide sequence in said STK6 or TPX2 gene. Said one or more polynucleotide probes can be polynucleotide probes on a microarray.


In still another embodiment, the invention provides a method for evaluating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said method comprising determining a level of abundance of a protein encoded by a STK6 or TPX2 gene in said cell, wherein said level of abundance of said protein above a predetermined threshold level indicates that said cell is resistant to the growth inhibitory effect of said KSP inhibitor. The invention also provides a method for evaluating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said method comprising determining a level of activity of a protein encoded by a STK6 or TPX2 gene in said cell, wherein said activity level above a predetermined threshold level indicates that said cell is resistant to the growth inhibitory effect of said KSP inhibitor. In a preferred embodiment, said cell is a human cell.


In still another embodiment, the invention provides a method for regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, comprising contacting said cell with a sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene. The invention also provides a method for regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor in a mammal, comprising administering to said mammal a therapeutically sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene. The invention further provides a method for regulating growth of a cell, comprising contacting said cell with i) a sufficient amount of an agent that regulates the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene; and ii) a sufficient amount of a KSP inhibitor. Preferably, the agent reduces the expression of said STK6 or TPX2 gene in said cell. In a preferred embodiment, said agent comprises an siRNA targeting said STK 6 gene. In another preferred embodiment, said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6. In another preferred embodiment, said agent comprises an siRNA targeting said TPX2 gene. In another embodiment, cell is a human cell, and the siRNA can be selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.


In still another embodiment, the invention provides a method of identifying an agent that is capable of regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, wherein said agent is capable of modulating the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene, said method comprising comparing inhibitory effect of said KSP inhibitor on cells expressing said STK6 or TPX2 gene in the presence of said agent with inhibitory effect of said KSP inhibitor on cells expressing said STK6 or TPX2 gene in the absence of said agent, wherein a difference in said inhibitory effect of said KSP inhibitor identifies said agent as capable of regulating resistance of said cell to the growth inhibitory effect of said KSP inhibitor.


The invention also provides a method of identifying an agent that is capable of regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, wherein said agent is capable of modulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, said method comprising: (a) contacting a first cell expressing said STK6 or TPX2 gene with said KSP inhibitor in the presence of said agent and measuring a first growth inhibitory effect; (b) contacting a second cell expressing said STK6 or TPX2 gene with said KSP inhibitor in the absence of said agent and measuring a second growth inhibitory effect; and (c) comparing said first and second inhibitory effects measured in said step (a) and (b), wherein a difference between said first and second inhibitory effects identifies said agent as capable of regulating resistance of a cell to the growth inhibitory effect of said KSP inhibitor. In a preferred embodiment, said agent is a molecule which reduces expression of said STK6 or TPX2 gene. In another preferred embodiment, said agent comprises an siRNA targeting said STK 6 gene. In still another preferred embodiment, said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6. In another preferred embodiment, said agent comprises an siRNA targeting said TPX2 gene. In another embodiment, the cell is a human cell, and the siRNA can be selected from the group consisting of siRNAs described by SEQ ID NO: SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.


In still another aspect, the invention provides a cell comprising one or more different small interfering RNAs (siRNAs) targeting a STK6 or TPX2 gene in said cell. The cell can be a human cell. The cell can also be a murine cell. In one embodiment, said cell is a human cell, and each of said one or more different siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6. In another embodiment, the cell is a human cell, and the siRNA can be selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239. In one embodiment, said cell is produced by transfection using a composition of said one or more different siRNAs, wherein the total siRNA concentration of said composition is an optimal concentration for silencing said STK6 or TPX2 gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%. In one embodiment, the concentration of each said different siRNA is about the same. In one embodiment, the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%. In another embodiment, none of the siRNAs in said composition has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs. In another embodiment, at least one siRNA in said composition has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs. In another embodiment, the number of different siRNAs and the concentration of each siRNA in said composition is chosen such that said composition causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.


In still another aspect, the invention provides a microarray for diagnosing resistance of a cell to the growth inhibitory effect of a KSP inhibitor. The microarray comprising one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in a STK6 or TPX2 gene.


In still another aspect, the invention provides kit for diagnosis of resistance of a cell to the growth inhibitory effect of a KSP inhibitor. The kit comprises in one or more containers one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in a STK6 or TPX2 gene. The invention also provides a kit for screening for agents which regulate resistance of a cell to the growth inhibitory effect of a KSP inhibitor. The kit comprises in one or more containers (i) a cell comprising one or more different small interfering RNAs (siRNAs) targeting a STK6 or TPX2 gene in said cell; and (ii) a KSP inhibitor. In still another aspect, the invention provides a kit for treating a mammal having a cancer, which comprises in one or more containers (i) a sufficient amount of an agent that regulates the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene; and (ii) a KSP inhibitor.


In the invention, the KSP inhibitor can be (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine as described in PCT application PCT/US03/18482, filed Jun. 12, 2003.


The invention also provides a method for identifying a gene which interacts with a primary target gene in a cell of a cell type. The method comprises (a) contacting one or more cells of said cell type with an agent, wherein said agent modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene, and wherein said one or more cells express a first small interference RNA (siRNA) targeting said primary target gene; (b) comparing the effect of said agent on said one or more cells of said clone to the effect of said agent on a cell of said cell type not expressing said first siRNA; and (c) identifying said secondary target gene as a gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said one or more cells expressing said first siRNA is different as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA.


In a specific embodiment, the method comprises (a) generating a clone of cells of said cell type which express a first small interference RNA (siRNA) targeting said primary target gene; (b) contacting one or more cells of said clone with an agent, wherein said agent modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; (c) comparing the effect of said agent on said one or more cells of said clone to the effect of said agent on a cell of said cell type not expressing said first siRNA; and (d) identifying said secondary target gene as a gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said one or more cells expressing said first siRNA is different as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA.


In some embodiments, the effect of said agent on said one or more cells expressing said first siRNA is enhanced as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA. In some other embodiments, the effect of said agent on said one or more cells expressing said first siRNA is reduced as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA. In one embodiment, said agent is an inhibitor of said secondary target gene. The effect of said agent can be a change in the sensitivity of cells of said cell type to a drug, e.g., to a DNA damaging agent, e.g., a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, an anti-metabolite, or ionizing radiation.


In another embodiment, said agent comprises one or more second siRNAs targeting and silencing said secondary target gene. Preferably, said one or more second siRNAs comprises at least k different siRNAs, e.g., at least 2, 3, 4, 5, 6 and 10 different siRNAs. In a preferred embodiment, the total siRNA concentration of the one or more second siRNAs is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the one or more second siRNAs is an optimal concentration for silencing the intended secondary target gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the one or more second siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the one or more second siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the one or more second siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more second siRNAs. In another preferred embodiment, none of the siRNAs in the one or more second siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more second siRNAs. In a preferred embodiment, the composition of the one or more second siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the one or more second siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA in the one or more second siRNAs has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA in the one or more second siRNAs has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA in the one or more second siRNAs has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the secondary target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the secondary target gene.


In one embodiment, said cell type is a cancer cell type. In another embodiment, said primary target gene is p53.


In a preferred embodiment, steps (b)-(d) of the method are repeated for each of a plurality of different secondary target genes. The plurality of secondary target genes can comprise at least 5, 10, 100, 1,000, and 5,000 different genes.


The invention also provides a method for treating a mammal having a cancer. The method comprises administering to said mammal a therapeutically sufficient amount of an agent, said agent regulating the expression of a gene and/or activity of a protein encoded by said gene, wherein said mammal is subject to a therapy comprising administering to said mammal a therapeutically sufficient amount of a composition comprising one or more DNA damaging agents. In one embodiment, the invention provides a method for treating a mammal having a cancer, comprising administering to said mammal i) a therapeutically sufficient amount of an agent, said agent regulating the expression of a gene and/or activity of a protein encoded by said gene, and ii) a therapeutically sufficient amount of a composition comprising one or more DNA damaging agents.


Preferably, said agent reduces the expression of said gene in cells of said cancer. In a preferred embodiment, said agent comprises an siRNA targeting said gene. In specific embodiment, said gene is EPHB3, WEE1, ELK1, STK6, CHEK1 or BRCA2. The agent can also be an agent that enhances the expression of said gene in cells of said cancer. The one or more DNA damaging agents can comprise a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, an anti-metabolite, or ionizing radiation.


The invention also provides a method for evaluating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent. The method comprises determining a transcript level of a gene in said cell, wherein said transcript level below a predetermined threshold level indicates that said cell is sensitive to the growth inhibitory effect of said DNA damaging agent. The DNA damaging agent can be a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, an anti-metabolite, or ionizing radiation. In a preferred embodiment, said gene is EPHB3, WEE1, ELK1, STK6, CHEK1 or BRCA2. In a preferred embodiment, said transcript level of said gene is determined by a method comprising measuring the transcript level of said gene using one or more polynucleotide probes, each of said one or more polynucleotide probes comprising a nucleotide sequence in said gene. In one embodiment, said one or more polynucleotide probes are polynucleotide probes on a microarray.


In another embodiment, the invention provides a method for evaluating sensitivity of a cell, e.g., a human cell, to the growth inhibitory effect of a DNA damaging agent. The method comprises determining a level of abundance of a protein encoded by a gene in said cell, wherein said level of abundance of said protein below a predetermined threshold level indicates that said cell is sensitive to the growth inhibitory effect of said DNA damaging agent. The invention also provides a method for evaluating sensitivity of a cell, e.g., a human cell, to the growth inhibitory effect of a DNA damaging agent, said method comprising determining a level of activity of a protein encoded by a gene in said cell, wherein said activity level above a predetermined threshold level indicates that said cell is sensitive to the growth inhibitory effect of said DNA damaging agent. The DNA damaging agent can be a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, an anti-metabolite, or ionizing radiation. In a preferred embodiment, said gene is EPHB3, WEE1, ELK1, STK6, CHEK1 or BRCA2.


The invention also provides a method for regulating sensitivity of a cell to DNA damage. The method comprises contacting said cell with a sufficient amount of an agent, said agent regulating the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene. The invention also provides a method for regulating growth of a cell, comprising contacting said cell with i) a sufficient amount of an agent that regulates the expression of a gene selected from the group consisting of EPHB33, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene; and ii) a sufficient amount of a DNA damaging agent. The DNA damaging agent can be a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, an anti-metabolite, or ionizing radiation.


In one embodiment, said agent reduces the expression of said gene in said cell. In a preferred embodiment, said agent comprises an siRNA targeting said gene. In another preferred embodiment, said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said gene. In a preferred embodiment, the total siRNA concentration of the different siRNAs targeting said is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the different siRNAs targeting said gene is an optimal concentration for silencing the gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the different siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the different siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the different siRNAs. In another preferred embodiment, none of the siRNAs in the different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the different siRNAs. In a preferred embodiment, the composition of the different siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the different siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA in the different siRNAs has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA in the different siRNAs has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the target gene.


The invention also provides a method of identifying an agent that is capable of regulating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, wherein said agent is capable of modulating the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene, said method comprising comparing inhibitory effect of said DNA damaging agent on cells expressing said gene in the presence of said agent with inhibitory effect of said DNA damaging agent on cells expressing said gene in the absence of said agent, wherein a difference in said inhibitory effect of said DNA damaging agent identifies said agent as capable of regulating sensitivity of said cell to the growth inhibitory effect of said DNA damaging agent. In one embodiment, the invention provides a method of identifying an agent that is capable of regulating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, wherein said agent is capable of modulating the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or activity of a protein encoded by said gene, said method comprising: (a) contacting a first cell expressing said gene with said DNA damaging agent in the presence of said agent and measuring a first growth inhibitory effect; (b) contacting a second cell expressing said gene with said DNA damaging agent in the absence of said agent and measuring a second growth inhibitory effect; and (c) comparing said first and second inhibitory effects measured in said step (a) and (b), wherein a difference between said first and second inhibitory effects identifies said agent as capable of regulating sensitivity of a cell to the growth inhibitory effect of said DNA damaging agent.


Preferably, said cell expresses an siRNA targeting a primary target gene. In one embodiment, said primary target gene is p53.


In a preferred embodiment, said agent is a molecule that reduces expression of said gene. In one embodiment, said agent comprises an siRNA targeting said gene. In another embodiment, said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said gene. In a preferred embodiment, the total siRNA concentration of the different siRNAs targeting said is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the different siRNAs targeting said gene is an optimal concentration for silencing the gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the different siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the different siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the different siRNAs. In another preferred embodiment, none of the siRNAs in the different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the different siRNAs. In a preferred embodiment, the composition of the different siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the different siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA in the different siRNAs has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA in the different siRNAs has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the target gene.


In the method, said DNA damaging agent can be a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, an anti-metabolite, or an ionizing radiation.


The invention also provides a cell comprising one or more different small interfering RNAs (siRNAs) targeting a gene selected from the group consisting of EPHB3, WEE1, ELK1, BRCA1, BRCA2, BARD1, and RAD51 in said cell. In one embodiment, said one or more different siRNAs comprises 2, 3, 4, 5, 6, or 10 different siRNAs. In a preferred embodiment, the total siRNA concentration of the one or more different siRNAs is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the one or more siRNAs is an optimal concentration for silencing the intended target gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the one or more siRNAs comprise each siRNA in equal proportion. In another preferred embodiment, the one or more siRNAs comprise each siRNA in proportions different from each other by less than 5%, 10%, 20% or 50%. In a preferred embodiment, at least one of the one or more siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more siRNAs. In another preferred embodiment, none of the siRNAs in the one or more siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration of the one or more siRNAs. In a preferred embodiment, the composition of the one or more siRNAs, including the number of different siRNAs and the concentration of each siRNA, is chosen such that the one or more siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In other embodiments, each siRNA in the one or more siRNAs has a concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, at least one siRNA in the one or more siRNAs has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA in the one or more siRNAs has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the target gene.


The invention also provides a microarray for diagnosing sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent. The microarray comprises one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in one or more genes selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.


The invention also provides a kit for diagnosis of sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent. The kit comprises in one or more containers one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.


The invention also provides a kit for screening for agents which regulate sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent. The kit comprises in one or more containers (i) a cell comprising one or more different small interfering RNAs (siRNAs) targeting a gene selected from the group consisting of EPHB3, WEE1, ELK1, BRCA1, BRCA2, BARD1, and RAD51 in said cell; and (ii) said DNA damaging agent.


The invention also provides a kit for treating a mammal having a cancer, comprising in one or more containers (i) a sufficient amount of an agent that regulates the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene; and (ii) a DNA damaging agent.


In the kit of the invention, the DNA damaging agent can be a topoisomerase I inhibitor, e.g., camptothecin, a topoisomerase II inhibitor, e.g., doxorubicin, a DNA binding agent, e.g., cisplatin, or an anti-metabolite.


The invention also provides a method of evaluating the responsiveness of cells of a cell type to treatment of a drug, comprising (a) contacting one or more cells of said cell type with said drug, wherein said one or more cells express a first small interference RNA (siRNA) targeting a primary target gene, and wherein said one or more cells are subject to treatment of a composition that modulates the expression of one or more secondary target genes and/or the activity of one or more proteins encoded respectively by said one or more secondary target genes; (b) contacting one or more cells of said cell type with said drug, wherein said one or more cells do not express a small interference RNA (siRNA) targeting said primary target gene, and wherein said one or more cells are subject to treatment of said agent that modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; and (c) comparing the effect of said drug on said one or more cells measured in step (a) to the effect of said drug on said one or more cells measured in step (b), thereby evaluating the responsiveness of said cells to treatment of said drug. In one embodiment, the method further comprises a step (d) repeating steps (a)-(b) for each of a plurality of different secondary target genes.


In a specific embodiment, the invention provides a method for evaluating the responsiveness of cells of a cell type to treatment of a drug, said method comprising (a) generating a clone of cells of said cell type which express a first small interference RNA (siRNA) targeting a primary target gene; (b) contacting one or more cells of said clone which express said first siRNA with said drug, wherein said one or more cells are subject to treatment of an agent that modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; (c) contacting one or more cells of said cell type which do not express a small interference RNA (siRNA) targeting said primary target gene with said drug, wherein said one or more cells are subject to treatment of said agent that modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; and (d) comparing the effect of said drug on said one or more cells measured in step (b) to the effect of said drug on said one or more cells measured in step (c), thereby evaluating the responsiveness of said cells to treatment of said drug. In one embodiment, the method further comprises a step (e) repeating steps (b)-(d) for each of a plurality of different secondary target genes.


In one embodiment, the effect of said drug on said one or more cells expressing said first siRNA is enhanced as compared to the effect of said drug on a cell of said cell type not expressing said first siRNA. In another embodiment, the effect of said drug on said one or more cells expressing said first siRNA is reduced as compared to the effect of said drug on a cell of said cell type not expressing said first siRNA.


In one embodiment, said composition comprises one or more inhibitors of said one or more secondary target gene. In a preferred embodiment, said composition comprises one or more second siRNAs targeting and silencing said one or more secondary target gene.


In one embodiment, said one or more second siRNAs comprises at least k different siRNAs, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10. In one embodiment, the total siRNA concentration of said at least k different siRNAs in said agent is an optimal concentration for silencing said secondary target gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%. In another embodiment, the concentration of each said at least k different siRNA is about the same. In another embodiment, the respective concentrations of said at least k different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%. In still another embodiment, none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs. In still another embodiment, at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said at least k different siRNAs. In still another embodiment, the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.


In some embodiment, said cell type is a cancer cell type, and said primary target gene is p53. In preferred embodiment, said plurality of secondary target genes comprises at least the number of different genes selected from the group consisting of 5, 10, 100, 1,000, and 5,000 different genes.


In one embodiment, said drug is a DNA damaging agent, e.g., a DNA damaging agent selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation. In a specific embodiment, said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin.




4. BRIEF DESCRIPTION OF FIGURES


FIG. 1 shows correlation between mRNA silencing and growth inhibition phenotype for STK6. HeLa cells were transfected with six individual siRNAs to STK6. At 24 hrs post transfection, one set of cells was harvested for RNA isolation and determination of STK6 mRNA levels by TaqMan analysis using an Assay on Demand (Applied Biosciences). Another set of cells was incubated further (72 hrs total) and cellular growth was assessed in triplicate wells using an Alamar Blue assay. Values for mRNA levels (X axis) and cell growth (Y axis) for each were normalized to a mock transfected control. For TaqMan analysis, each data point represents a single RNA sample assayed in triplicate (and normalized to GUS); variation between replicates was generally <10%. For growth assay determinations, each data point represents the average of triplicate determinations that generally varied from the mean by <20%. The solid line represents an ideal 1:1 relationship between silencing and phenotype.



FIG. 2 shows synthetic lethal interactions between STK6 and KSP. HeLa cells were transfected with increasing concentrations of siRNA to luciferase (negative control) and STK6 (top panel) or PTEN (bottom panel) and tested for growth relative to control (luciferase-treated) in the three-day Alamar Blue assay. Where indicated, cells were also treated with 25 nM KSPi, (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine; the EC50 for HeLa cells assayed under these conditions was ˜80 nM. Shown are the mean±SD (error bars) of triplicate determinations.



FIG. 3 demonstrates that stable expression of a TP53 shRNA effectively silences the target gene. HCT116 cells were transfected with a TP53-targeting shRNA plasmid (pRS-p53). Shown are the TP53 mRNA levels in wild type (WT) cells and in two independent clones (A5 and A11) of cells stably transfected with pRS-p53. TP53 mRNA levels were silenced >95% in clones A5 and A11 (Middle bars). Transient introduction of the pRS-p53 into HCT116 cells achieves ˜80% silencing 24 hr post transfection (Right bar).



FIG. 4 shows maintenance of mRNA silencing by stable shRNA expression following siRNA supertransfection. (A) pRS-p53 does not affect CHEK1 silencing by siRNAs and vice versa. A pool of three siRNAs targeting CHEK1 was transiently transfected into WT and pRS-p53 stably transfected HCT116 cells (clone A11). CHEK1 and TP53 mRNA levels were measured by Taqman analysis (left and right panels, respectively). (B) Supertransfected KNSL1 siRNAs do not competitively inhibit silencing by pRS-STK6. STK6 and KNSL1 siRNAs were transiently co-transfected into WT SW480 cells and KNSL1 siRNAs were supertransfected into pRS-STK6 stably transfected SW480 cells. STK6 mRNA levels were measured by Taqman analysis. For the left set of bars, STK6 siRNA (10 nM) was used alone or together with one of three different individual KNSL1 siRNAs (10 nM each). The KNSL1 siRNAs variably inhibit silencing by STK6 siRNAs. For the right two sets of bars, KNSL1 siRNAs were used as competitors at 10 or 100 nM against the stably expressed STK6 shRNA.



FIG. 5 demonstrates that siRNA library screens in the absence of DNA damage show good correlation between cells with and without a shRNA targeting p53. (x axis) pRS (vector alone) cells were supertransfected with pools of three siRNAs each targeting one of 800 genes and tested for growth related phenotypes; (y axis) pRS-p53 cells assayed in the same manner. The tight correlation between the two sets of data indicates that the performance of the siRNA pools is likely not affected by the presence of the shRNA suggesting that the shRNA does not compete with the siRNAs.



FIG. 6 shows that CHEK1 silencing decreases G2 checkpoint arrest in pRS-p53 cells. A549 cells stably transfected with vector only (pRS) or pRS-p53 cells were supertransfected with control (luc, luciferase) siRNA or with a pool of three siRNAs to CHEK1. Doxorubicin (200 ng/ml) was added 24 hr post-transfection and cell cycle profiles were analyzed 48 hr after doxorubicin addition. TP53 mRNA levels in pRS-p53 cells was reduced ˜90% compared with pRS cells.



FIG. 7 illustrates the identification of genes that sensitize to Cisplatin. HeLa cells grown in 384 well plates were transfected with siRNA pools representing ˜800 human genes (3 siRNAs/gene, total siRNA concentration 100 nM). Four hours post-transfection, cells were treated with either medium alone (or plus vehicle) (− drug) or medium plus an EC10 concentration of Cisplatin (Cis, + drug). Cell growth was then measured 72 hrs post-transfection using an Alamar Blue assay and is expressed as % growth measured in wells transfected with luciferase siRNA. Each point represents the average of 2-4 replicate determinations.



FIG. 8 shows a comparison of genes that sensitize to different drug treatments. HeLa cells were transfected with siRNAs as shown in FIG. 1 and treated with either medium alone (or plus vehicle), or medium plus an EC10 concentration of Cis, Doxorubicin (Dox) or Camoptothecin (Campto). Cell growth was measured and is expressed the ratio of growth—drug/growth+drug. Dotted red lines indicate two-fold sensitization. Selected genes are indicated.



FIGS. 9A-9C show that silencing of WEE1 sensitizes HeLa cells to DNA damage induced by Dox, Campto, and Cis. FIGS. 9D-91 show that silencing of WEE1 sensitizes p53−A549 cells to DNA damage induced by Dox, Campto, and Cis, but does not sensitize p53+A549 cells to such DNA damage.



FIGS. 10A-10C show that silencing of EPHB3 sensitizes HeLa cells and p53−A549 C7, and to a lesser extent p53+ A549 pRS cells, to DNA damage induced by Dox, Campto, and Cis.



FIGS. 11A-11C show that silencing of STK6 sensitizes HeLa cells and p53−A549 C7, and to a lesser extent p53+ A549 pRS cells to DNA damage induced by Dox, Campto, and Cis.



FIGS. 12A-12C show that silencing of BRCA1 sensitizes HeLa cells and p53−A549 C7 cells to DNA damage induced by Dox, Campto, and Cis. Silencing of BRCA also sensitizes p53+ A549 pRS cells to DNA damage induced by Cis to a lesser extent, but does not sensitize p53+ A549 pRS cells to DNA damage induced by Dox and Campto.



FIGS. 13A-13B show that silencing of BRCA2 sensitizes HeLa cells and p53−A549 C7 cells to DNA damage induced by Dox, Campto, and Cis. FIG. 13C shows that silencing of BRCA2 sensitize p53+ A549 pRS cells to DNA damage induced by Cis to a lesser extent, but not dox and Campto.



FIGS. 14A-14B show that silencing of CHUK sensitizes HeLa cells to DNA damage induced by Dox, Campto, and Cis. FIG. 14C shows that silencing of CHUK sensitizes p53−A549 C7 cells to DNA damage induced by Campto and Cis. FIG. 14D shows that silencing of CHUK does not sensitize p53+ A549 pRS cells to DNA damage induced by Campto and Cis.


FIGS. 15A-C shows results of CHEK1 silencing on the sensitivity of cells to DNA damage. 15A CHEK1 silencing/inhibition sensitizes HeLa cells to DNA damage. 15B CHEK1 silencing/inhibition sensitizes p53−A549 cells. 15C CHEK1 silencing does not sensitize HREP cells to Doxorubicin.



FIG. 16 shows that siRNA mediated knockdown of PLK gene results in a cell cycle arrest and apoptosis.



FIG. 17 shows results of screens for genes that sensitize to KSPi.



FIG. 18 shows results of screens for genes that sensitize to Taxol.



FIG. 19 BRCA complexes enhance cisplatin activity. HeLa cells were transfected in 384 well format with siRNAs pools to ˜2,000 genes (3 siRNAs/gene) and then treated with (Y axis) or without (X axis) cisplatin. Two different cisplatin concentrations were tested, 100 ng/ml (˜EC10, left panel) or 400 ng/ml (˜EC50, right panel). Cell growth was measured 72 hrs post transfection using an Alamar Blue assay. Diagonal lines indicate concordance between the two treatments (black lines), or 2- and 3-fold sensitization by cisplatin treatment (magenta and red lines, respectively).



FIG. 20 Silencing of BRCA1 preferentially sensitizes TP53− cells to DNA damage. A549 cells stably transfected with empty vector (pRS, left panel) or an shRNA targeting TP53 (pRS-TP53, right panel) were supertransfected with siRNAs to luciferase, BRCA1, or BRCA2 prior to treatment with the DNA damaging agent, cisplatin. Cell growth was measured 72 hrs post-transfection using Alamar Blue.



FIG. 21 Silencing of BRCA1 selectively sensitizes TP53-cells to DNA damage. Matched TP53-negative (left column) or positive (right column) A549 cells were transfected with an siRNA to luciferase (top row) or BRCA1 (bottom row) prior to treatment with the DNA damaging agent, bleomycin. Seventy-two hours after transfection, cells were fixed, stained with propidium iodide and analyzed for cell cycle distribution by flow cytometry. The relative fluorescence of cells having 2N or 4N DNA content is indicated with arrows. The gates labeled in red indicate the number of sub-G1 (dead) cells.



FIG. 22 shows results that demonstrate that RAD51/Doxorubicin synergy is greater in TP53-cells.




5. DETAILED DESCRIPTION OF THE INVENTION

The invention provides methods and compositions for identifying interactions, e.g., lethal/synthetic lethal interactions, between a gene or its product and an agent, e.g., a drug, using RNA interference. As used herein, the term “gene product” includes mRNA transcribed from the gene and protein encoded by the gene. The invention also provides methods and compositions for treating cancer utilizing synthetic lethal interactions between STK6 kinase (also known as Aurora A kinase) and KSP (a kinesin-like motor protein, also known as KNSL1 or EG5) inhibitors (KSPi's). In this disclosure, a KSPi (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine
embedded image

(see, PCT application PCT/US03/18482, filed Jun. 12, 2003, which is incorporated herein by reference in its entirety), is often used. Other KSPi's can also be used in the invention. It is envisioned that methods utilize such other KSPi's are also encompassed by the present invention. The invention also provides methods and compositions for treating cancer utilizing interactions between a DNA damage response gene and a DNA damaging agent.


5.1. Methods of Screening of Interaction Using RNA Interference

The invention provides a method of identifying one or more genes in a cell of a cell type which interact with, e.g., modulate the effect of, an agent, e.g., a drug. As used herein, interaction of a gene with an agent or another gene includes interactions of the gene and/or its products with the agent or another gene/gene product. For example, an identified gene may confer resistance or sensitivity to a drug, i.e., reduces or enhances the effect of the drug. Such gene or genes can be identified by knocking down a plurality of different genes in cells of the cell type using a plurality of small interfering RNAs (knockdown cells), each of which targets one of the plurality of different genes, and determining which gene or genes among the plurality of different genes whose knockdown modulates the response of the cell to the agent. In one embodiment, a plurality of different knockdown cells (a knockdown library) are generated, each knockdown cell in the knockdown library comprising a different gene that is knockdown, e.g., by an siRNA. In another embodiment, a plurality of different knockdown cells (a knockdown library) are generated, each knockdown cell in the knockdown library comprising 2 or more different genes that are knockdown, e.g., by shRNA and siRNA targeting different genes. In one embodiment, the knockdown library comprises a plurality of cells, each of which expresses an siRNA targeting a primary gene and is supertransfected with one or more siRNAs targeting a secondary gene. It will be apparent to one skilled in the art that a knockdown cell may also be generated by other means, e.g., by using antisense, ribozyme, antibody, or a small organic or inorganic molecule that target the gene or its product. It is envisioned that any of these other means and means utilizing siRNA can be used alone or in combination to generate a knockdown library of the invention. Any method for siRNA silencing may be used, including methods that allow tuning of the level of silencing of the target gene. Section 5.2., infra, describes various methods that can be used.


In one embodiment, the method of the invention is practiced using an siRNA knockdown library comprising a plurality of cells of a cell type each comprising one of a plurality of siRNAs, each of the plurality of siRNAs targeting and silencing (i.e., knocking down) one of a plurality of different genes in the cell (i.e., knockdown cells). Any known method of introducing siRNAs into a cell can be used for this purpose. Preferably, each of the plurality of cells is generated and maintained separately such that they can be studied separately. Each of the plurality of cells is then treated with an agent, and the effect of the agent on the cell is determined. The effect of the agent on a cell comprising a gene silenced by an siRNA is then compared with the effect of the agent on cells of the cell type which do not comprise an siRNA, i.e., normal cells of the cell type. Knockdown cell or cells which exhibit a change in response to the agent are identified. The gene which is silenced by the comprised siRNA in such a knockdown cell is a gene which modulates the effect of the agent. Preferably, the plurality of siRNAs comprises siRNAs targeting and silencing at least 5, 10, 100, or 1,000 different genes in the cells. In a preferred embodiment, the plurality of siRNAs target and silence endogenous genes.


In a preferred embodiment, the knockdown library comprises a plurality of different knockdown cells having the same gene knocked down, e.g., each cell having a different siRNA targeting and silencing a same gene. The plurality of different knockdown cells having the same gene knocked down can comprises at least 2, 3, 4, 5, 6 or 10 different knockdown cells, each of which comprises an siRNA targeting a different region of the knocked down gene. In another preferred embodiment, the knockdown library comprises a plurality of different knockdown cells, e.g., at least 2, 3, 4, 5, 6, or 10, for each of a plurality of different genes represented in the knockdown library. In still another preferred embodiment, the knockdown library comprises a plurality of different knockdown cells, e.g., at least 2, 3, 4, 5, 6, or 10, for each of all different genes represented in the knockdown library.


In another preferred embodiment, the knockdown library comprises a plurality of different knockdown cells having different genes knocked down, each of the different knockdown cells has two or more different siRNA targeting and silencing a same gene. In preferred embodiment, each different knockdown cell can comprises at least 2, 3, 4, 5, 6 or 10 different siRNAs targeting the same gene at different regions.


In a preferred embodiment, the interaction of a gene with an agent is evaluated based on responses of a plurality of different knockdown cells having the gene knocked down, e.g., each cell having a different siRNA targeting and silencing a same gene. Utilizing the responses of a plurality of different siRNAs allows determination of the on-target and off-target effect of different siRNAs (see, e.g., International application No. PCT/U.S. 2004/015439 by Jackson et al., filed on May 17, 2004).


The effect of the agent on a cell of a cell type may be reduced in a knockdown cell as compared to that of a normal cell of the cell type, i.e., the knockdown of the gene mitigates the effect of the agent. The gene which is knocked down in such a cell is said to confer sensitivity to the agent. Thus, in one embodiment, the method of the invention is used for identifying one or more genes that confer sensitivity to an agent.


The effect of the agent on a cell of a cell type may be enhanced in a knockdown cell as compared to that of a normal cell of the cell type. The gene which is knocked down in such a cell is said to confer resistance to the agent. Thus, in another embodiment, the method of the invention is used for identifying a gene or genes that confers resistance to an agent. The enhancement of an effect of an agent may be additive or synergistic. In one embodiment, the invention provides a method for identifying one or more genes capable of regulating and/or enhancing the growth inhibitory effect of an anti-cancer drug in a cancer cell, e.g., a KSP inhibitor in cancer cells.


The method of the invention can be used for evaluating a plurality of different agents. For example, sensitivity to a plurality of different DNA damaging agents described in Section 5.4.2., infra, may be evaluated by the method of the invention. In a preferred embodiment, sensitivity of each knockdown cell in the knockdown library to each of the plurality of different agents is evaluated to generate a two-dimensional responsiveness matrix comprising measurement of effect of each agent on each knockdown cell. A cut at the gene axis at a particular gene index gives a profile of responses of the particular knockdown cell (in which the particular gene is knocked down) to different drugs. A cut at the drug axis at a particular drug gives a gene responsiveness profile to the drug, i.e., a profile comprising measurements of effect of the drug on different knockdown cells in the knockdown library. Tables IIA-IIC are examples of gene responsiveness profiles to cisplatin (Table IIA), camptothecin (Table IIB), and doxorubicin (Table IIC).


The method of the invention may be used for identifying interaction between different genes by using an agent that regulates, e.g., suppresses or enhances, the expression of a gene and/or an activity of a protein encoded by the gene. Examples of such agents include but are not limited to siRNA, antisense, ribozyme, antibody, and small organic or inorganic molecules that target the gene or its product. The gene targeted by such an agent is termed the primary target. Such an agent can be used in conjunction with a knockdown library to identify gene or genes which modulates the response of the cell to the agent. The primary target can be different from any of the plurality of genes represented in the knockdown library (secondary genes). The gene or genes identified as modulating the effect of the agent are therefore gene or genes that interact with the primary target.


In a preferred embodiment, the invention provides a method for indentifying interaction between different genes using a dual siRNA approach. In a preferred embodiment, dual RNAi screens is achieved through the use of stable in vivo delivery of an shRNA disrupting the primary target gene and supertransfection of an siRNA targeting a secondary target gene. This approach provides matched (isogenic) cell line pairs (plus or minus the shRNA) and does not result in competition between the shRNA and siRNA. In the method, short hairpin RNAs (shRNAs) are expressed from recombinant vectors introduced either transiently or stably integrated into the genome (see, e.g., Paddison et al., 2002, Genes Dev 16:948-958; Sui et al., 2002, Proc Natl Acad Sci USA 99:5515-5520; Yu et al., 2002, Proc Natl Acad Sci USA 99:6047-6052; Miyagishi et al., 2002, Nat Biotechnol 20:497-500; Paul et al., 2002, Nat Biotechnol 20:505-508; Kwak et al., 2003, J Pharmacol Sci 93:214-217; Brummelkamp et al., 2002, Science 296:550-553; Boden et al., 2003, Nucleic Acids Res 31:5033-5038; Kawasaki et al., 2003, Nucleic Acids Res 31:700-707). The siRNA that disrupts the primary gene can be expressed (via an shRNA) by any suitable vector which encodes the shRNA. The vector can also encode a marker which can be used for selecting clones in which the vector or a sufficient portion thereof is integrated in the host genome such that the shRNA is expressed. Any standard method known in the art can be used to deliver the vector into the cells. In one embodiment, cells expressing the shRNA are generated by transfecting suitable cells with a plasmid containing the vector. Cells can then be selected by the appropriate marker. Clones are then picked, and tested for knockdown. In a preferred embodiment, the expression of the shRNA is under the control of an inducible promoter such that the silencing of its target gene can be turned on when desired. Inducible expression of an siRNA is particularly useful for targeting essential genes.


In one embodiment, the expression of the shRNA is under the control of a regulated promoter that allows tuning of the silencing level of the target gene. This allows screening against cells in which the target gene is partially knocked out. As used herein, a “regulated promoter” refers to a promoter that can be activated when an appropriate inducing agent is present. An “inducing agent” can be any molecule that can be used to activate transcription by activating the regulated promoter. An inducing agent can be, but is not limited to, a peptide or polypeptide, a hormone, or an organic small molecule. An analogue of an inducing agent, i.e., a molecule that activates the regulated promoter as the inducing agent does, can also be used. The level of activity of the regulated promoter induced by different analogues may be different, thus allowing more flexibility in tuning the activity level of the regulated promoter. The regulated promoter in the vector can be any mammalian transcription regulation system known in the art (see, e.g., Gossen et al, 1995, Science 268:1766-1769; Lucas et al, 1992, Annu. Rev. Biochem. 61:1131; Li et al., 1996, Cell 85:319-329; Saez et al., 2000, Proc. Natl. Acad. Sci. USA 97:14512-14517; and Pollock et al., 2000, Proc. Natl. Acad. Sci. USA 97:13221-13226). In preferred embodiments, the regulated promoter is regulated in a dosage and/or analogue dependent manner. In one embodiment, the level of activity of the regulated promoter is tuned to a desired level by a method comprising adjusting the concentration of the inducing agent to which the regulated promoter is responsive. The desired level of activity of the regulated promoter, as obtained by applying a particular concentration of the inducing agent, can be determined based on the desired silencing level of the target gene.


In one embodiment, a tetracycline regulated gene expression system is used (see, e.g., Gossen et al, 1995, Science 268:1766-1769; U.S. Pat. No. 6,004,941). A tet regulated system utilizes components of the tet repressor/operator/inducer system of prokaryotes to regulate gene expression in eukaryotic cells. Thus, the invention provides methods for using the tet regulatory system for regulating the expression of an shRNA linked to one or more tet operator sequences. The methods involve introducing into a cell a vector encoding a fusion protein that activates transcription. The fusion protein comprises a first polypeptide that binds to a tet operator sequence in the presence of tetracycline or a tetracycline analogue operatively linked to a second polypeptide that activates transcription in cells. By modulating the concentration of a tetracycline, or a tetracycline analogue, expression of the tet operator-linked shRNA is regulated.


In other embodiments, an ecdyson regulated gene expression system (see, e.g., Saez et al., 2000, Proc. Natl. Acad. Sci. USA 97:14512-14517), or an MMTV glucocorticoid response element regulated gene expression system (see, e.g., Lucas et al, 1992, Annu. Rev. Biochem. 61:1131) may be used to regulate the expression of the shRNA.


In one embodiment, a pRETRO-SUPER (pRS) vector which encodes a puromycin-resistance marker and drives shRNA expression from an H1 (RNA Pol III) promoter is used. The pRS-shRNA plasmid can be generated by any standard method known in the art. In one embodiment, the pRS-shRNA is deconvoluted from a library plasmid pool for a chosen gene by transforming bacteria with the pool and looking for clones containing only the plasmid of interest. Preferably, a 19mer siRNA sequence is used along with suitable forward and reverse primers for sequence specific PCR. Plasmids are identified by sequence specific PCR, and confirmed by sequencing. Cells expressing the shRNA are generated by transfecting suitable cells with the pRS-shRNA plasmid. Cells are selected by the appropriate marker, e.g., puromycin, and maintained until colonies are evident. Clones are then picked, and tested for knockdown.


In another embodiment, an shRNA is expressed by a plasmid, e.g., a pRS-shRNA. The knockdown by the pRS-shRNA plasmid, can be achieved by transfecting cells using Lipofectamine 2000 (Invitrogen).


In a preferred embodiment, matched cell lines (+/− primary target gene) are generated by selecting stable clones containing either empty pRS vector or pRS-shRNA.


Silencing of the secondary target gene are then carried out using cells of a generated shRNA primary target clone. Silencing of the secondary target gene can be achieved using any known method of RNA interference (see, e.g., Section 5.2.). For example, secondary target gene can be silenced by transfection with siRNA and/or plasmid encoding an shRNA. In one embodiment, cells of a generated shRNA primary target clone are supertransfected with one or more siRNAs targeting a secondary target gene. In one embodiment, the one or more siRNAs targeting the secondary gene are transfected into the cells directly. In another embodiment, the one or more siRNAs targeting the secondary gene are transfected into the cells via shRNAs using one or more suitable plasmids. RNA can be harvested 24 hours post transfection and knockdown assessed by TaqMan analysis. In a preferred embodiment, an siRNA pool containing at least k (k=2, 3, 4, 5, 6 or 10) different siRNAs targeting the secondary target gene at different sequence regions is used to supertransfect the cells. In another preferred embodiment, an siRNA pool containing at least k (k=2, 3, 4, 5, 6 or 10) different siRNAs targeting two or more different secondary target genes is used to supertransfect the cells.


In a preferred embodiment, the total siRNA concentration of the pool is about the same as the concentration of a single siRNA when used individually, e.g., 100 nM. Preferably, the total concentration of the pool of siRNAs is an optimal concentration for silencing the intended target gene. An optimal concentration is a concentration further increase of which does not increase the level of silencing substantially. In one embodiment, the optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 5%, 10% or 20%. In a preferred embodiment, the composition of the pool, including the number of different siRNAs in the pool and the concentration of each different siRNA, is chosen such that the pool of siRNAs causes less than 30%, 20%, 10% or 5%, 1%, 0.1% or 0.01% of silencing of any off-target genes. In another preferred embodiment, the concentration of each different siRNA in the pool of different siRNAs is about the same. In still another preferred embodiment, the respective concentrations of different siRNAs in the pool are different from each other by less than 5%, 10%, 20% or 50%. In still another preferred embodiment, at least one siRNA in the pool of different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration in the pool. In still another preferred embodiment, none of the siRNAs in the pool of different siRNAs constitutes more than 90%, 80%, 70%, 50%, or 20% of the total siRNA concentration in the pool. In other embodiments, each siRNA in the pool has an concentration that is lower than the optimal concentration when used individually. In a preferred embodiment, each different siRNA in the pool has an concentration that is lower than the concentration of the siRNA that is effective to achieve at least 30%, 50%, 75%, 80%, 85%, 90% or 95% silencing when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In another preferred embodiment, each different siRNA in the pool has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the gene when used in the absence of other siRNAs or in the absence of other siRNAs designed to silence the gene. In a preferred embodiment, each siRNA has a concentration that causes less than 30%, 20%, 10% or 5% of silencing of the target gene when used alone, while the plurality of siRNAs causes at least 80% or 90% of silencing of the target gene.


In one embodiment, the invention provides a method for identifying one or more genes which exhibit synthetic lethal interaction with a primary target gene. In the method, an agent that is an inhibitor of the primary target gene in the cell type is used to screen against a knockdown library. The gene or genes identified as enhancing the effect of the agent are therefore gene or genes that have synthetic lethal interaction with the primary target. In a preferred embodiment, the agent is an siRNA targeting and silencing the primary target.


The method for determining the effect of an agent on cells depends on the particular effect to be evaluated. For example, if the agent is an anti-cancer drug, and the effect to be evaluated is the growth inhibitory effect of the drug, an MTT assay or an alamarBlue assay may be used (see, e.g., Section 5.2). One skilled person in the art will be able to choose a method known in the art based on the particular effect to be evaluated.


In another embodiment, the invention provides a method of determining the effect of an agent on the growth of cells having the primary target gene and the secondary target gene silenced. In a preferred embodiment, matched cell lines (+/− primary target gene) are generated as described above. Both cell lines are then supertransfected with either a control siRNA (e.g., luciferase) or one or more siRNAs targeting a secondary target gene. The cell cycle profiles are examined with or without exposure to the agent. Cell cycle analysis can be carried out using standard method known in the art (see, Section 5.2., infra). In one embodiment, the supernatant from each well is combined with the cells that have been harvested by trypsinization. The mixture is then centrifuged at a suitable speed. The cells are then fixed with ice cold 70% ethanol for a suitable period of time, e.g., ˜30 minutes. Fixed cells can be washed once with PBS and resuspended, e.g., in 0.5 ml of PBS containing Propidium Iodide (10 microgram/ml) and RNase A(1 mg/ml), and incubated at a suitable temperature, e.g., 37° C., for a suitable period of time, e.g., 30 min. Flow cytometric analysis is carried out using a flow cytometer. In one embodiment, the Sub-G1 cell population is used to measure cell death. An increase of sub-G1 cell population in cells having the primary target gene and the secondary target gene silenced indicates synthetic lethality between the primary and secondary target genes in the presence of the agent.


In a specific embodiment, the invention provides a method for identifying gene or genes whose knockdown enhances the growth inhibitory effect of a KSP inhibitor on tumor cells. In one embodiment, the method was used to identify genes whose knockdown inhibits tumor cell growth in the presence of suboptimal concentrations of a KSPi, i.e., concentrations lower than EC10. In one embodiment, an siRNA knockdown library contained 3 siRNAs targeting each of the following 11 genes: CDC20, ROCK2, TTK, FZR1, BUB1, BUB3, BUB1B, MAD1L1, MAD2L1, DNCH1 and STK6 are generated and used (see Table I). Each of these siRNAs were transfected into HeLa cells in the presence or absence of an <EC10 concentration (25 nM) of a KSPi, (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine (see, PCT application PCT/US03/18482, filed Jun. 12, 2003) (EC50˜80 nM) and the response of the cell was determined. One siRNA to STK6 (STK6-1) showed significant inhibition of tumor cell growth in the presence of KSPi.


The growth inhibitory activity was further examined using three additional siRNAs to STK6 and the abilities of all six siRNAs to induce STK6 silencing and growth inhibition were evaluated. Amongst the different siRNAs, there was a good correlation between the level of STK6 silencing and growth inhibition (FIG. 1). This correlation suggested that growth inhibition was due to on target activity (i.e., STK6 disruption). STK6-1 was then titrated with control siRNAs targeting luciferase (negative control) in the presence or absence of (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine as described in PCT application PCT/US03/18482, filed Jun. 12, 2003. (FIG. 2). The addition of KSPi shifted the STK6-1 dose response curve 5-10-fold to the left. This concentration of KSPi did not augment effects on cell growth caused by a luciferase siRNA. In contrast, the dose response curve to a siRNA targeting PTEN with similar effects on cell growth as STK6-1 was not shifted by KSPi. Other siRNAs targeting STK6 also enhanced the effect of KSPi on cell growth. Thus, disruption of STK6 enhances the effect of KSPi on cell growth. Further support for this was obtained by studies using combinations of siRNAs to STK6 and KSP (Table I), which showed greater growth inhibitory activity than either siRNA alone. Because the concentrations of KSPi used in these experiments did not affect cell growth on its own, the effects of KSPi on STK6 siRNA activity appeared synergistic rather than additive.


In another specific embodiment, the invention provides a method for determining synthetic lethality between p53 and CHEK1. Stable clones having p53 gene silenced was generated. The pRS-TP53 1026 shRNA plasmid was deconvoluted from a library plasmid pool for TP53 by transforming bacteria with the pool and looking for clones containing only the plasmid of interest. The sequences used are as follows: pRS-p53 1026 19mer sequence: GACTCCAGTGGTAATCTAC (SEQ ID NO:43); primers for sequence specific PCR: Forward: GTAGATTACCACTGGAGTC (SEQ ID NO:44), Reverse: CCCTTGAACCTCCTCGTTCGACC (SEQ ID NO:45). Plasmids were identified by sequence specific PCR, and confirmed by sequencing. Stable p53-clones were generated by transfecting HCT116 cells using FuGENE 6 (Roche) with the pRS-TP53 1026 plasmid. Cells were split into 10 cm dishes plus 1 ug/ml puromycin 48 hours later, and maintained until colonies were evident (5-7 days). Clones were picked into a 96 well plate, maintained in 1 ug/ml puromycin, and tested for knockdown by TaqMan using the TP53 and hGUS Pre-Developed Assay Reagent (Applied Biosystems). To measure transient knockdown by the pRS-TP53 1026 plasmid, HCT116 cells were transfected using Lipofectamine 2000 (Invitrogen), and RNA harvested 24 hours later. TP53 transcript levels were assessed by TaqMan.


Analysis of multiple puromycin-resistant TP53 shRNA clones (pRS-p53) derived from the colon tumor line HCT116 showed varying levels of target silencing (50% to 96% as determined by TaqMan). FIG. 3 shows the level of TP53 expression in clones A5 and A11, which exhibited the highest levels of silencing. TP53 silencing achieved in these clones exceeded that observed 24 hr after delivery of pRS-p53 into HCT116 cells by transient transfection (FIG. 3). It is possible that transfection efficiency limits the effectiveness of TP53 shRNA in transient assays. Alternatively, cells having greater levels of TP53 silencing gain a growth advantage during clonal growth. With an shRNA that targets STK6 (pRS-STK6: pRS-STK6 2178 19mer sequence: CATTGGAGTCATAGCATGT (SEQ ID NO:46)), a range of silencing in stable clones was also observed. These clones, however, did not achieve as high a degree of silencing observed in the TP53 lines, nor was silencing greater than that achieved in transient assays. This may indicate selection against high level of STK6 silencing because STK6 is an essential gene for tumor cell growth in culture.


To test whether TP53 silencing in HCT116 clone A11 was subject to competition with siRNAs, cells of this clone were supertransfected with a pool of CHEK1-specific siRNAs. CHK1 pool contains the following three siRNAs: CUGAAGAAGCAGUCGCAGUTT (SEQ ID NO:99); AUCGAUUCUGCUCCUCUAGTT (SEQ ID NO:98); and UGCCUGAAAGAGACUUGUGTT (SEQ ID NO:100). This siRNA pool had been found to competitively reduce silencing activity of a TP53 targeted siRNA. siRNAs were transfected using Oligofectamine (Invitrogen) at 10 nM or 100 nM where indicated. For the CHK1 pool, three siRNAs were transfected simultaneously at 33.3 nM each for a total delivery of 100 nM. RNA was harvested 24 hours post transfection and knockdown was assessed by TaqMan analysis using the CHK1 or TP53 and hGUS Pre-Developed Assay Reagent (Applied Biosystems). As shown in FIG. 4A, the shRNA and the siRNA pool did not competitively inhibit silencing of each other's targets. Inhibition by known competitive siRNAs of either a transiently transfected siRNA or a stably expressed shRNA of the same sequence was then assayed. As shown in FIG. 4B, three individual siRNAs targeting KNSL1 (KNSLI 210: GACCUGUGCCUUUUAGAGATT (SEQ ID NO:47); KNSLI 211: GACUUCAUUGACAGUGGCCTT (SEQ ID NO:48); KNSLI 212: AAAGGACAACUGCAGCUACTT (SEQ ID NO:49)) competitively inhibited the silencing achieved by co-transfected siRNA targeting STK6 (left bars). In contrast, silencing by the homologous STK6 shRNA in stably transfected lines was unaffected by supertransfection of the KNSL1 siRNAs, even when the competitor siRNAs were added at ten fold higher concentrations (middle and right bars). These experiments suggested that there was little competition between stably expressed shRNAs and transiently transfected siRNAs. This is in contrast to the observation that two different siRNAs targeting distinct mRNAs compete with each other when transfected together, effectively decreasing the efficacy of one or both of the siRNAs used. pRS and pRS-p53 HCT116 cells were transiently transfected with siRNA pools for ˜800 genes (see Example 3, infra) and measured effects on cellular growth by Alamar Blue assay. Nearly identical responses to the ˜800 siRNA pools in pRS cells and in pRS-p53 cells, with no suggestion of competitive inhibition of silencing were observed.


Next, supertransfection of the CHEK1 siRNA pool into cells stably expressing TP53 shRNAs was evaluated to determine if it could be used to investigate genetic interactions (SL) between these molecules. Matched cell lines +/−TP53 expression were generated by selecting stable clones of A549 lung cancer cell lines containing either empty pRS vector or pRS-p53. The latter cells showed >90% silencing of TP53 mRNA. Both cell lines were then supertransfected with either control luciferase siRNA (luc, 100 nM) or the CHEK1 siRNA pool (100 nM total; 33 nM each of 3 siRNAs) and their cell cycle profiles examined with or without exposure to the DNA damaging agent, doxorubicin (Dox, FIG. 5). Cell cycle profiles of pRS-p53 cells were not appreciably different from those of pRS cells in the absence of Dox. Transient transfection of CHEK1 siRNAs also did not affect cell cycle profiles in the absence of Dox. In the presence of Dox, however, pRS-transfected cells exhibited G1 and G2/M arrest as is expected of cells expressing functional TP53. Supertransfection of CHEK1 siRNAs resulted in an override of the G2 checkpoint and an increase in the number of cells blocked at G1. Because the cells retained TP53 function, they stopped in G1 and did not proceed back into S phase.


In contrast, pRS-p53 cells lost the ability to arrest at G1 and arrested primarily at G2 in response Dox treatment, consistent with the role of TP53 in the G1 checkpoint. The cell cycle profile of pRS-p53 cells was unchanged by supertransfection of luc siRNA (FIG. 5). The failure of luc siRNA to cause even partial restoration of the TP53 response (and a corresponding increase in the G1 peak) suggests that there was little competitive inhibition of TP53 silencing and phenotype by this siRNA. Therefore, competitive inhibition of TP53 silencing by the CHEK1 siRNA pool was not expected to exist. Indeed, in response to Dox treatment, pRS-p53 cells transiently transfected with CHEK1 showed profound alterations in their cell cycle profile with large increases in the fraction of cells in S and with sub-G1 (dead cells) amounts of DNA. Similar findings were also observed in pRS and pRS-p53 stably transfected HCT116 cells. Thus, simultaneous disruption of the G1 checkpoint mediated by TP53 and the G2 checkpoint mediated by CHEK1 is lethal to TP53− but not TP53+ tumor cells.


In another embodiment, the invention provides a method for determining synthetic lethality between p53 and a member of the BRCC complex, e.g., BRCA1, BRCA2, BARD1 and RAD51. In this embodiment, a matched pair of TP53 positive and negative cells generated by stable expression of short hairpin RNAs (shRNAs) targeting TP53 was used. TP53-positive or negative cells were supertransfected with siRNA pools to BRCA1 or BRCA2, treated with cisplatin and analyzed for cell growth using Alamar Blue (FIG. 20). TP53-negative cells were ˜10-fold more sensitive to cisplatin when transfected with BRCA1 or BRCA2 siRNAs (IC50˜0.1 nM) than with control siRNA (luciferase, IC50-˜1 nM). The sensitization to cisplatin following BRCA1 or BRCA2 disruption was even more pronounced at lower cisplatin concentrations. TP53-positive cells were less sensitized to cisplatin following BRCA1 or BRCA2 disruption (IC50 ˜0.4 nM). Sensitization to cisplatin following BRCA1 or BRCA2 disruption was similar in magnitude in this assay to the sensitization seen following disruption of CHEK1 (data not shown). Sensitization to DNA damaging agents following BRCA1 and BRCA2 disruption can also be investigated using cell cycle analysis. TP53-positive and negative cells were supertransfected with siRNA pools to BRCA1 or BRCA2, treated with one of several DNA damaging agents (cisplatin, camptothecin, doxorubicin and bleomycin) and analyzed for cell cycle distribution by flow cytometry. In all cases, TP53-negative cells were more sensitive to DNA damage following BRCA1 or BRCA2 disruption than in luciferase-transfected cells (data not shown). The response of these cells to bleomycin following BRCA1 disruption is shown in FIG. 21. BRCA1 disruption resulted in more sub-G1 cells (dead cells) following bleomycin treatment of TP53-negative than TP53-positive cells. The results show that cells lacking TP53 are more sensitive to DNA damage following BRCA1 disruption.


The cell lines used can be HeLa cells, TP53-positive A549 cells or TP53-negative A549 cells. In one embodiment, matched pair of TP53 positive and negative cells were generated by stable transfection of short hairpin RNAs (shRNAs) targeting TP53 (monthly highlt highlight, November 2003). The cells were transfected using pools of siRNAs (pool of 3 siRNA per gene) at 100 nM (each siRNA at 33 nM), or with single siRNA at 100 nM. The following siRNAs were used: Luc control, BRCA1, BRCA2 and BARD1 pool. These transfected cells were then treated with varying concentrations of DNA damaging agents. The concentration for each agent used in the cell cycle analysis is as follows: for HeLa cells, Doxorubicin (10 nM), Camptothecin (6 nM), Cisplatin (400 ng/ml), Mitomycin C (40 nM), Bleomycin (100 ng/ml); for the other cell lines, Doxorubicin (200 nM), Camptothecin (200 nM), Cisplatin (2 ug/ml), Mitomycin C (400 nM), Bleomycin (5 ug/ml).


In one embodiment, siRNA transfection was carried out as follows: one day prior to transfection, 2000 (or 100) microliters of a chosen cell line, e.g., cervical cancer HeLa cells (ATCC, Cat. No. CCL-2), grown in DMEM/10% fetal bovine serum (Invitrogen, Carlsbad, Calif.) to approximately 90% confluency were seeded in a 6-well (or 96-well) tissue culture plate at 45,000 (or 2000) cells/well. For each transfection 70 microliters of OptiMEM (Invitrogen) was mixed with 5 microliter of siRNA (Dharmacon, Lafayette, Colo.) from a 20 micromolar stock. For each transfection, a ratio of 20 microliter of OptiMEM was mixed with 5 microliter of Oligofectamine reagent (Invitrogen) and incubated 5 minutes at room temperature. Then 25-microliter OptiMEM/Oligofectamine mixture was mixed with the 75-microliter of OptiMEM/siRNA mixture, and incubated 15-20 minutes at room temperature. 100 (or 10) microliter of the transfection mixture was aliquoted into each well of the 6-well (or 96-well) plate and incubated for 4 hours at 37° C. and 5% CO2.


After 4 hours, 100 microliter/well of DMEM/10% fetal bovine serum with or without DNA damage agents was added to each well to reach the final concentration of each agents as described above. The plates were incubated at 37° C. and 5% CO2 for another 68 hours. Samples from the 6-well plates were analyzed for cell cycle profiles and samples from 96-well plates were analyzed for cell growth with Alamar Blue assay.


For cell cycle analysis, the supernatant from each well was combined with the cells that were harvested by trypsinization. The mixture was then centrifuged at 1200 rpm for 5 minutes. The cells were then fixed with ice cold 70% ethanol for ˜30 minutes. Fixed cells were washed once with PBS and resuspended in 0.5 ml of PBS containing Propidium Iodide (10 microgram/ml) and RNase A (1 mg/ml), and incubated at 37° C. for 30 min. Flow cytometric analysis was carried out using a FACSCalibur flow cytometer (Becton Dickinson) and the data was analyzed using FlowJo software (Tree Star, Inc). The Sub-G1 cell population was used to measure cell death. If the summation of the Sub-G1 population from the (siRNA+DMSO) sample and (Luc+drug) sample is larger than the Sub-G1 population of (siRNA+drug) sample, we define that as sensitization of siRNA silencing to DNA damage.


For Alamar Blue assay, the media from the 96-well plates was removed, and 100 uL/well complete media containing 10% (vol/vol) alamarBlue reagent (BioSource International, Inc) and 1/100th volume 1M Hepes buffer tissue culture reagent was added. The plates were then incubated 1-4 hours at 37° C. and fluorescence was measured by exciting at 544 nm and detecting emission at 590 nm with SPECTRAMax Gemini-Xs Spectrofluorometer (Molceular Devices). The fluorescence signal was corrected for background (no cells). Cell response (survival) in the presence of DNA damaging agents was measured as a percentage of control cell growth in the absence of DNA damaging agents.


5.2. Methods and Compositions for RNA Interference and Cell Assays

Any standard method for gene silencing can be used in the present invention (see, e.g., Guo et al., 1995, Cell 81:611-620; Fire et al., 1998, Nature 391:806-811; Grant, 1999, Cell 96:303-306; Tabara et al., 1999, Cell 99:123-132; Zamore et al., 2000, Cell 101:25-33; Bass, 2000, Cell 101:235-238; Petcherski et al., 2000, Nature 405:364-368; Elbashir et al., Nature 411:494-498; Paddison et al., Proc. Natl. Acad. Sci. USA 99:1443-1448). The siRNAs targeting a gene can be designed according to methods known in the art (see, e.g., U.S. Provisional Patent Application No. 60/572,314 by Jackson et al., filed on May 17, 2004, and Elbashir et al., 2002, Methods 26:199-213, each of which is incorporated herein by reference in its entirety).


SiRNAs having only partial sequence homology to a target gene can also be used (see, e.g., International application No. PCT/U.S. 2004/015439 by Jackson et al., filed on May 17, 2004, which is incorporated herein by reference in its entirety). In one embodiment, an siRNA that comprises a sense strand contiguous nucleotide sequence of 11-18 nucleotides that is identical to a sequence of a transcript of a gene but the siRNA does not have full length homology to any sequences in the transcript is used to silence the gene. Preferably, the contiguous nucleotide sequence is in the central region of the siRNA molecules. A contiguous nucleotide sequence in the central region of an siRNA can be any continuous stretch of nucleotide sequence in the siRNA which does not begin at the 3′ end. For example, a contiguous nucleotide sequence of 11 nucleotides can be the nucleotide sequence 2-12, 3-13, 4-14, 5-15, 6-16, 7-17, 8-18, or 9-19. In preferred embodiments, the contiguous nucleotide sequence is 11-16, 11-15, 14-15, 11, 12, or 13 nucleotides in length.


In another embodiment, an siRNA that comprises a 3′ sense strand contiguous nucleotide sequence of 9-18 nucleotides which is identical to a sequence of a transcript of a gene but which siRNA does not have full length sequence identity to any contiguous sequences in the transcript is used to silence the gene. In this application, a 3′ 9-18 nucleotide sequence is a continuous stretch of nucleotides that begins at the first paired base, i.e., it does not comprise the two base 3′ overhang. Thus, when it is stated that a particular nucleotide sequence is at the 3′ end of the siRNA, the 2 base overhang is not considered. In preferred embodiments, the contiguous nucleotide sequence is 9-16, 9-15, 9-12, 11, 10, or 9 nucleotides in length.


Any method known in the art can be used for carrying out RNA interference. In one embodiment, gene silencing is induced by presenting the cell with the siRNA, mimicking the product of Dicer cleavage (see, e.g., Elbashir et al., 2001, Nature 411, 494-498; Elbashir et al., 2001, Genes Dev. 15, 188-200, all of which are incorporated by reference herein in their entirety). Synthetic siRNA duplexes maintain the ability to associate with RISC and direct silencing of mRNA transcripts. siRNAs can be chemically synthesized, or derived from cleavage of double-stranded RNA by recombinant Dicer. Cells can be transfected with the siRNA using standard method known in the art.


In one embodiment, siRNA transfection is carried out as follows: one day prior to transfection, 100 microliters of chosen cells, e.g., cervical cancer HeLa cells (ATCC, Cat. No. CCL-2), grown in DMEM/10% fetal bovine serum (Invitrogen, Carlsbad, Calif.) to approximately 90% confluency are seeded in a 96-well tissue culture plate (Corning, Corning, N.Y.) at 1500 cells/well. For each transfection 85 microliters of OptiMEM (Invitrogen) is mixed with 5 microliter of serially diluted siRNA (Dharma on, Denver) from a 20 micro molar stock. For each transfection 5 microliter OptiMEM is mixed with 5 microliter Oligofectamine reagent (Invitrogen) and incubated 5 minutes at room temperature. The 10 microliter OptiMEM/Oligofectamine mixture is dispensed into each tube with the OptiMEM/siRNA mixture, mixed and incubated 15-20 minutes at room temperature. 10 microliter of the transfection mixture is aliquoted into each well of the 96-well plate and incubated for 4 hours at 37° C. and 5% CO2.


Another method for gene silencing is to introduce an shRNA, for short hairpin RNA (see, e.g., Paddison et al., 2002, Genes Dev. 16, 948-958; Brummelkamp et al., 2002, Science 296, 550-553; Sui, G. et al. 2002, Proc. Natl. Acad. Sci. USA 99, 5515-5520, all of which are incorporated by reference herein in their entirety), which can be processed in the cells into siRNA. In this method, a desired siRNA sequence is expressed from a plasmid (or virus) as an inverted repeat with an intervening loop sequence to form a hairpin structure. The resulting RNA transcript containing the hairpin is subsequently processed by Dicer to produce siRNAs for silencing. Plasmid-based shRNAs can be expressed stably in cells, allowing long-term gene silencing in cells both in vitro and in vivo, e.g., in animals (see, McCaffrey et al. 2002, Nature 418, 38-39; Xia et al., 2002, Nat. Biotech. 20, 1006-1010; Lewis et al., 2002, Nat. Genetics 32, 107-108; Rubinson et al., 2003, Nat. Genetics 33, 401-406; Tiscomia et al., 2003, Proc. Natl. Acad. Sci. USA 100, 1844-1848, all of which are incorporated by reference herein in their entirety). Thus, in one embodiment, a plasmid-based shRNA is used.


In a preferred embodiment, shRNAs are expressed from recombinant vectors introduced either transiently or stably integrated into the genome (see, e.g., Paddison et al., 2002, Genes Dev 16:948-958; Sui et al., 2002, Proc Natl Acad Sci USA 99:5515-5520; Yu et al., 2002, Proc Natl Acad Sci USA 99:6047-6052; Miyagishi et al., 2002, Nat Biotechnol 20:497-500; Paul et al., 2002, Nat Biotechnol 20:505-508; Kwak et al., 2003, J Pharmacol Sci 93:214-217; Brummelkamp et al., 2002, Science 296:550-553; Boden et al., 2003, Nucleic Acids Res 31:5033-5038; Kawasaki et al., 2003, Nucleic Acids Res 31:700-707). The siRNA that disrupts the target gene can be expressed (via an shRNA) by any suitable vector which encodes the shRNA. The vector can also encode a marker which can be used for selecting clones in which the vector or a sufficient portion thereof is integrated in the host genome such that the shRNA is expressed. Any standard method known in the art can be used to deliver the vector into the cells. In one embodiment, cells expressing the shRNA are generated by transfecting suitable cells with a plasmid containing the vector. Cells can then be selected by the appropriate marker. Clones are then picked, and tested for knockdown. In a preferred embodiment, the expression of the shRNA is under the control of an inducible promoter such that the silencing of its target gene can be turned on when desired. Inducible expression of an siRNA is particularly useful for targeting essential genes.


In one embodiment, the expression of the shRNA is under the control of a regulated promoter that allows tuning of the silencing level of the target gene. This allows screening against cells in which the target gene is partially knocked out. As used herein, a “regulated promoter” refers to a promoter that can be activated when an appropriate inducing agent is present. An “inducing agent” can be any molecule that can be used to activate transcription by activating the regulated promoter. An inducing agent can be, but is not limited to, a peptide or polypeptide, a hormone, or an organic small molecule. An analogue of an inducing agent, i.e., a molecule that activates the regulated promoter as the inducing agent does, can also be used. The level of activity of the regulated promoter induced by different analogues may be different, thus allowing more flexibility in tuning the activity level of the regulated promoter. The regulated promoter in the vector can be any mammalian transcription regulation system known in the art (see, e.g., Gossen et al, 1995, Science 268:1766-1769; Lucas et al, 1992, Annu. Rev. Biochem. 61:1131; L1 et al., 1996, Cell 85:319-329; Saez et al., 2000, Proc. Natl. Acad. Sci. USA 97:14512-14517; and Pollock et al., 2000, Proc. Natl. Acad. Sci. USA 97:13221-13226). In preferred embodiments, the regulated promoter is regulated in a dosage and/or analogue dependent manner. In one embodiment, the level of activity of the regulated promoter is tuned to a desired level by a method comprising adjusting the concentration of the inducing agent to which the regulated promoter is responsive. The desired level of activity of the regulated promoter, as obtained by applying a particular concentration of the inducing agent, can be determined based on the desired silencing level of the target gene.


In one embodiment, a tetracycline regulated gene expression system is used (see, e.g., Gossen et al, 1995, Science 268:1766-1769; U.S. Pat. No. 6,004,941). A tet regulated system utilizes components of the tet repressor/operator/inducer system of prokaryotes to regulate gene expression in eukaryotic cells. Thus, the invention provides methods for using the tet regulatory system for regulating the expression of an shRNA linked to one or more tet operator sequences. The methods involve introducing into a cell a vector encoding a fusion protein that activates transcription. The fusion protein comprises a first polypeptide that binds to a tet operator sequence in the presence of tetracycline or a tetracycline analogue operatively linked to a second polypeptide that activates transcription in cells. By modulating the concentration of a tetracycline, or a tetracycline analogue, expression of the tet operator-linked shRNA is regulated.


In other embodiments, an ecdyson regulated gene expression system (see, e.g., Saez et al., 2000, Proc. Natl. Acad. Sci. USA 97:14512-14517), or an MMTV glucocorticoid response element regulated gene expression system (see, e.g., Lucas et al, 1992, Annu. Rev. Biochem. 61:1131) may be used to regulate the expression of the shRNA.


In one embodiment, the pRETRO-SUPER (pRS) vector which encodes a puromycin-resistance marker and drives shRNA expression from an H1 (RNA Pol III) promoter is used. The pRS-shRNA plasmid can be generated by any standard method known in the art. In one embodiment, the pRS-shRNA is deconvoluted from the library plasmid pool for a chosen gene by transforming bacteria with the pool and looking for clones containing only the plasmid of interest. Preferably, a 19mer siRNA sequence is used along with suitable forward and reverse primers for sequence specific PCR. Plasmids are identified by sequence specific PCR, and confirmed by sequencing. Cells expressing the shRNA are generated by transfecting suitable cells with the pRS-shRNA plasmid. Cells are selected by the appropriate marker, e.g., puromycin, and maintained until colonies are evident. Clones are then picked, and tested for knockdown. In another embodiment, an shRNA is expressed by a plasmid, e.g., a pRS-shRNA. The knockdown by the pRS-shRNA plasmid, can be achieved by transfecting cells using Lipofectamine 2000 (Invitrogen).


In yet another method, siRNAs can be delivered to an organ or tissue in an animal, such a human, in vivo (see, e.g., Song et al. 2003, Nat. Medicine 9, 347-351; Sorensen et al., 2003, J. Mol. Biol. 327, 761-766; Lewis et al., 2002, Nat. Genetics 32, 107-108, all of which are incorporated by reference herein in their entirety). In this method, a solution of siRNA is injected intravenously into the animal. The siRNA can then reach an organ or tissue of interest and effectively reduce the expression of the target gene in the organ or tissue of the animal.


Any suitable proliferation or growth inhibition assays known in the art can be used to assay cell growth. In a preferred embodiment, an MTT proliferation assay (see, e.g., van de Loosdrechet, et al., 1994, J. Immunol. Methods 174: 311-320; Ohno et al., 1991, J. Immunol. Methods 145:199-203; Ferrari et al., 1990, J. Immunol. Methods 131: 165-172; Alley et al., 1988, Cancer Res. 48: 589-601; Carmichael et al., 1987, Cancer Res. 47:936-942; Gerlier et al., 1986, J. Immunol. Methods 65:55-63; Mosmann, 1983, J. Immunological Methods 65:55-63) is used to assay the effect of one or more agents in inhibiting the growth of cells. The cells are treated with chosen concentrations of one or more candidate agents for a chosen period of time, e.g., for 4 to 72 hours. The cells are then incubated with a suitable amount of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) for a chosen period of time, e.g., 1-8 hours, such that viable cells convert MTT into an intracellular deposit of insoluble formazan. After removing the excess MTT contained in the supernatant, a suitable MTT solvent, e.g., a DMSO solution, is added to dissolved the formazan. The concentration of MTT, which is proportional to the number of viable cells, is then measured by determining the optical density at e.g., 570 nm. A plurality of different concentrations of the candidate agent can be assayed to allow the determination of the concentrations of the candidate agent or agents which causes 50% inhibition.


In another preferred embodiment, an alamarBlue™ Assay for cell proliferation is used to screen for one or more candidate agents that can be used to inhibit the growth of cells (see, e.g., Page et al., 1993, Int. J. Oncol. 3:473-476). An alamarBlue™ assay measures cellular respiration and uses it as a measure of the number of living cells. The internal environment of proliferating cells is more reduced than that of non-proliferating cells. For example, the ratios of NADPH/NADP, FADH/FAD, FMNH/FMN, and NADH/NAF increase during proliferation. AlamarBlue can be reduced by these metabolic intermediates and, therefore, can be used to monitor cell proliferation. The cell number of a treated sample as measured by alamarBlue can be expressed in percent relative to that of an untreated control sample. alamarBlue reduction can be measured by either absorption or fluorescence spectroscopy. In one embodiment, the alamarBlue reduction is determined by absorbance and calculated as percent reduced using the equation:
%Reduced=(ɛoxλ2)(Aλ1)-(ɛoxλ1)(Aλ2)(ɛredλ1)(Aλ2)-(ɛredλ2)(Aλ1)×100(1)

where:

  • λ1=570 nm
  • λ2=600 nm
  • red λ1)=155,677 (Molar extinction coefficient of reduced alamarBlue at 570 nm)
  • red λ2)=14,652 (Molar extinction coefficient of reduced alamarBlue at 600 nm)
  • ox λ1)=80,586 (Molar extinction coefficient of oxidized alamarBlue at 570 nm)
  • ox λ2)=117,216 (Molar extinction coefficient of oxidized alamarBlue at 600 nm)
  • (A λ1)=Absorbance of test wells at 570 nm
  • (A λ2)=Absorbance of test wells at 600 nm
  • (A′λ1)=Absorbance of negative control wells which contain medium plus alamar Blue but to which no cells have been added at 570 nm.
  • (A′λ2)=Absorbance of negative control wells which contain medium plus alamar Blue but to which no cells have been added at 600 nm. Preferably, the % Reduced of wells containing no cell was subtracted from the % Reduced of wells containing samples to determine the % Reduced above background.


Cell cycle analysis can be carried out using standard method known in the art. In one embodiment, the supernatant from each well is combined with the cells that have been harvested by trypsinization. The mixture is then centrifuged at a suitable speed. The cells are then fixed with, e.g., ice cold 70% ethanol for a suitable period of time, e.g., ˜30 minutes. Fixed cells can be washed once with PBS and resuspended, e.g., in 0.5 ml of PBS containing Propidium Iodide (10 microgram/ml) and RNase A (1 mg/ml), and incubated at a suitable temperature, e.g., 37° C., for a suitable period of time, e.g., 30 min. Flow cytometric analysis is then carried out using a flow cytometer. In one embodiment, the Sub-G1 cell population is used as a measure of cell death. For example, the cells are said to have been sensitized to an agent if the Sub-G1 population from the sample treated with the agent is larger than the Sub-G1 population of sample not treated with the agent.


5.3. Uses of KSP Interacting Genes and their Products

The invention provides methods and compositions for utilizing a gene that interacts with KSP (“KSP interacting gene”), e.g., STK6 or TPX2 gene, its product and antibodies for identifying proteins or other molecules that interact with the KSP interacting gene or protein. In preferred embodiment, the invention provides STK6 or TPX2 gene as such KSP interacting gene. The invention also provides methods and compositions for utilizing the the KSP interacting gene, e.g., STK6 or TPX2 gene, product and antibodies for screening for agents that regulate expression of the KSP interacting gene or modulating interaction of the KSP interacting gene or protein with other proteins or molecules. The invention further provides methods and compositions for utilizing the KSP interacting gene, e.g., STK6 or TPX2 gene, product and antibodies for screening for agents that are useful in regulating resistance to the growth inhibitory effect of a KSP inhibitor (KSPi) and/or in enhancing the growth inhibitory effect of a KSP inhibitor in a cell or organism. The invention also provides methods and compositions for utilizing the KSP interacting gene, e.g., STK6 or TPX2 gene, product and antibodies for diagnosing resistance to the growth inhibitory effect of KSP inhibitors mediated by the KSP interacting gene, and for treatment of diseases in conjunction with a therapy using a KSP inhibitor.


5.3.1. Methods of Determining Proteins or Other Molecules that Interact with a KSP Interacting Gene or Its Product

Any method suitable for detecting protein-protein interactions may be employed for identifying interaction of a KSP interacting protein, e.g., STK6 or TPX2 protein, with another cellular protein. The interaction between a KSP interacting gene e.g., STK6 or TPX2 gene, and other cellular molecules, e.g., interaction between a KSP interacting gene and its regulators, may also be determined using methods known in the art.


Among the traditional methods which may be employed are co-immunoprecipitation, crosslinking and co-purification through gradients or chromatographic columns. Utilizing procedures such as these allows for the identification of cellular proteins which interact with a KSP interacting gene product. Once isolated, such a cellular protein can be identified and can, in turn, be used, in conjunction with standard techniques, to identify proteins it interacts with. For example, at least a portion of the amino acid sequence of the cellular protein which interacts with a KSP interacting gene product can be ascertained using techniques well known to those of skill in the art, such as via the Edman degradation technique (see, e.g., Creighton, 1983, “Proteins: Structures and Molecular Principles”, W.H. Freeman & Co., N.Y., pp. 34-49). The amino acid sequence obtained may be used as a guide for the generation of oligonucleotide mixtures that can be used to screen for gene sequences encoding such cellular proteins. Screening may be accomplished, for example, by standard hybridization or PCR techniques. Techniques for the generation of oligonucleotide mixtures and the screening are well-known. (See, e.g., Ausubel, supra., and PCR Protocols: A Guide to Methods and Applications, 1990, Innis, M. et al., eds. Academic Press, Inc., New York).


Additionally, methods may be employed which result in the simultaneous identification of genes which encode the cellular protein interacting with the KSP interacting protein. These methods include, for example, probing expression libraries with a labeled KSP interacting protein, using the KSP interacting protein in a manner similar to the well known technique of antibody probing of λgt11 libraries.


One method which detects protein interactions in vivo, the two-hybrid system, is described in detail for illustration only and not by way of limitation. One version of this system has been described (Chien et al., 1991, Proc. Natl. Acad. Sci. USA, 88:9578-9582) and is commercially available from Clontech (Palo Alto, Calif.).


Briefly, utilizing such a system, plasmids are constructed that encode two hybrid proteins: one consists of the DNA-binding domain of a transcription activator protein fused to a KSP interacting gene product and the other consists of the transcription activator protein's activation domain fused to an unknown protein that is encoded by a cDNA which has been recombined into this plasmid as part of a cDNA library. The DNA-binding domain fusion plasmid and the cDNA library are transformed into a strain of the yeast Saccharomyces cerevisiae that contains a reporter gene (e.g., HBS or lacZ) whose regulatory region contains the transcription activator's binding site. Either hybrid protein alone cannot activate transcription of the reporter gene: the DNA-binding domain hybrid cannot because it does not provide activation function and the activation domain hybrid cannot because it cannot localize to the activator's binding sites. Interaction of the two hybrid proteins reconstitutes the functional activator protein and results in expression of the reporter gene, which is detected by an assay for the reporter gene product.


The two-hybrid system or related methodology may be used to screen activation domain libraries for proteins that interact with the “bait” gene product. By way of example, and not by way of limitation, KSP interacting gene products may be used as the bait gene product. Total genomic or cDNA sequences are fused to the DNA encoding an activation domain. This library and a plasmid encoding a hybrid of a bait KSP interacting gene product fused to the DNA-binding domain are cotransformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene. For example, and not by way of limitation, a bait KSP interacting gene sequence, such as the coding sequence of a KSP interacting gene can be cloned into a vector such that it is translationally fused to the DNA encoding the DNA-binding domain of the GAL4 protein. These colonies are purified and the library plasmids responsible for reporter gene expression are isolated. DNA sequencing is then used to identify the proteins encoded by the library plasmids.


A cDNA library of the cell line from which proteins that interact with bait KSP interacting gene product are to be detected can be made using methods routinely practiced in the art. According to the particular system described herein, for example, the cDNA fragments can be inserted into a vector such that they are translationally fused to the transcriptional activation domain of GALA. This library can be co-transformed along with the bait KSP interacting gene-GALA fusion plasmid into a yeast strain which contains a lacZ gene driven by a promoter which contains GALA activation sequence. A cDNA encoded protein, fused to GALA transcriptional activation domain, that interacts with bait KSP interacting gene product will reconstitute an active GALA protein and thereby drive expression of the HIS3 gene. Colonies which express HIS3 can be detected by their growth on petri dishes containing semi-solid agar based media lacking histidine. The cDNA can then be purified from these strains, and used to produce and isolate the bait KSP interacting gene-interacting protein using techniques routinely practiced in the art.


The interaction between a KSP interacting gene and its regulators may be determined by a standard method known in the art.


5.3.2. Methods of Screening for Agents

The invention provides methods for screening for agents that regulate the expression or modulate interaction of a KSP interacting protein, e.g., STK6 or TPX2, with other proteins or molecules.


5.3.2.1. Screening Assays

The following assays are designed to identify compounds that bind to a KSP interacting gene or gene products, bind to other cellular proteins that interact with a KSP interacting gene product, bind to cellular constituents, e.g., proteins, that are affected by a KSP interacting gene product, or bind to compounds that interfere with the interaction of the KSP interacting gene or gene product with other cellular proteins and to compounds which modulate the activity of a KSP interacting gene (i.e., modulate the level of STK6 or TPX2 gene expression and/or modulate the activity level of a STK6 or TPX2 gene product). Assays may additionally be utilized which identify compounds which bind to a KSP interacting gene regulatory sequences (e.g., promoter sequences), see e.g., Platt, K. A., 1994, J. Biol. Chem. 269:28558-28562, which is incorporated herein by reference in its entirety, which may modulate the level of expression of a KSP interacting gene. Compounds may include, but are not limited to, small organic molecules which are able to affect expression of the KSP interacting gene or some other gene involved in the pathways involving the KSP interacting gene, or other cellular proteins. Methods for the identification of such cellular proteins are described, above, in Section 5.3.1. Such cellular proteins may be involved in the regulation of the growth inhibitory effect of a KSP inhibitor. Further, among these compounds are compounds which affect the level of expression of a KSP interacting gene and/or activity of its gene product and which can be used in the regulation of resistance to the growth inhibitory effect of a KSP inhibitor.


Compounds may include, but are not limited to, peptides such as, for example, soluble peptides, including but not limited to, Ig-tailed fusion peptides, and members of random peptide libraries; (see, e.g., Lam, K. S. et al., 1991, Nature 354:82-84; Houghten, R. et al., 1991, Nature 354:84-86), and combinatorial chemistry-derived molecular library made of D- and/or L-configuration amino acids, phosphopeptides (including, but not limited to members of random or partially degenerate, directed phosphopeptide libraries; see, e.g., Songyang, Z. et al., 1993, Cell 72:767-778), antibodies (including, but not limited to, polyclonal, monoclonal, humanized, anti-idiotypic, chimeric or single chain antibodies, and FAb, F(ab′)2 and FAb expression library fragments, and epitope-binding fragments thereof), and small organic or inorganic molecules.


Compounds identified via assays such as those described herein may be useful, for example, in regulating the biological function of the KSP interacting gene product, and for ameliorating resistance to the growth inhibitory effect of a KSP inhibitor and/or enhancing the growth inhibitory effect of a KSP inhibitor. Assays for testing the effectiveness of compounds are discussed, below, in Section 5.3.2.2.


In vitro systems may be designed to identify compounds capable of binding the KSP interacting gene products of the invention. Compounds identified may be useful, for example, in modulating the activity of wild type and/or mutant of KSP interacting gene products, may be useful in elaborating the biological function of the KSP interacting gene product, may be utilized in screens for identifying compounds that disrupt normal KSP interacting gene product interactions, or may in themselves disrupt such interactions.


The principle of the assays used to identify compounds that bind to a KSP interacting gene product involves preparing a reaction mixture of the KSP interacting gene product and the test compound under conditions and for a time sufficient to allow the two components to interact and bind, thus forming a complex which can be removed and/or detected in the reaction mixture. These assays can be conducted in a variety of ways. For example, one method to conduct such an assay would involve anchoring the KSP interacting gene product or the test substance onto a solid phase and detecting the KSP interacting gene product/test compound complexes anchored on the solid phase at the end of the reaction. In one embodiment of such a method, the KSP interacting gene product may be anchored onto a solid surface, and the test compound, which is not anchored, may be labeled, either directly or indirectly.


In practice, microtiter plates may conveniently be utilized as the solid phase. The anchored component may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished by simply coating the solid surface with a solution of the protein and drying. Alternatively, an immobilized antibody, preferably a monoclonal antibody, specific for the protein to be immobilized may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.


In order to conduct the assay, the nonimmobilized component is added to the coated surface containing the anchored component. After the reaction is complete, unreacted components are removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the previously nonimmobilized component is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the previously nonimmobilized component is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the previously nonimmobilized component (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).


Alternatively, a reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for a KSP interacting gene product or the test compound to anchor any complexes formed in solution, and a labeled antibody specific for the other component of the possible complex to detect anchored complexes.


The KSP interacting gene or gene products may interact in vivo with one or more intracellular or extracellular molecules, such as proteins. Such molecules may include, but are not limited to, nucleic acid molecules and those proteins identified via methods such as those described, above, in Section 5.3.1. For purposes of this discussion, such molecules are referred to herein as “binding partners”. Compounds that disrupt the binding of a KSP interacting gene product may be useful in regulating the activity of the KSP interacting gene product. Compounds that disrupt the binding of a KSP interacting gene product may be useful in regulating the expression of the KSP interacting gene, such as by regulating the binding of a regulator of KSP interacting gene. Such compounds may include, but are not limited to molecules such as peptides, and the like, as described, for example, in Section 5.3.2.1. above, which would be capable of gaining access to the KSP interacting gene product.


The basic principle of the assay systems used to identify compounds that interfere with the interaction between a KSP interacting gene product and its intracellular or extracellular binding partner or partners involves preparing a reaction mixture containing the KSP interacting gene product, and the binding partner under conditions and for a time sufficient to allow the two to interact and bind, thus forming a complex. In order to test a compound for inhibitory activity, the reaction mixture is prepared in the presence and absence of the test compound. The test compound may be initially included in the reaction mixture, or may be added at a time subsequent to the addition of the KSP interacting gene product and its binding partner. Control reaction mixtures are incubated without the test compound or with a placebo. The formation of any complexes between the KSP interacting protein and the binding partner is then detected. The formation of a complex in the control reaction, but not in the reaction mixture containing the test compound, indicates that the compound interferes with the interaction of the KSP interacting protein and the interactive binding partner. Additionally, complex formation within reaction mixtures containing the test compound and normal KSP interacting protein may also be compared to complex formation within reaction mixtures containing the test compound and a mutant KSP interacting protein. This comparison may be important in those cases wherein it is desirable to identify compounds that disrupt interactions of mutant but not normal KSP interacting proteins.


The assay for compounds that interfere with the interaction of the KSP interacting gene products and binding partners can be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring either the KSP interacting gene product or the binding partner onto a solid phase and detecting complexes anchored on the solid phase at the end of the reaction. In homogeneous assays, the entire reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested. For example, test compounds that interfere with the interaction between the KSP interacting gene products and the binding partners, e.g., by competition, can be identified by conducting the reaction in the presence of the test substance; i.e., by adding the test substance to the reaction mixture prior to or simultaneously with the KSP interacting protein and interactive binding partner. Alternatively, test compounds that disrupt preformed complexes, e.g. compounds with higher binding constants that displace one of the components from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed. The various formats are described briefly below.


In a heterogeneous assay system, either the KSP interacting gene product or the interactive binding partner, is anchored onto a solid surface, while the non-anchored species is labeled, either directly or indirectly. In practice, microtiter plates are conveniently utilized. The anchored species may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the KSP interacting gene product or binding partner and drying. Alternatively, an immobilized antibody specific for the species to be anchored may be used to anchor the species to the solid surface. The surfaces may be prepared in advance and stored.


In order to conduct the assay, the partner of the immobilized species is exposed to the coated surface with or without the test compound. After the reaction is complete, unreacted components are removed (e.g., by washing) and any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the non-immobilized species is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the non-immobilized species is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the initially non-immobilized species (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody). Depending upon the order of addition of reaction components, test compounds which inhibit complex formation or which disrupt preformed complexes can be detected.


Alternatively, the reaction can be conducted in a liquid phase in the presence or absence of the test compound, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for one of the binding components to anchor any complexes formed in solution, and a labeled antibody specific for the other partner to detect anchored complexes. Again, depending upon the order of addition of reactants to the liquid phase, test compounds which inhibit complex or which disrupt preformed complexes can be identified.


In an alternate embodiment of the invention, a homogeneous assay can be used. In this approach, a preformed complex of the KSP interacting protein and the interactive binding partner is prepared in which either the KSP interacting gene product or its binding partners is labeled, but the signal generated by the label is quenched due to complex formation (see, e.g., U.S. Pat. No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays). The addition of a test substance that competes with and displaces one of the species from the preformed complex will result in the generation of a signal above background. In this way, test substances which disrupt KSP interacting protein/binding partner interaction can be identified.


In a particular embodiment, the KSP interacting gene product can be prepared for immobilization using recombinant DNA techniques. For example, the coding region of a KSP interacting gene can be fused to a glutathione-5-transferase (GST) gene using a fusion vector, such as pGEX-5X-1, in such a manner that its binding activity is maintained in the resulting fusion protein. The interactive binding partner can be purified and used to raise a monoclonal antibody, using methods routinely practiced in the art. This antibody can be labeled with the radioactive isotope 125I, for example, by methods routinely practiced in the art. In a heterogeneous assay, the GST fusion protein, e.g., the GST-STK6 or GST-TPX2 fusion protein, can be anchored to glutathione-agarose beads. The interactive binding partner can then be added in the presence or absence of the test compound in a manner that allows interaction and binding to occur. At the end of the reaction period, unbound material can be washed away, and the labeled monoclonal antibody can be added to the system and allowed to bind to the complexed components. The interaction between the KSP interacting protein and the interactive binding partner can be detected by measuring the amount of radioactivity that remains associated with the glutathione-agarose beads. A successful inhibition of the interaction by the test compound will result in a decrease in measured radioactivity.


Alternatively, the fusion protein, e.g., the GST-STK6 gene fusion protein, and the interactive binding partner can be mixed together in liquid in the absence of the solid glutathione-agarose beads. The test compound can be added either during or after the species are allowed to interact. This mixture can then be added to the glutathione-agarose beads and unbound material is washed away. Again the extent of inhibition of the KSP interacting gene product/binding partner interaction can be detected by adding the labeled antibody and measuring the radioactivity associated with the beads.


In another embodiment of the invention, these same techniques can be employed using peptide fragments that correspond to the binding domains of the KSP interacting protein and/or the interactive binding partner (in cases where the binding partner is a protein), in place of one or both of the full length proteins. Any number of methods routinely practiced in the art can be used to identify and isolate the binding sites. These methods include, but are not limited to, mutagenesis of the gene encoding one of the proteins and screening for disruption of binding in a co-immunoprecipitation assay. Compensating mutations in the gene encoding the second species in the complex can then be selected. Sequence analysis of the genes encoding the respective proteins will reveal the mutations that correspond to the region of the protein involved in interactive binding. Alternatively, one protein can be anchored to a solid surface using methods described in this Section above, and allowed to interact with and bind to its labeled binding partner, which has been treated with a proteolytic enzyme, such as trypsin. After washing, a short, labeled peptide comprising the binding domain may remain associated with the solid material, which can be isolated and identified by amino acid sequencing. Also, once the gene coding for the binding partner is obtained, short gene segments can be engineered to express peptide fragments of the protein, which can then be tested for binding activity and purified or synthesized.


For example, and not by way of limitation, a STK6 or TPX2 gene product can be anchored to a solid material as described, above, in this Section by making a GST-STK6 or GST-TPX2 fusion protein and allowing it to bind to glutathione agarose beads. The interactive binding partner can be labeled with a radioactive isotope, such as 35S, and cleaved with a proteolytic enzyme such as trypsin. Cleavage products can then be added to the anchored GST-STK6 or GST-TPX2 fusion protein and allowed to bind. After washing away unbound peptides, labeled bound material, representing the binding partner binding domain, can be eluted, purified, and analyzed for amino acid sequence by well-known methods. Peptides so identified can be produced synthetically or fused to appropriate facilitative proteins using recombinant DNA technology.


5.3.2.2. Screening Compounds that Regulate and/or Enhance the Growth Inhibitory Effect of a KSP Inhibitor

Any agents that regulate the expression of a KSP interacting gene and/or the interaction of a KSP interacting protein with its binding partners, e.g., compounds that are identified in Section 5.3.2.1., antibodies to a KSP interacting protein, and so on, can be further screened for its ability to regulate and/or enhance the growth inhibitory effect of a KSP inhibitor in cells. Any suitable proliferation or growth inhibition assays known in the art can be used for this purpose. In one embodiment, a candidate agent and a KSP inhibitor are applied to cells of a cell line, and a change in growth inhibitory effect is determined. Preferably, changes in growth inhibitory effect are determined using different concentrations of the candidate agent in conjunction with different concentrations of the KSPi such that one or more combinations of concentrations of the candidate agent and KSPi which cause 50% inhibition, i.e., the IC50, are determined.


In a preferred embodiment, an MTT proliferation assay (see, e.g., van de Loosdrechet, et al., 1994, J. Immunol. Methods 174: 311-320; Ohno et al., 1991, J. Immunol. Methods 145:199-203; Ferrari et al., 1990, J. Immunol. Methods 131: 165-172; Alley et al., 1988, Cancer Res. 48: 589-601; Carmichael et al., 1987, Cancer Res. 47:936-942; Gerlier et al., 1986, J. Immunol. Methods 65:55-63; Mosmann, 1983, J. Immunological Methods 65:55-63) is used to screen for a candidate agent that can be used in conjunction with a KSPi to inhibit the growth of cells. The cells are treated with chosen concentrations of the candidate agent and a KSPi for 4 to 72 hours. The cells are then incubated with a suitable amount of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) for 1-8 hours such that viable cells convert MTT into an intracellular deposit of insoluble formazan. After removing the excess MTT contained in the supernatant, a suitable MTT solvent, e.g., a DMSO solution, is added to dissolved the formazan. The concentration of MTT, which is proportional to the number of viable cells, is then measured by determining the optical density at 570 nm. A plurality of different concentrations of the candidate agent can be assayed to allow the determination of the concentrations of the candidate agent and the KSPi which causes 50% inhibition.


In another preferred embodiment, an alamarBlue™ Assay for cell proliferation is used to screen for a candidate agent that can be used in conjunction with a KSPi to inhibit the growth of cells (see, e.g., Page et al., 1993, Int. J. Oncol. 3:473-476). AlamarBlue assay is described in Section 5.2., supra. In specific embodiment, the alamarBlue™ assay is performed to determine whether transfection titration curves of an siRNA targeting a KSP interacting gene were changed by the presence of a KSPi of a chosen concentration, e.g., 25 nM of the KSP inhibitor (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine. Cells were transfected with an STK6 siRNA. 4 hours after siRNA transfection, 100 microliter/well of DMEM/10% fetal bovine serum with or without the KSPi was added and the plates were incubated at 37° C. and 5% CO2 for 68 hours. The medium was removed from the wells and replaced with 100 microliter/well DMEM/10% Fetal Bovine Serum (Invitrogen) containing 10% (vol/vol) alamarBlue™ reagent (Biosource International Inc., Camarillo, Calif.) and 0.001 volumes of 1M Hepes buffer tissue culture reagent (Invitrogen). The plates were incubated for 2 hours at 37° C. before they were read at 570 and 600 nm wavelengths on a SpectraMax plus plate reader (Molecular Devices, Sunnyvale, Calif.) using Softmax Pro 3.1.2 software (Molecular Devices). The percent reduced for wells transfected with a titration of STK6 siRNA with or without 25 nM (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine were compared to luciferase siRNA-transfected wells. The number calculated for % Reduced for 0 nM luciferase siRNA-transfected wells without the KSPi was considered to be 100%.


5.3.2.3. Compounds Identified

The compounds identified in the screen include compounds that demonstrate the ability to selectively modulate the expression of a KSP interacting gene and regulate and/or enhance the growth inhibitory effect of a KSP inhibitor in cells. These compounds include but are not limited to siRNA, antisense, ribozyme, triple helix, antibody, and polypeptide molecules, aptamrs, and small organic or inorganic molecules.


The compounds identified in the screen also include compounds that modulate interaction of a KSP interacting with other proteins or molecules. In one embodiment, the compounds identified in the screen are compounds that modulate the interaction of a KSP interacting protein with its interaction partner. In another embodiment, the compounds identified in the screen are compounds that modulate the interaction of a KSP interacting gene with a transcription regulator.


5.3.3. Diagnostics

A variety of methods can be employed for the diagnostic and prognostic evaluation of cell or cells for their resistance to the growth inhibitory effect of a KSP inhibitor, e.g., (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine, resulting from defective regulation of a KSP interacting gene, e.g., STK6 or TPX2, and for the identification of subjects having a predisposition to resistance to the growth inhibitory effect of a KSP inhibitor.


In one embodiment, the method comprises determining an expression level of a KSP interacting gene in the cell, in which an expression level above a predetermined threshold level indicates that the cell is KSPi resistant. Preferably, the predetermined threshold level is at least 2-fold, 4-fold, 8-fold, or 10-fold of the normal expression level of the KSP interacting gene. In another embodiment, the invention provides a method for evaluating KSPi resistance in a cell comprising determining a level of abundance of a protein encoded by a KSP interacting gene in the cell, in which a level of abundance of the protein above a predetermined threshold level indicates that the cell is KSPi resistant. In still another embodiment, the invention provides a method for evaluating KSPi resistance in a cell comprising determining a level of activity of a protein encoded by a KSP interacting gene in cells of the mammal, in which an activity level above a predetermined threshold level indicates that the cell is KSPi resistant. Preferably, the predetermined threshold level of abundance or activity is at least 2-fold, 4-fold, 8-fold, or 10-fold of the normal level of abundance or activity of the KSP interacting protein.


Such methods may, for example, utilize reagents such as the KSP interacting gene nucleotide sequences and antibodies directed against KSP interacting gene products, including peptide fragments thereof. Specifically, such reagents may be used, for example, for: (1) the detection of the presence of mutations in a KSP interacting gene, or the detection of either over- or under-expression of an mRNA of a KSP interacting gene relative to the normal expression level; and (2) the detection of either an over- or an under-abundance of a KSP interacting gene product relative to the normal level of a KSP interacting protein.


The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one specific KSP interacting gene nucleic acid or anti-KSP interacting protein antibody reagent described herein, which may be conveniently used, e.g., in clinical settings, to diagnose patients exhibiting disorder or abnormalities related to a KSP interacting gene.


For the detection of mutations in a KSP interacting gene, any nucleated cell can be used as a starting source for genomic nucleic acid. For the detection of the expression of a KSP interacting gene or KSP interacting gene products, any cell type or tissue in which the KSP interacting gene is expressed may be utilized.


Nucleic acid-based detection techniques are described, below, in Section 5.3.3.1. Peptide detection techniques are described, below, in Section 5.3.3.2.


5.3.3.1. Detection of Expression of a KSP Interacting Gene

The expression of a KSP interacting gene, e.g., STK6 or TPX2, in cells or tissues, e.g., the cellular level of KSP interacting gene transcripts and/or the presence or absence of mutations, can be detected by utilizing a number of techniques. Nucleic acid from any nucleated cell can be used as the starting point for such assay techniques, and may be isolated according to standard nucleic acid preparation procedures which are well known to those of skill in the art. For example, the expression level of the KSP interacting gene can determined by measuring the expression level of the KSP interacting gene using one or more polynucleotide probes, each of which comprises a nucleotide sequence in the KSP interacting gene. In particularly preferred embodiments of the invention, the method is used to diagnose resistance of a cancer to a treatment using KSPi in a human.


DNA may be used in hybridization or amplification assays of biological samples to detect abnormalities involving the structure of a KSP interacting gene, including point mutations, insertions, deletions and chromosomal rearrangements. Such assays may include, but are not limited to, Southern analyses, single stranded conformational polymorphism analyses (SSCP), DNA microarray analyses, and PCR analyses.


Such diagnostic methods for the detection of KSP interacting gene-specific mutations can involve, for example, contacting and incubating nucleic acids including recombinant DNA molecules, cloned genes or degenerate variants thereof, obtained from a sample, e.g., derived from a patient sample or other appropriate cellular source, with one or more labeled nucleic acid reagents including recombinant DNA molecules, cloned genes or degenerate variants thereof, under conditions favorable for the specific annealing of these reagents to their complementary sequences within the KSP interacting gene. Preferably, the lengths of these nucleic acid reagents are at least 15 to 30 nucleotides. After incubation, all non-annealed nucleic acids are removed from the nucleic acid: KSP interacting gene molecule hybrid. The presence of nucleic acids which have hybridized, if any such molecules exist, is then detected. Using such a detection scheme, the nucleic acid from the cell type or tissue of interest can be immobilized, for example, to a solid support such as a membrane, or a plastic surface such as that on a microtiter plate or polystyrene beads. In this case, after incubation, non-annealed, labeled nucleic acid reagents are easily removed. Detection of the remaining, annealed, labeled KSP interacting gene nucleic acid reagents is accomplished using standard techniques well-known to those in the art. The KSP interacting gene sequences to which the nucleic acid reagents have annealed can be compared to the annealing pattern expected from a normal KSP interacting gene sequence in order to determine whether a KSP interacting gene mutation is present.


Alternative diagnostic methods for the detection of a KSP interacting gene specific nucleic acid molecules, in patient samples or other appropriate cell sources, may involve their amplification, e.g., by PCR (the experimental embodiment set forth in Mullis, K. B., 1987, U.S. Pat. No. 4,683,202), followed by the detection of the amplified molecules using techniques well known to those of skill in the art. The resulting amplified sequences can be compared to those which would be expected if the nucleic acid being amplified contained only normal copies of the KSP interacting gene in order to determine whether a KSP interacting gene mutation exists.


Among the nucleic acid sequences of a KSP interacting gene which are preferred for such hybridization and/or PCR analyses are those which will detect the presence of the KSP interacting gene splice site mutation.


Additionally, well-known genotyping techniques can be performed to identify individuals carrying a mutation in a KSP interacting gene. Such techniques include, for example, the use of restriction fragment length polymorphisms (RFLPs), which involve sequence variations in one of the recognition sites for the specific restriction enzyme used. Additionally, improved methods for analyzing DNA polymorphisms which can be utilized for the identification of mutations in a KSP interacting gene have been described which capitalize on the presence of variable numbers of short, tandemly repeated DNA sequences between the restriction enzyme sites. For example, Weber (U.S. Pat. No. 5,075,217, which is incorporated herein by reference in its entirety) describes a DNA marker based on length polymorphisms in blocks of (dC-dA)n-(dG-dT)n short tandem repeats. The average separation of (dC-dA)n-(dG-dT)n blocks is estimated to be 30,000-60,000 bp. Markers which are so closely spaced exhibit a high frequency co-inheritance, and are extremely useful in the identification of genetic mutations, such as, for example, mutations within the KSP interacting gene, and the diagnosis of diseases and disorders related to mutations in the KSP interacting.


Also, Caskey et al. (U.S. Pat. No. 5,364,759, which is incorporated herein by reference in its entirety) describe a DNA profiling assay for detecting short tri and tetra nucleotide repeat sequences. The process includes extracting the DNA of interest, such as the KSP interacting gene, amplifying the extracted DNA, and labelling the repeat sequences to form a genotypic map of the individual's DNA.


The expression level of a KSP interacting gene can also be assayed. For example, RNA from a cell type or tissue known, or suspected, to express the KSP interacting gene, such as a cancer cell type which exhibits KSPi resistance, may be isolated and tested utilizing hybridization or PCR techniques such as are described, above. The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be a necessary step in the assessment of cells to be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the KSP interacting gene. Such analyses may reveal both quantitative and qualitative aspects of the expression pattern of the KSP interacting gene, including activation or inactivation of the expression of the KSP interacting gene.


In one embodiment of such a detection scheme, a cDNA molecule is synthesized from an RNA molecule of interest (e.g., by reverse transcription of the RNA molecule into cDNA). A sequence within the cDNA is then used as the template for a nucleic acid amplification reaction, such as a PCR amplification reaction, or the like. The nucleic acid reagents used as synthesis initiation reagents (e.g., primers) in the reverse transcription and nucleic acid amplification steps of this method are chosen from among the KSP interacting gene nucleic acid reagents. The preferred lengths of such nucleic acid reagents are at least 9-30 nucleotides. For detection of the amplified product, the nucleic acid amplification may be performed using radioactively or non-radioactively labeled nucleotides. Alternatively, enough amplified product may be made such that the product may be visualized by utilizing any suitable nucleic acid staining method, e.g., by standard ethidium bromide staining.


Additionally, it is possible to perform such KSP interacting gene expression assays “in situ”, i.e., directly upon tissue sections (fixed and/or frozen) of patient tissue obtained from biopsies or resections, such that no nucleic acid purification is necessary. Nucleic acids from a KSP interacting gene may be used as probes and/or primers for such in situ procedures (see; for example, Nuovo, G. J., 1992, “PCR In Situ Hybridization: Protocols And Applications”, Raven Press, NY).


Alternatively, if a sufficient quantity of the appropriate cells can be obtained, standard Northern analysis can be performed to determine the level of mRNA expression of the KSP interacting gene.


The expression of KSP interacting gene in cells or tissues, e.g., the cellular level of KSP interacting transcripts and/or the presence or absence of mutations, can also be evaluated using DNA microarray technologies. In such technologies, one or more polynucleotide probes each comprising a sequence of the KSP interacting gene are used to monitor the expression of the KSP interacting gene. The present invention therefore provides DNA microarrays comprising polynucleotide probes comprising sequences of the KSP interacting gene.


Any formats of DNA microarray technologies can be used in conjunction with the present invention. In one embodiment, spotted cDNA arrays are prepared by depositing PCR products of cDNA fragments, e.g., full length cDNAs, ESTs, etc., of the KSP interacting gene onto a suitable surface (see, e.g., DeRisi et al., 1996, Nature Genetics 14:457-460; Shalon et al., 1996, Genome Res. 6:689-645; Schena et al., 1995, Proc. Natl. Acad. Sci U.S.A. 93:10539-11286; and Duggan et al., Nature Genetics Supplement 21:10-14). In another embodiment, high-density oligonucleotide arrays containing oligonucleotides complementary to sequences of the KSP interacting gene are synthesized in situ on the surface by photolithographic techniques (see, e.g., Fodor et al., 1991, Science 251:767-773; Pease et al., 1994, Proc. Natl. Acad. Sci. U.S.A. 91:5022-5026; Lockhart et al., 1996, Nature Biotechnology 14:1675; McGall et al., 1996, Proc. Natl. Acad. Sci. U.S.A. 93:13555-13560; U.S. Pat. Nos. 5,578,832; 5,556,752; 5,510,270; 5,858,659; and 6,040,138). This format of microarray technology is particular useful for detection of single nucleotide polymorphisms (SNPs) (see, e.g., Hacia et al., 1999, Nat Genet. 22:164-7; Wang et al., 1998, Science 280:1077-82). In yet another embodiment, high-density oligonucleotide arrays containing oligonucleotides complementary to sequences of the KSP interacting gene are synthesized in situ on the surface by inkjet technologies (see, e.g., Blanchard, International Patent Publication WO 98/41531, published Sep. 24, 1998; Blanchard et al., 1996, Biosensors and Bioelectronics 11:687-690; Blanchard, 1998, in Synthetic DNA Arrays in Genetic Engineering, Vol. 20, J. K. Setlow, Ed., Plenum Press, New York at pages 111-123). In still another embodiment, DNA microarrays that allow electronic stringency control can be used in conjunction with polynucleotide probes comprising sequences of the KSP interacting gene (see, e.g., U.S. Pat. No. 5,849,486).


5.3.3.2. Detection of KSP Interacting Gene Products

Antibodies directed against wild type or mutant KSP interacting gene products or conserved variants or peptide fragments thereof may be used as diagnostics and prognostics of KSPi resistance, as described herein. Such diagnostic methods may be used to detect abnormalities in the expression level of a KSP interacting gene, or abnormalities in the structure and/or temporal, tissue, cellular, or subcellular location of a KSP interacting gene product.


Because KSP interacting gene products are intracellular gene products, the antibodies and immunoassay methods described below have important in vitro applications in assessing the efficacy of treatments for disorders resulting from defective regulation of KSP interacting gene such as proliferative diseases. Antibodies, or fragments of antibodies, such as those described below, may be used to screen potentially therapeutic compounds in vitro to determine their effects on KSP interacting gene expression and KSP interacting peptide production. The compounds which have beneficial effects on disorders related to defective regulation of KSP interacting can be identified, and a therapeutically effective dose determined.


In vitro immunoassays may also be used, for example, to assess the efficacy of cell-based gene therapy for disorders related to defective regulation of a KSP interacting gene. Antibodies directed against KSP interacting peptides may be used in vitro to determine the level of KSP interacting gene expression achieved in cells genetically engineered to produce KSP interacting peptides. Given that evidence disclosed herein indicates that the KSP interacting gene product is an intracellular gene product, such an assessment is, preferably, done using cell lysates or extracts. Such analysis will allow for a determination of the number of transformed cells necessary to achieve therapeutic efficacy in vivo, as well as optimization of the gene replacement protocol.


The tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the KSP interacting gene, such as, a KSPi resistant cancer cell type. The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.), which is incorporated herein by reference in its entirety. The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be used to test the effect of compounds on the expression of the KSP interacting gene.


Preferred diagnostic methods for the detection of KSP interacting gene products or conserved variants or peptide fragments thereof, may involve, for example, immunoassays wherein the KSP interacting gene products or conserved variants or peptide fragments are detected by their interaction with an anti-KSP interacting gene product-specific antibody.


For example, antibodies, or fragments of antibodies, that bind a KSP interacting protein, may be used to quantitatively or qualitatively detect the presence of KSP interacting gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody (see below, this Section) coupled with light microscopic, flow cytometric, or fluorimetric detection. Such techniques are especially preferred if such KSP interacting gene products are expressed on the cell surface.


The antibodies (or fragments thereof) useful in the present invention may, additionally, be employed histologically, as in immunofluorescence or immunoelectron microscopy, for in situ detection of KSP interacting gene products or conserved variants or peptide fragments thereof. In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody of the present invention. The antibody (or fragment) is preferably applied by overlaying the labeled antibody (or fragment) onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the KSP interacting gene product, or conserved variants or peptide fragments, but also its distribution in the examined tissue. Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.


Immunoassays for KSP interacting gene products or conserved variants or peptide fragments thereof will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of identifying KSP interacting gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.


The biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled KSP interacting protein specific antibody. The solid phase support may then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on solid support may then be detected by conventional means.


By “solid phase support or carrier” is intended any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention. The support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to an antigen or antibody. Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tub, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc. Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.


The binding activity of a given lot of anti-KSP interacting gene product antibody may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.


One of the ways in which the KSP interacting gene peptide-specific antibody can be detectably labeled is by linking the same to an enzyme and use in an enzyme immunoassay (EIA) (Voller, A., “The Enzyme Linked Immunosorbent Assay (ELISA)”, 1978, Diagnostic Horizons 2:1-7, Microbiological Associates Quarterly Publication, Walkersville, Md.); Voller, A. et al., 1978, J. Clin. Pathol. 31:507-520; Butler, J. E., 1981, Meth. Enzymol. 73:482-523; Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC Press, Boca Raton, Fla.,; Ishikawa, E. et al., (eds.), 1981, Enzyme Immunoassay, Kgaku Shoin, Tokyo). The enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. The detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.


Detection may also be accomplished using any of a variety of other immunoassays. For example, by radioactively labeling the antibodies or antibody fragments, it is possible to detect KSP interacting peptides through the use of a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March 1986, which is incorporated by reference herein). The radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.


It is also possible to label the antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.


The antibody can also be detectably labeled using fluorescence emitting metals such as 152Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).


The antibody also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.


Likewise, a bioluminescent compound may be used to label the antibody of the present invention. Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.


5.3.4. Methods of Regulating Expression of KSP Interacting Genes

A variety of therapeutic approaches may be used in accordance with the invention to modulate expression of a KSP interacting gene, e.g., STK6 or TPX2, in vivo. For example, siRNA molecules may be engineered and used to silence the KSP interacting gene in vivo. Antisense DNA molecules may also be engineered and used to block translation of a KSP interacting mRNA in vivo. Alternatively, ribozyme molecules may be designed to cleave and destroy the KSP interacting mRNAs in vivo. In another alternative, oligonucleotides designed to hybridize to the 5′ region of the KSP interacting gene (including the region upstream of the coding sequence) and form triple helix structures may be used to block or reduce transcription of the KSP interacting gene. If desired, oligonucleotides can also be designed to hybridize and form triple helix structures with the binding site of a negative regulator so as to block the binding of the negative regulator and to enhance the transcription of the KSP interacting gene.


In a preferred embodiment, siRNA, antisense, ribozyme, and triple helix nucleotides are designed to inhibit the translation or transcription of one or more KSP interacting protein isoforms with minimal effects on the expression of other genes that may share one or more sequence motif with the KSP interacting gene. To accomplish this, the oligonucleotides used should be designed on the basis of relevant sequences unique to the KSP interacting gene.


For example, and not by way of limitation, the oligonucleotides should not fall within those region where the nucleotide sequence of a KSP interacting gene is most homologous to that of the other genes. In the case of antisense molecules, it is preferred that the sequence be chosen from the list above. It is also preferred that the sequence be at least 18 nucleotides in length in order to achieve sufficiently strong annealing to the target mRNA sequence to prevent translation of the sequence. Izant et al., 1984, Cell, 36:1007-1015; Rosenberg et al., 1985, Nature, 313:703-706.


In the case of the “hammerhead” type of ribozymes, it is also preferred that the target sequences of the ribozymes be chosen from the list above. Ribozymes are RNA molecules which possess highly specific endoribonuclease activity. Hammerhead ribozymes comprise a hybridizing region which is complementary in nucleotide sequence to at least part of the target RNA, and a catalytic region which is adapted to cleave the target RNA. The hybridizing region contains nine (9) or more nucleotides. Therefore, the hammerhead ribozymes of the present invention have a hybridizing region which is complementary to the sequences listed above and is at least nine nucleotides in length. The construction and production of such ribozymes is well known in the art and is described more fully in Haseloff et al., 1988, Nature, 334:585-591.


The ribozymes of the present invention also include RNA endoribonucleases (hereinafter “Cech-type ribozymes”) such as the one which occurs naturally in Tetrahymena Thermophila (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224:574-578; Zaug and Cech, 1986, Science, 231:470-475; Zaug, et al., 1986, Nature, 324:429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been et al., 1986, Cell, 47:207-216). The Cech endoribonucleases have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.


In the case of oligonucleotides that hybridize to and form triple helix structures at the 5′ terminus of the KSP interacting gene and can be used to block transcription, it is preferred that they be complementary to those sequences in the 5′ terminus of KSP interacting gene which are not present in the other genes whose expression level is not to be affected. It is also preferred that the sequences do not include those regions of the promoter of a KSP interacting gene which are even slightly homologous to that of such other genes. The foregoing compounds can be administered by a variety of methods which are known in the art including, but not limited to the use of liposomes as a delivery vehicle. Naked DNA or RNA molecules may also be used where they are in a form which is resistant to degradation such as by modification of the ends, by the formation of circular molecules, or by the use of alternate bonds including phosphothionate and thiophosphoryl modified bonds. In addition, the delivery of nucleic acid may be by facilitated transport where the nucleic acid molecules are conjugated to poly-lysine or transferrin. Nucleic acid may also be transported into cells by any of the various viral carriers, including but not limited to, retrovirus, vaccinia, AAV, and adenovirus.


Alternatively, a recombinant nucleic acid molecule which encodes, or is, such antisense, ribozyme, triple helix, or KSP interacting gene nucleic acid molecule can be constructed. This nucleic acid molecule may be either RNA or DNA. If the nucleic acid encodes an RNA, it is preferred that the sequence be operatively attached to a regulatory element so that sufficient copies of the desired RNA product are produced. The regulatory element may permit either constitutive or regulated transcription of the sequence. In vivo, that is, within the cells or cells of an organism, a transfer vector such as a bacterial plasmid or viral RNA or DNA, encoding one or more of the RNAs, may be transfected into cells e.g. (Llewellyn et al., 1987, J. Mol. Biol., 195:115-123; Hanahan et al. 1983, J. Mol. Biol., 166:557-580). Once inside the cell, the transfer vector may replicate, and be transcribed by cellular polymerases to produce the RNA or it may be integrated into the genome of the host cell. Alternatively, a transfer vector containing sequences encoding one or more of the RNAs may be transfected into cells or introduced into cells by way of micromanipulation techniques such as microinjection, such that the transfer vector or a part thereof becomes integrated into the genome of the host cell.


RNAi can also be used to knock down the expression of a KSP interacting gene. In one embodiment, double-stranded RNA molecules of 21-23 nucleotides which hybridize to a homologous region of mRNAs transcribed from the KSP interacting gene are used to degrade the mRNAs, thereby “silence” the expression of the KSP interacting gene. Preferably, the dsRNAs have a hybridizing region, e.g., a 19-nucleotide double-stranded region, which is complementary to a sequence of the coding sequence of the KSP interacting gene. Any siRNA targeting an appropriate coding sequence of a KSP interacting gene, e.g., a human STK6 or TPX2 gene, can be used in the invention. As an exemplary embodiment, 21-nucleotide double-stranded siRNAs targeting the coding regions of KSP interacting gene are designed according to standard selection rules (see, e.g., Elbashir et al., 2002, Methods 26:199-213, which is incorporated herein by reference in its entirety).


Any standard method for introducing siRNAs into cells can be used. In one embodiment, gene silencing is induced by presenting the cell with the siRNA targeting the KSP interacting gene (see, e.g., Elbashir et al., 2001, Nature 411, 494-498; Elbashir et al., 2001, Genes Dev. 15, 188-200, all of which are incorporated by reference herein in their entirety). The siRNAs can be chemically synthesized, or derived from cleavage of double-stranded RNA by recombinant Dicer. Another method to introduce a double stranded DNA (dsRNA) for silencing of the KSP interacting gene is shRNA, for short hairpin RNA (see, e.g., Paddison et al., 2002, Genes Dev. 16, 948-958; Brummelkamp et al., 2002, Science 296, 550-553; Sui, G. et al. 2002, Proc. Natl. Acad. Sci. USA 99, 5515-5520, all of which are incorporated by reference herein in their entirety). In this method, an siRNA targeting a KSP interacting gene is expressed from a plasmid (or virus) as an inverted repeat with an intervening loop sequence to form a hairpin structure. The resulting RNA transcript containing the hairpin is subsequently processed by Dicer to produce siRNAs for silencing. Plasmid-based shRNAs can be expressed stably in cells, allowing long-term gene silencing in cells both in vitro and in vivo (see, McCaffrey et al. 2002, Nature 418, 38-39; Xia et al., 2002, Nat. Biotech. 20, 1006-1010; Lewis et al., 2002, Nat. Genetics 32, 107-108; Rubinson et al., 2003, Nat. Genetics 33, 401-406; Tiscomia et al., 2003, Proc. Natl. Acad. Sci. USA 100, 1844-1848, all of which are incorporated by reference herein in their entirety). SiRNAs targeting the KSP interacting gene can also be delivered to an organ or tissue in a mammal, such a human, in vivo (see, e.g., Song et al. 2003, Nat. Medicine 9, 347-351; Sorensen et al., 2003, J. Mol. Biol. 327, 761-766; Lewis et al., 2002, Nat. Genetics 32, 107-108, all of which are incorporated by reference herein in their entirety). In this method, a solution of siRNA is injected intravenously into the mammal. The siRNA can then reach an organ or tissue of interest and effectively reduce the expression of the target gene in the organ or tissue of the mammal.


5.3.5. Methods of Regulating Activity of a KSP Interacting Protein and/or Its Pathways

The activity of a KSP interacting protein can be regulated by modulating the interaction of the KSP interacting protein with its binding partners. In one embodiment, agents, e.g., antibodies, aptamers, small organic or inorganic molecules, can be used to inhibit binding of such a binding partner such that KSPi resistance is regulated. In another embodiment, agents, e.g., antibodies, aptamers, small organic or inorganic molecules, can be used to inhibit the activity of a protein in a KSP interacting protein regulatory pathway such that KSPi resistance is regulated.


5.3.6. Cancer Therapy by Targeting KSP Interacting Gene and/or Gene Product

The methods and/or compositions described above for modulating expression and/or activity of a KSP interacting gene or protein, e.g., STK6 or TPX2 gene or protein, may be used to treat patients who have a cancer in conjunction with a KSPi. In particular, the methods and/or compositions may be used in conjunction with a KSPi for treatment of a patient having a cancer which exhibits the KSP interacting gene or protein mediated KSPi resistance. Such therapies may be used to treat cancers, including but not limted to, rhabdomyosarcoma, neuroblastoma and glioblastoma, small cell lung cancer, osteoscarcoma, pancreatic cancer, breast and prostate cancer, murine melanoma and leukemia, and B-cell lymphoma.


In preferred embodiments, the methods and/or compositions of the invention are used in conjunction with a KSPi for treatment of a patient having a cancer which exhibits STK6 or TPX2 mediated KSPi resistance. In such embodiments, the expression and/or activity of STK6 or TPX2 are modulated to confer cancer cells sensitivity to a KSPi, thereby conferring or enhancing the efficacy of KSPi therapy.


In a combination therapy, one or more compositions of the present invention can be administered before, at the same time of, or after the administration of a KSPi. In one embodiment, the compositions of the invention are administered before the administration a KSPi. The time intervals between the administration of the compositions of the invention and a KSPi can be determined by routine experiments that are familiar to one skilled person in the art. In one embodiment, a KSPi is given after the KSP interacting protein level reaches a desirable threshold. The level of KSP interacting protein can be determined by using any techniques described supra.


In another embodiment, the compositions of the invention are administered at the same time with the KSPi.


In still another embodiment, one or more of the compositions of the invention are also administered after the administration of a KSPi. Such administration can be beneficial especially when the KSPi has a longer half life than that of the one or more compositions of the invention used in the treatment.


It will be apparent to one skilled person in the art that any combination of different timing of the administration of the compositions of the invention and a KSPi can be used. For example, when the KSPi has a longer half life than that of the composition of the invention, it is preferable to administer the compositions of the invention before and after the administration of the KSPi.


The frequency or intervals of administration of the compositions of the invention depends on the desired level of the KSP interacting protein, which can be determined by any of the techniques described supra. The administration frequency of the compositions of the invention can be increased or decreased when the KSP interacting protein level changes either higher or lower from the desired level.


The effects or benefits of administration of the compositions of the invention alone or in conjunction with a KSPi can be evaluated by any methods known in the art, e.g., by methods that are based on measuring the survival rate, side effects, dosage requirement of the KSPi, or any combinations thereof. If the administration of the compositions of the invention achieves any one or more of the benefits in a patient, such as increasing the survival rate, decreasing side effects, lowing the dosage requirement for the KSPi, the compositions of the invention are said to have augmented the KSPi therapy, and the method is said to have efficacy.


5.3.7. Cancer Therapy by Targeting STK6 Gene in Combination with Other Drugs that Target Mitosis

The inventors have also discovered that STK6 also interacts with other drugs that target mitosis, e.g., taxol. FIG. 18 shows that STK6 sensitize HeLa cells to taxol treatment. Thus, the invention also provides methods and compositions described above for modulating STK6 expression and/or activity for treating patients who have a cancer in conjunction with a drug that targets mitosis, e.g., taxol. In particular, the methods and/or compositions may be used in conjunction with taxol for treatment of a patient having a cancer which exhibits STK6-mediated taxol resistance. Such therapies may be used to treat cancers, including but not limted to, rhabdomyosarcoma, neuroblastoma and glioblastoma, small cell lung cancer, osteoscarcoma, pancreatic cancer, breast and prostate cancer, murine melanoma and leukemia, and B-cell lymphoma.


5.4. Genes and Gene Products Interacting with a DNA Damaging Agent and Their Uses

The invention provides methods and compositions for utilizing the genes and gene products that interact with DNA damaging agents in treating diseases. Such a gene is often referred to as a “DNA damage response gene.” A gene product, e.g., a protein, encoded by such a gene is often referred to as a “DNA damage response gene product.” The invention also provides methods and compositions for utilizing these genes and their products for screening for agents that regulate the expression/activity of the genes/gene products, and/or modulating interaction of the genes or proteins with other proteins or molecules. The invention further provides methods and compositions for utilizing these genes and gene products for screening for agents that are useful in regulating sensitivity of cells to the growth inhibitory effect of DNA damaging agents and/or in enhancing the growth inhibitory effect of DNA damaging agent in a cell or organism. The invention also provides methods and compositions for utilizing these gene and gene products for diagnosing resistance or sensitivity to the growth inhibitory effect of DNA damaging agents, and for treatment of diseases in conjunction with a therapy using one or more DNA damaging agents.


5.4.1. Genes and Gene Products Interacting with a DNA Damaging Agent

The invention provides genes that are capable of reducing or enhancing cell killing by DNA damaging agents. These genes can be used in conjunction with the DNA damaging agents described in Section 5.4.2., infra. Uses of these genes are described in Sections 5.4.3 and 5.4.4., infra.


In one embodiment, the invention provides genes that are capable of reducing or enhancing cell killing by a DNA damaging agent, e.g., cis, dox, or campto, by at least 1.5 fold, 1.6 fold, 1.7 fold, 1.8 fold, and 1.9 fold. In a preferred embodiment, the invention provides the following genes whose silencing enhances cell killing by a DNA damaging agent by at least 2.0 fold: BRCA2, EPHB3, WEE1, and ELK1. FIG. 8 shows that silencing of BRCA2, EPHB3, WEE1, and ELK1 enhances cell killing due to a DNA damaging agent by at least 2 fold. The invention provides method of treatment of cancer by regulating, e.g., enhancing or reducing, the expression of such genes and/or activity of a protein encoded by such genes, in conjunction with a therapy involving administration of a DNA damaging agent.


The invention also provides genes that are capable of reducing or enhancing cell killing by a particular type of DNA damaging agents. Table IIA shows genes whose silencing enhances or reduces cell killing by a DNA binding agent such as DNA groove binding agent, e.g., DNA minor groove binding agent; DNA crosslinking agent; intercalating agent; and DNA adduct forming agent. In one embodiment, the invention provides genes whose silencing enhances cell killing by a DNA binding agent, e.g., cis, by at least 1.5 fold, 1.6 fold, 1.7 fold, 1.8 fold, and 1.9 fold as listed in Table IIA, e.g., gene IDs 752-806 (1.5 fold), gene IDs 771-806 (1.6 fold), gene IDs 784-806 (1.7 fold), gene IDs 789-806 (1.8 fold), and gene IDs 793-806 (1.9 fold). In a preferred embodiment, the invention provides following genes whose silencing enhances cell killing by a DNA binding agent, e.g., cis, by at least 2 fold: BRCA1, BRCA2, EPHB3, WEE1, ELK1, RPS6KA6, BRAF, GPRK6, MCM3, CDC42, KIF2C, CENPE, CDC25B, and C20orf97. In another embodiment, the invention provides following genes whose silencing reduces cell killing by a DNA binding agent, e.g., cis, by at least 2 fold: PLK (see FIG. 16). The invention provides method of treatment of cancer by regulating, e.g., enhancing or reducing, the expression of such genes and/or activity of a protein encoded by such genes, in conjunction with a therapy involving administration of a DNA binding agent.


The invention also provides genes that are capable of reducing or enhancing cell killing by Topo I inhibitor, such as camptothecin. In one embodiment, the invention provides genes whose silencing enhances cell killing by a topo I inhibitor, e.g., campto, by at least 1.5 fold, 1.6 fold, 1.7 fold, 1.8 fold, and 1.9 fold as listed in Table IIB, e.g., gene IDs 635-807 (1.5 fold), gene IDs 673-807 (1.6 fold), gene IDs 702-807 (1.7 fold), gene IDs 727-807 (1.8 fold), and gene IDs 749-807 (1.9 fold). In a preferred embodiment, the invention provides genes whose silencing enhances cell killing by a Topo I inhibitor, e.g., campto, by at least 2 fold, e.g., NM139286, TOP3B, WASL, STAT4, CHEK1, BCL2, NM016263, TOP2B, TGFBR1, MAPK8, RHOK, NM017719, TERT, ANAPC5, NM021170, SGK2, C20orf97, CSF1R, EGR2, AATK, TCF3, CDC45L, STAT3, PRKY, BMPR1B, KIF2C, PTTG1, NM019089, FOXO1A, STK4, SRC, ELK1, NM018492, RASA2, GPRK6, BLK, ABL1, HSPCB, PRKACA, CCNE2, CTNNBIP1, NM013367, FRAT1, PIK3C2A, NM017769, XM170783, NM016457, XM064050, STK6, RALBP1, ELK1, NF1, STAT5A, WEE1, PTK6, RPS6KA6, BRCA1, EPHB3, and BRCA2. In another preferred embodiment, the invention provides genes whose silencing enhances cell killing by a Topo I inhibitor, e.g., campto, by at least 3 fold, e.g., XM064050, STK6, RALBP1, ELK1, NF1, STAT5A, WEE1, PTK6, RPS6KA6, BRCA1, EPHB3, and BRCA2. In another embodiment, the invention provides genes whose silencing reduces cell killing by a topo I inhibitor, e.g., campto, by at least 2 fold, e.g., PLK, CCNA2, MADH4, NFKB1, RRM2B, TSG101, DCK, CDC5L, CDCA8, NM006101, INSR. The invention provides method of treatment of cancer by regulating, e.g., enhancing or reducing, the expression of such genes and/or activity of a protein encoded by such genes, in conjunction with a therapy involving administration of a Topo I inhibitor.


The invention also provides genes that are capable of reducing or enhancing cell killing by Topo II inhibitor, such as doxorubicin. In one embodiment, the invention provides genes whose silencing enhances cell killing by a DNA binding agent, e.g., dox, by at least 1.5 fold, 1.6 fold, 1.7 fold, 1.8 fold, and 1.9 fold as listed in Table IIC, e.g., gene IDs 657-830 (1.5 fold), gene IDs 685-830 (1.6 fold), gene IDs 723-830 (1.7 fold), gene IDs 750-830 (1.8 fold), and gene IDs 767-830 (1.9 fold). In a preferred embodiment, the invention provides genes whose silencing enhances cell killing by a Topo II inhibitor, e.g., dox, by at least 2 fold, e.g., PTK2, KRAS2, BRA, FZD4, RASAL2, CENPE, CCNH, MAP4K3, MAP4K2, ERBB3, RHOK, MYO3A, AXIN1, INPP5D, NM018401, NEK1, TGFBR1, XM064050, STAT4, MAP3K1, CCNE2, STK6, HDAC4, CTNNA1, EIF4EBP1, ACVR2B, CDC42, MAPK8, BLK, WEE1, KIF26A, TCF1, NM019089, NOTCH4, HDAC3, PIK3CB, CCNG2, TLK2, XM066649, MCM3, ELK1, PTK6, ABL1, FZD4, XM170783, CHUK, SRC, NM016263, and C20orf97. In another preferred embodiment, the invention provides genes whose silencing enhances cell killing by a Topo II inhibitor, e.g., dox, by at least 3 fold, e.g., ELK1, PTK6, ABL1, FZD4, XM170783, CHUK, SRC, NM016263, and C20orf97. In another embodiment, the invention provides genes whose silencing reduces cell killing by a Topo II inhibitor, e.g., dox, by at least 2 fold, e.g., PLK (see FIG. 16). The invention provides method of treatment of cancer by regulating, e.g., enhancing or reducing, the expression of such genes and/or activity of a protein encoded by such genes, in conjunction with a therapy involving administration of a Topo II inhibitor.


In a preferred embodiment, the invention provides CHEK1, BRCA1, BARD1, and RAD51 as genes that are capable of enhancing killing of p53− cells by DNA damaging agents.


In another preferred embodiment, the invention provides WEE1 as a gene that is capable of reducing or enhancing cell killing by DNA damaging agents. Wee1 is a negative mitosis regulator protein first identified in fission yeast Schizosaccharmomyces pombe (Russell and Nurse, 1987 Cell 49:559-67). Wee1 mutants have a short G2 period and enter mitosis at half the size (hence the name wee) of wild type cells. In cells that overexpress cdc25, a mitotic inducer, wee1 activity is required to prevent lethality by premature mitosis (mitotic catastrophe). The human homolog of wee1 was cloned by transcomplementation of a S. pombe temperature-dependent wee1−1, cdc25 over-expressing mutant (Igarashi et al., 1991, Nature 353:80-83). Overexpression of the human wee1 in fission yeast generates elongated cells from inhibition of the G2-M transition of the cell cycle. This human Wee1 clone was significantly smaller than its yeast counterpoint, and was later found to be missing a portion of the amino terminus sequence (Watanabe et al., 1995, EMBO 14:1878-91).


The single copy human wee1 gene is located on chromosome 11 (Taviaux and Demaille, 1993, Genomics 15:194-196). The wee1 gene is 16.96 kb with 11 exons, encoding a 4.23 kb mRNA transcript. The 94 kDa human Wee1 protein comprises 646 amino acids. According to Aceview, an integrated analysis of publicly available experimental cDNA data (http://www.ncbi.nlm.nih.gov/IEB/Research/Acembly/av.cgi?c=locusid&org=9606&1=7465) there may be six smaller Wee1 protein isoforms produced by alternative splicing. Wee1 expression has been found in wide range of human cells, such as lung fibroblasts, embryonic fibroblasts, cervical cancer HeLa cells, colon adenocarcinoma, bladder carcinoma (Igarashi et al., 1991, Nature 353:80-83), uterine, blood vessel, liver, eye, spleen, gall bladder, skin, cartilage, and various tumor cell lines (UniGene, http://www.ncbi.nlm.nih.gov/UniGene/). Wee1-like proteins have also been identified in mouse, rat, C. elegans, Drosphila, and S. cerevisiae, with the mouse and rat 646 amino acid proteins having the highest degree of similarity (89% and 91% respectively) (UniGene). Full-length human Wee1 sequence has five stretches with high PEST scores, and the catalytic kinase domain is in the C-terminus (Watanabe et al., 1995, EMBO 14:1878-91). The conserved Lys114 residue appears to be critical for Wee1 kinase activity (McGowan and Russell, 1993, EMBO 12:75-85).


Other Wee1-related kinases have been identified in multiple species. Xenopus Wee1 is expressed maternally (oocytes), while Wee2 is expressed in zygotes in non-dividing tissue. In vertebrates, the related Myt1 has similar phosphorylating activity to Wee1 (reviewed in Kellogg, 2003, J. Cell Sci. 116:4883-4890). A Wee1B has also been identified in humans, which is almost exclusively expressed in mature oocytes (Nakanishi et al., 2000, Genes to Cells 5:839-847).


Wee1 is a nuclear tyrosine kinase belonging to the family of Ser/Thr family of protein kinases. Wee1 ensures the completion of DNA replication prior to mitosis by inhibiting Cdc2-cyclin B kinase at the G2/M transition of the cell cycle. Phosphorylation of the Thr14 and Tyr15 residues in the ATP-binding site of Cdc2 inhibits its activity; Wee1 tyrosine kinase phosphorylates the Tyr15 residue at the N-terminus. A second related protein kinase, Mik1 (Myt1), phosphorylates Cdc2 on both Thr14 and Tyr15. Cdc2 activity is required for progression into mitosis. Dephosphorylation of the critical Tyr15 residue is catalyzed by Cdc25, functioning in opposition to Wee1. Balance of Wee1 and Cdc25 activities determines entry into mitosis (reviewed in Kellogg, 2003, J. Cell Sci. 116:4883-4890; Pendergast, 1996, Curr. Opin. Cell Biol. 8:174-181).


Wee1 activity is highly regulated during the cell cycle. During S and G2 phases, Wee1 activity increases, paralleling increases in protein levels. Wee1 activity is suppressed at mitosis as a result of hyperphosphorylation and degradation of Wee1 (Watanabe et al., 1995, EMBO 14:1878-91; McGowan and Russell, 1993, EMBO 12:75-85). Recent work in Xenopus and fission yeast has demonstrated that Cdk1 (Cdc2) can phosphorylate Wee1, suggesting a positive-feedback loop model in which a small amount of mitotic Cdk1 inactivates Wee1, and subsequently triggers a significant increase in mitotic Cdk1. Tome-1 also promotes mitotic entry by targeting Wee1 for proteolytic destruction by SCF in G2 phase. APC CDH allows Wee1 reinstatement in S phase by destruction of Tome-1 and cyclin B during G1 phase (reviewed by Lim and Surana, 2003, Mol. Cell 11:845-851).


A new role has also been suggested for Wee1 in apoptosis. Crk, which has been implicated in apoptosis in Xenopus, can bind with Wee1 via its SH2 domain. Exogenous Wee1 accelerated Xenopus egg apoptosis in a Crk dependent manner (Smith et al., 2000, J. Cell Biol. 151:1391-1400). These Crk-Wee1 complexes, in the absence of nuclear export factor Crm1 binding, also promoted apoptosis in mammalian cells (Smith et al, 2002, Mol. Cell. Biol. 22:1412-1423). Studies involving the HIV protein R (Vpr) have also involved Wee1 in apoptotic events (Yuan, et al., 2003, J. Virol. 77:2063-2070). Vpr causes G2 arrest which is associated with Cdc2 inactivation, and prolonged G2 arrest leads to apoptosis. Wee1 was depleted in Vpr induced apoptotic HeLa cells and gamma-irradiated apoptotic HeLa cells. Overexpression of Wee1 attenuated Vpr-induced apoptosis, and depletion of Wee1 by siRNA induced apoptotic death. The apparent conflict between Wee1 levels and apoptotic events in these studies, and the mechanisms of apoptosis induction by Wee1 have not been elucidated.


The role of cell cycle inhibitors is important if DNA is damaged. The block in cell division allows time for DNA repair and minimizes the replication and segregation of damaged DNA. The two cell cycle “checkpoints” for genetic integrity are at the G1 phase (before DNA synthesis) and G2 phase (just before mitosis). Loss of these checkpoint controls facilitates the evolution of cells into cancer (reviewed by Hartwell and Kastan, 1994, Science 266:1821-8).


Defective Wee1 expression may abrogate the G2 checkpoint, facilitating tumor cell proliferation. Wee1 has been found to be significantly suppressed in colon carcinoma cells (reviewed by Lee and Yang, 2001, Cell. Mol. Life Sci. 58:1907-1922). Absence of Wee1 expression was also associated with poorer prognosis and higher recurrency of non-small-cell lung cancer (Yoshida et al., 2004, Ann. Onco. 15:252-256).


In contrast, Wee1 levels and kinase activity was also elevated in hepatocellular carcinoma compared to the surrounding cirrhotic tissue (Masaki et al., 2003, Hepatology 37:534-543).


Alternatively, abrogation of the G2 checkpoint may enhance chemotherapy against G1 checkpoint defective tumor cells. Many tumor cells lack a functional p53 gene, and do not demonstrate a G1 checkpoint. While normal cells would arrest at G1 after DNA damage from irradiation or chemotherapy, the cancer cells would rely upon G2 checkpoint for DNA repair. Abrogation of the G2 checkpoint would therefore be more detrimental to cancer cells than normal cells. A chemical library screen for compounds which selectively inhibit Wee1 has been used to search for anti-cancer agents which inhibit G2 checkpoint because of Wee1's negative regulation of Cdc2 and Wee1's attenuation of apoptosis (Wang et al., 2001, Cancer Res. 61:8211-8217). PD0166285 Wee1 kinase inhibitor demonstrated inhibition of Cdc2 phosphorylation, abrogation of G2 arrest, and sensitized killing of p53 mutant cell lines by radiation. In one embodiment, the invention provides a method of treating a cancer using PD166285 in conjunction with a DNA damaging agent.


Wee1 activation may also be involved in the pathology of rheumatoid arthritis. Growth of rheumatoid synovial cells is tumor-like; cells possess abundant cytoplasm, large nuclei, and karyotypic changes. These transformed cells are found in the cartilage and bone of human RA and animal models. Rheumatoid synovial cell growth is disorganized and anchorage-independent. C-Fos/Ap-1 trasncription factor was increased in rheumatoid synovium. Kawasaki et al. (Kawasaki et al., 2003, Onco. 22:6839-6844) demonstrated that Wee1 is transactivated by c-Fos/AP-1; c-Fos and Wee1 was significantly increased in rheumatoid synovial cells compared to osteoarthritis cells. These synovial cells also displayed increased tetraploidy. Inactivating Wee1 may alleviate some of the joint destruction that occurs in RA.


U.S. 20030087847 A1 describes a method for using nucleic acids molecules to inhibit Chk1 activity, as a way to abrogate the G2 checkpoint and selectively sensitive p53 deficient tumors to chemotherapy. Chk1 phosphorylates an inhibitory residue on Cdc25, which is an activator of Cdc2. EP1360281 A2 describes Wee1 nucleotide and amino acid sequences, methods for expression of recombinant Wee1, and identifying compounds that modulate Wee1 activity.


In another preferred embodiment, the invention provides EPHB3 as a gene that is capable of reducing or enhancing cell killing by DNA damaging agents. Receptor tyrosine kinases (RTK) are membrane spanning proteins with an extra-cellular ligand binding domain and intracellular kinase domain. With 14 members, the Eph receptors comprise the largest subfamily of RTK. The extracellular region of The extracellular portion of Eph receptors is composed of a putative immunoglobulin (Ig) region (ligand binding domain), followed by a cysteine-rich region, and two fibronectin type III repeats near the single transmembrane segment (Connor and Pasquale, 1995 Oncogene 11:2429-2438; Labrador et al., 1997, EMBO 16:3889-3897). The cytoplasmic portion contains a highly conserved tyrosine kinase domain flanked by a juxtamembrane region and a C-terminal tail (sterile a motif and PDZ-binding motif), which are less conserved. Eph receptors are divided into two groups based on the sequence homologies of their extracellular domains. The EphA receptors interact with high affinity to ephrin-A ligands, which are tethered to the cell surface by a glycosylphophatidylinositol (GPI) anchor. EphB receptors preferentially bind the transmembrane ephrin-B ligands. With each group, receptors can bind to more than one ligand, and each ligand can bind to more than one receptor. There is less receptor-ligand cross-talk between the A and B subgroups (reviewed in Orioli and Klein, 1997 Trends in Genetics 13:354-359; Pasquale, 1997 Curr. Biol. 9:608-615). Eph receptors can only be activated by membrane-bound or artificially-clustered ephrins; while soluble ligands do bind the receptors, they do not trigger receptor autophosphorylation (Davis et al., 1994 Science 266: 816-819). Eph receptors and ephrins are unique in that they mediate bi-directional signaling. Due to their membrane-bound states, Eph receptors and ephrins are thought to mediated cell-to-cell interactions rather than long-range functions.


Expression of the Eph receptors is distinct, but overlapping, suggesting unique but redundant functions. Expression of Eph receptors is highest in the nervous tissue, but can be found in numerous tissues. Expression is higher in the developing embryo, but is also present in adult tissues. Receptor-ligand interactions often result in cell repulsion, and these repulsive effects have been implicated in axonal guidance, synapse formation, segmental patterning of the nervous system, angiogenesis, and cell migration in development. These receptors may also be involved in neural cells, angiogenesis, and tumorigenesis in adults (reviewed in Dodelet and Pasquale, 2000 Oncogene 19:5614-5619; Zhou, 1998 Pharmacol. Ther. 77:151-181; Pasquale, 1997 Curr. Opin. Cell Biol. 9:608-615). Cellular repulsion or de-adhesion appears to be mediated through interaction between the Eph receptor and numerous signaling molecules such as Nck, Ras-GAP, Src, SHEP1, and SHP2 (Wilkinson, 2001 Neurosci. Rev. 2:155-164).


There are eight EphA receptors (EphA1-8) and six EphB (EphB1-6) receptors, all of which encode a protein of about 1000 amino acids. Eph genes have been identified in a number of species such as chicken, rat, mouse, and human. EphB3, also known as Hek2, Sek4, Mdk5, Cek10, or Tyro 6, can interact with ligands ephrin-B1-3 (Pasquale, 1997, Curr. Opin. Cell Biol. 9:608-615). EphB3 sequences are highly conserved among different species (>95% amino acid homology). The single copy 20.2 kb EphB3 gene is located on human chromosome 3 and has 16 exons. The human protein consists of 998 amino acids (ref. seq. NM004443). High levels of mouse EphB3 transcripts are found throughout embryonic development and in adult brain, intestine, placenta, muscle, heart, and with lesser intensity lung and kidney (Ciossek et al., 1995 Oncogene 11:2085-2095). EphB3 transcripts were found in adult human brain, lung, pancreas, liver, placenta, kidney, skeletal muscle, and heart (Bohme et al, 1993 Oncogene 8:2857-2862).


An EphB3 splice variant has been identified in the chicken, which has a 15 amino acid insertion in the juxtamembrane domain (Sajjadi and Pasquale, 1993 Oncogene 8:1807-1813). In addition to the major 4.8 kb full-length EphB3 transcript, smaller 2.8 kb, 2.3 kb, and 1.9 kb transcripts were found in mouse tissues (Ciossek et al., 1995 Oncogene 11:2085-2095). Only one transcript size has been observed thus far in human EphB3 (Bohme et al., Oncogene 1993 8:2857-2862). However, a human EphB2 splice variant has been identified, suggesting that additional isoforms of other human Eph receptors may be found (Tang et al., 1998 Oncogene 17:521-526).


Considerable characterization of Eph receptors has been done in embryo development. Adams et al. (Genes & Dev. 13:295-306), showed that EphB3 is expressed in the yolk sacs and developing arteries and veins of embryonic mice. They also demonstrated that EphB2/EphB3 double mutant mice display defects in yolk sac vascularization, extended pericardial sacs, defective vascular development, and defective angiogenesis of the head, heart, and somites. Adams et al. also determined that ephrin-B ligands are able to induce capillary sprouting in an in vitro assay.


EphB3 deficient mice implicate the receptor's involvement in the formation of brain commissures, specifically the corpus callosum which connects the two cerebral hemispheres. Furthermore EphB2/EphB3 double mutants have cleft palates, suggesting their involving in facial development as well (Orioli et al., 1996 EMBO 15:6035-6049).


Within the intestinal epithelium, stem cells produce precursors that migrate in specific patterns as they differentiate. Mutational activation of β-catenin/TCF in intestinal epithelial cells results in polyp formation. Batle et al. showed that β-catenin/TCF signaling events control EphB3 expression in colorectal cancer cells and along the crypt-villus axis. In EphB3 null mice, Paneth cells, which normally migrate to occupy the bottom of the intestinal crypts, were randomly localized throughout the crypt, suggesting a deficiency in sorting cell populations. Furthermore, in EphB2/EphB3 double mutants, proliferative and differentiated cells intermingled in the intestinal epithelium (Batle et al., 2002 Cell 111:251-263).


EphB3 expression has also been found in adult mouse cochlea, suggesting a possible role in the peripheral auditory system. EphB3 knockout mice exhibited significantly lower distortion-product otoacoustic emissions DPOAE levels compared to wild type controls (Howard et al., 2003 Hear. Res. 178:118-130). DPOAE measurements reflect cochlear function at the level of outer hair cells.


Willson et al. demonstrated upregulation of EphB3 expression in the injured spinal cords of adult rats, at the injury site (Willson et al., 2003, Cell Transpl. 12:279-290). Expression of EphB3 receptors was co-localized in regions of the CNS which also had a high level of ephrin B ligands. The complementary expression of both EphB3 receptor and ligand at the site of injury may contribute to an environment that inhibits axonal regeneration after injury.


EphB3 has been detected in tumor cell lines of breast and epidermoid origin (Bohme et al., 1993, Oncogene 8:2857-2862). Expression levels of other Eph receptors are upregulated in various tumor types as well (reviewed in Dodelet and Pasquale, Oncogene 2000 19:5614-5619). Some evidence suggests that upregulation of Eph receptors does not appear to drive proliferation (Lhotak and Pawson, 1993, Mol. Cell. Biol. 13:7071-7079), but rather elevated expression appears to correlate with metastatic potential (Andres et al., 1994 Oncogene 1461-1467; Vogt et al., 1998 Clin. Cancer Res. 4:791-797).


Tissue disorganization and abnormal cell adhesion are hallmarks of advanced tumors. Overexpression Eph receptors may make tumors highly sensitive to ephrin activation, promoting decreased cell adhesion, cell motility, and invasiveness. Eph receptors have been found to influence cell-matrix attachment by modulating integrin activity. Maio et al. (2000 Nature Cell. Biol. 2:62-69) has shown that activation of EphA2 with the ephrinA1 ligand on prostate carcinoma cells transiently inhibits integrin-mediated cell attachment. Additionally, in early Xenopus embryos, ectopic expression of ephrin-B1 or activated EphA4 interfered with cadherin dependent cell attachment (Jones et al, 1998 Proc. Natl. Acad. Sci. USA 95:576-581; Winning et al, 1996 Dev. Biol., 179:309-319).


Links between Eph receptors and cytoskeletal changes, a key aspect of cellular motility, have also been established. Activation of EphB4 by ephrin-B2 ligand induces Rac-mediated membrane ruffling in Eph expressing cells (Marston et al., 2003 Nat. Cell Biol. 5:879-888). Wahl et al. (2000 J. Cell Biol. 149:263-270) has demonstrated that ephrin-A5 induces collapse of neural growth cones in a Rho-dependent manner. Both Rho and Rac have been implicated in the cellular changes involved in a tumor formation (reviewed in Schmitz et al., 2000 Exp. Cell Res. 261:1-12). Activation of these signaling pathways by Eph receptors may contribute to tumor invasion and metastasis.


Given the role of Eph receptors and their ligands in embryonic vascular development, and angiogenesis (reviewed in Sullivan and Bicknell, 2003 Br. J. Cancer 89:228-231), these molecules may also be involved in tumor growth by contributing to vascularization of tumors. Eph receptor ligands have been shown to promote organization and assembly of endothelial cells into capillary structures, and to induce capillary sprouting from existing blood vessels (Daniel et al., 1996 Kidney Intl. Suppl. 57:S73-81; Pandey et al., 1995 Science 268:567-569). Secreted ephrin ligands may also act as diffusible chemoattractants for endothelial cells; eph receptors expressed on tumor cells may guide the construction of new vessels from incoming endothelial cells (Pandey et al., 1995 Science 268:567-569).


Because of its upregulation in tumor cells (Bohme et al., 1993 Oncogene 8:2857), and its potential involvement in tumor angiogenesis and metastasis, EphB3 may make an attractive target for cancer diagnosis or therapeutic intervention. Soluble EphA-Fc receptors inhibited tumor angiogenesis in cutaneous window assays and in vivo in mice which were injected with 4T1 tumor cells Brantley et al, 2002 Oncogene 21:7011-7026).


Alternatively, there may be situations where enhancement of the angiogenesis properties of Eph receptors may be desirable, such as for treatment for coronary vessel blockage.


The expression of EphB3 in injured spinal cords may also serve as an attractive therapeutic target for CNS injury. The cell repulsive effects of EphB3 may contribute to inability of injured spinal cord axons to regrow. Studies have demonstrated axonal regrowth in the injured spinal cord when other molecules inhibitory for axonal regeneration are blocked by antibodies (Bregman et al., 1995 Nature 378:498-501; GrandPre et al., 2002 Nature 417:547-551).


Eph receptor autophosphorylation is a key event for subsequent interaction with other signaling molecules with SH2 of phosphotyrosine binding domains (reviewed in Bruckner et al, 1998 Curr. Opion. Neuro. 8:375-382).


Binns et al. (Binns, et al., 2000, Mol. Cell. Biol. 20:4791-4805) describes a cellular assay system for studying ephrin-stimulation of EphB2 on neuronal cells. Briefly, an NG108-15 cell line stably expressing EphB2 (NG-EphB2WT cells) was established. NG108-15 cells display characteristics of motor neurons, a cell type which expresses EphB2 during embryonic development. NG108-15 cells, however, do not endogenously express EphB2 or respond to ephrin-B ligands. Stimulation of NG-EphB2WT cells with Fc-ephrin-B1 results in neurite retraction and disassembly of polymerized actin structures. Wildtype NG108-15 cells and cells expressing tyrosine-to-phenylalanine substitutions (key phosphorylation sites) in the juxtamembrane motif do not exhibit the cytoskeletal remodeling in response to ligand stimulation. Variation in phosphorylation of tyrosine residues in wt EphB2 vs. EphB2(Y→F) transformed cells was also monitored with anti-p Tyr antibodies. Decreased EphB2 receptor function also resulted in decreased phosphorylation of p62dok, a component of the eph signaling cascade.


U.S. Pat. No. 6,169,167 also describes methods of determining hek4 activation with Hek4 ligands using a cell-cell autophosphorylation assay. Following receptor-ligand interaction, Hek4 receptors are immunoprecipitated from lysates of CHO cells expressing Hek4 DNA. The lysates are used in Western blots with anti-phosphotyrosine antibodies.


In still another preferred embodiment, the invention provides RAD51 as a gene that is capable of reducing or enhancing cell killing by DNA damaging agents. In mammalian cells, double strand DNA breaks (DSBs) can be repaired by non-homologous end joining (NHEJ) or by homologous recombination. NHEJ involves the re-ligation of broken DNA ends without a template and may result in mutations or deletions at the break site. Homologous recombination requires a template, an intact sister duplex, and results in high fidelity repair. Homologous recombination can also repair stalled or broken replication forks in DNA. Repair of DSBs is vital as impaired function or apoptosis may occur if they are left undone or repaired inaccurately. Genetic instability, a key characteristic of tumor cells, may also result without the high fidelity of homologous recombinational repair. The initial steps of homologous recombination, homologous pairing and strand exchange, involve a protein belonging to the RecA/Rad51 recombinase family (reviewed in Baumann and West, 1998, Trends Biochem. Sci. 23:247-251; Henning and Stürzbecher, 2003, Toxicology 193:91-109).


The E. coli protein RecA acts as a regulator of the SOS response to DNA damage and promotes homologous pairing and strand exchange (reviewed in Baumann and West, 1998, Trends Biochem. Sci. 23:247-251). A DSB repair gene rad51 was identified in Saccharomyces cerevisiae and is homologous to recA (Shinohara et al., 1992, Cell 69:457-470). The rad51 gene was also cloned from human and mouse (Yoshimura et al., 1993, Nucleic Acids Res. 21:1665; Shinohara et al., 1993, Nature Genet. 4:239-243). The single copy human rad51 gene is located on chromosome 15 (Shinohara et al, 1993, Nature Genet. 4:239-243). The rad51 gene consists of 10 exons, encoding a 339 amino acid protein. The amino acid sequence of the two mammalian Rad51 proteins is 83% homologous to the yeast Rad51, and 56% homologous to the E. coli RecA protein. The regions of homology between RecA and Rad51 include functional domains for recombination, UV resistance, and oligomer formation (positions 31-260 of RecA) (Yoshimura et al., 1993, Nucleic Acids Res. 21:1665; Shinohara et al., 1993, Nature Genet. 4:239-243). Mouse Rad51 transcripts were found at high levels in thymus, spleen, testis, and ovary, and at lower levels in the brain (Shinohara et al, 1993, Nature Genet. 4:239-243). Rad51 expression also appears to be cell cycle regulated, with transcriptional upregulation at S and G2 phases (Flygare et al., 1996, Biochim. Biophys. Acta 1312:231-236). Additionally, five Rad51 paralogs have been identified (XRCC2, XRCC3, Rad51B-D) that have 20-30% identity with Rad51. These paralogs may promote Rad51 focus formation (reviewed in Thompson and Schild, 2001, Mutat. Res. 477:131-153).


Rad51 functions as a long helical polymer that wraps around DNA to form a nucleoprotein filament. Rad51 binds to single stranded DNA produced by nucleolytic resection at the DSB site, and this interaction is enhanced by Rad52. Invasion of a re-sected end of the DSB into a homologous duplex occurs in the Rad51 nucleoprotein filament, requiring ATP-binding but not hydrolysis. The second re-sected end is also captured by Rad51. The invading re-sected ends function as primers for DNA re-synthesis. Holliday-junction resolution and ligation allow the repaired duplexes to separate (reviewed by West, 2003, Nat. Rev. Mol. Cell. Biol. 4:435-445). Pellegrini et al. (2002, Nature 420:287-293) reported that a conserved repeat sequence in BRCA2, BRC4, mimics a motif in Rad51 and serves as an interface for oligomerization of Rad51 monomers. Through this BRC4-Rad51-complex, BRCA2 is able to control the assembly of the Rad51 nucleoprotein filament. Rad51 activity is also regulated by other mechanisms. P53 has been found to down-modulate homologous recombination promoted by Rad51 (Linke et al., 2003, Cancer Res. 63:2596-2605; Yoon et al., 2004, J. Mol. Biol. 336:639-654). Rad54 has been found to disassemble Rad51 nucleoprotein filaments formed on double stranded DNA (dsDNA) and may be involved in turnover of Rad51-dsDNA filaments, which is important during DNA strand exchange reactions. In yeast, Srs2 has been found to inhibit recombination by disrupting Rad51 filament formation on single stranded DNA (Veaute et al., 2003, Nature 423:309-312; Krejci et al., 2003, Nature 423:305-309).


Splice variants of Rad51 have been identified. One transcript (NM133487) lacks an internal segment corresponding to exons 4, 5 and the 5′ portion of exon 6, resulting in a protein that lacks an internal region of 97 amino acids. The transcript identified by the Genbank accession number AY425955 also suggests the existence of a further truncated splice variant in testis. Rad51 splice variants have also been found in other species, such as C. elegans (Rinaldo et al., 1998, Mol. Gen. Genet. 260:289-294).


A couple of studies have demonstrated that a Rad51 135C polymorphism significantly elevates the risk of breast cancer in carriers of BRCA2 but not BRCA1 (Levy-Lahad et al., 2001, Proc. Natl. Acad. Sci. USA 98:3232-3236; Kadouri et al., 2004, Br. J. Cancer 90:2002-2005). A missense mutation (Gln150Arg) was reported in two patients with bilateral breast cancer, but otherwise, Rad51 mutations were not found in most tumors (Kato et al., 2000, J. Hum. Genet. 45:133-137; Schmutte et al., 1999, Cancer Res. 59:4564-4569). Rad51 knockout mice die early during embryonic development, though heterozygotes are viable and fertile, and rad51−/− mouse cell lines could not be established, indicating an essential role for this gene (Tsuzuki et al., 1996, Proc. Natl. Acad. Sci. USA 93:6236-6240). Sonoda et al. (1998, EMBO J., 17:598-608) generated a rad51−/− chicken B lymphocyte DT40 cell line by using a Rad51 transgene controlled by a repressible promoter. Inhibition of the rad51 transgene in DT40 cells resulted in high levels of chromosome breakage, cell cycle arrest at the G2/M phase, and cell death. Several studies have also investigated Rad51 overexpression in cell lines. Vispe et al. (1998, Nucleic Acids Res. 26:2859-2864) found that Rad51 overexpression in CHO cells resulted in a 20-fold increase in homologous recombination between two adjacent homologous alleles and increased resistance to ionizing radiation in the late S/G2 cell cycle phase. Work done by Richardson et al. (2004, Oncogene 23:546-553) presents evidence for a link between increased levels of Rad51 in tumor cells and chromosomal instability associated with tumor progression. Rad51 levels transiently upregulated 2-4-fold during induction of DSB in a mouse ES cell line produced novel recombinational repair products and generation of abnormal karyotypes.


Elevated Rad51 levels have been reported in tumors, suggesting that Rad51 up-regulation may confer an advantage to tumor progression. Maacke et al. (2000, Int. J. Cancer 88:907-913) reported a positive correlation between Rad51 overexpression and breast tumor grading. A 2-7-fold increase of Rad51 was also observed in a wide range of tumor cell lines compared to nonmalignant control cell lines (Raderschall et al., 2002, Cancer Res. 62:219-225). Rad51 overexpression was also found in 66% of human pancreatic adenocarcinoma tissue samples (Maacke et al., 2000, Oncogene 19:2791-2795). It is speculated that Rad51 overexpression in cancer cells may protect cells from DNA damage or contribute to genomic instability and diversity. Elevated expression of Rad51 and increased recombination was also observed during immortalization of human fibroblasts (Xia et al., 1997, Mol. Cell Biol. 17:7151-7158).


A number of studies have suggested a functional role for Rad51 in tumor resistance. Hansen et al. (2003, Int. J. Cancer 105:472-479) demonstrated that Rad51 levels positively correlated with etoposide resistance in small cell lung cancer (SCLC) cells. Furthermore, down or upregulation of Rad51 using sense or antisense constructs altered etoposide sensitivity in SCLC cells. Chlorambucil treatment was found to induce Rad51 expression in B-cell chronic lymphocytic leukemia cells (Christodoulopoulos et al., 1999, Clin. Cancer Res. 5:2178-2184). Antisense Rad51 oligonucleotides enhanced DNA damage by irradiation in both a mouse embryonic skin cell line and malignant gliomas (Taki et al., 1996, Biochem. Biophys. Res. Commun. 223:434-438; Ohnishi et al., 1998, Biochem. Biophys. Res. Commun. 245:319-324). Downregulation of Rad51 with ribozymes also increased the sensitivity of prostate cancer cells to irradiation (Collis et al., 2001, Nucleic Acids Res. 29:1534-1538). Disruption of Rad51 function through its interaction with BRC repeats on BRCA2 also leads to radiation and methyl methanesulfonate hypersensitivity in cancer cells (Chen et al., 1999, J. Biol. Chem. 274:32931-32935; Chen et al., 1998, Proc. Natl. Acad. Sci. USA 95:5287-5292). Slupianek et al. (2001, Mol. Cell 8:795-806) showed that Bcr/Abl regulation of Rad51 expression is important for cisplatin and mitomycin C resistance in myeloid cells. These studies suggest Rad51 as an attractive target to improve the efficacy of cancer therapy.


5.4.2. DNA Damaging Agents

The invention can be practiced with any known DNA damaging agent, including but are not limited to any topoisomerase inhibitor, DNA binding agent, anti-metabolite, ionizing radiation, or a combination of two or more of such known DNA damaging agents.


A topoisomerase inhibitor that can be used in conjunction with the invention can be a topoisomerase I (Topo I) inhibitor, a topoisomerase II (Topo II) inhibitor, or a dual topoisomerase I and II inhibitor. A topo I inhibitor can be from any of the following classes of compounds: camptothecin analogue (e.g., karenitecin, aminocamptothecin, lurtotecan, topotecan, irinotecan, BAY 56-3722, rubitecan, G114721, exatecan mesylate), rebeccamycin analogue, PNU 166148, rebeccamycin, TAS-103, camptothecin (e.g., camptothecin polyglutamate, camptothecin sodium), intoplicine, ecteinascidin 743, J-107088, pibenzimol. Examples of preferred topo I inhibitors include but are not limited to camptothecin, topotecan (hycaptamine), irinotecan (irinotecan hydrochloride), belotecan, or an analogue or derivative thereof.


A topo II inhibitor that can be used in conjunction with the invention can be from any of the following classes of compounds: anthracycline antibiotics (e.g., carubicin, pirarubicin, daunorubicin citrate liposomal, daunomycin, 4-iodo-4-doxydoxorubicin, doxorubicin, n,n-dibenzyl daunomycin, morpholinodoxorubicin, aclacinomycin antibiotics, duborimycin, menogaril, nogalamycin, zorubicin, epirubicin, marcellomycin, detorubicin, annamycin, 7-cyanoquinocarcinol, deoxydoxorubicin, idarubicin, GPX-100, MEN-10755, valrubicin, KRN5500), epipodophyllotoxin compound (e.g., podophyllin, teniposide, etoposide, GL331, 2-ethylhydrazide), anthraquinone compound (e.g., ametantrone, bisantrene, mitoxantrone, anthraquinone), ciprofloxacin, acridine carboxamide, amonafide, anthrapyrazole antibiotics (e.g., teloxantrone, sedoxantrone trihydrochloride, piroxantrone, anthrapyrazole, losoxantrone), TAS-103, fostriecin, razoxane, XK469R, XK469, chloroquinoxaline sulfonamide, merbarone, intoplicine, elsamitrucin, CI-921, pyrazoloacridine, elliptinium, amsacrine. Examples of preferred topo II inhibitors include but are not limited to doxorubicin (Adriamycin), etoposide phosphate (etopofos), teniposide, sobuzoxane, or an analogue or derivative thereof.


DNA binding agents that can be used in conjunction with the invention include but are not limited to DNA groove binding agent, e.g., DNA minor groove binding agent; DNA crosslinking agent; intercalating agent; and DNA adduct forming agent. A DNA minor groove binding agent can be an anthracycline antibiotic, mitomycin antibiotic (e.g., porfiromycin, KW-2149, mitomycin B, mitomycin A, mitomycin C), chromomycin A3, carzelesin, actinomycin antibiotic (e.g., cactinomycin, dactinomycin, actinomycin F1), brostallicin, echinomycin, bizelesin, duocarmycin antibiotic (e.g., KW 2189), adozelesin, olivomycin antibiotic, plicamycin, zinostatin, distamycin, MS-247, ecteinascidin 743, amsacrine, anthramycin, and pibenzimol, or an analogue or derivative thereof.


DNA crosslinking agents include but are not limited to antineoplastic alkylating agent, methoxsalen, mitomycin antibiotic, psoralen. An antineoplastic alkylating agent can be a nitrosourea compound (e.g., cystemustine, tauromustine, semustine, PCNU, streptozocin, SarCNU, CGP-6809, carmustine, fotemustine, methylnitrosourea, nimustine, ranimustine, ethylnitrosourea, lomustine, chlorozotocin), mustard agent (e.g., nitrogen mustard compound, such as spiromustine, trofosfamide, chlorambucil, estramustine, 2,2,2-trichlorotriethylamine, prednimustine, novembichin, phenamet, glufosfamide, peptichemio, ifosfamide, defosfamide, nitrogen mustard, phenesterin, mannomustine, cyclophosphamide, melphalan, perfosfamide, mechlorethamine oxide hydrochloride, uracil mustard, bestrabucil, DHEA mustard, tallimustine, mafosfamide, aniline mustard, chlomaphazine; sulfur mustard compound, such as bischloroethylsulfide; mustard prodrug, such as TLK286 and ZD2767), ethylenimine compound (e.g., mitomycin antibiotic, ethylenimine, uredepa, thiotepa, diaziquone, hexamethylene bisacetamide, pentamethylmelamine, altretamine, carzinophilin, triaziquone, meturedepa, benzodepa, carboquone), alkylsulfonate compound (e.g., dimethylbusulfan, Yoshi-864, improsulfan, piposulfan, treosulfan, busulfan, hepsulfam), epoxide compound (e.g., anaxirone, mitolactol, dianhydrogalactitol, teroxirone), miscellaneous alkylating agent (e.g., ipomeanol, carzelesin, methylene dimethane sulfonate, mitobronitol, bizelesin, adozelesin, piperazinedione, VNP40101M, asaley, 6-hydroxymethylacylfulvene, EO9, etoglucid, ecteinascidin 743, pipobroman), platinum compound (e.g., ZD0473, liposomal-cisplatin analogue, satraplatin, BBR 3464, spiroplatin, ormaplatin, cisplatin, oxaliplatin, carboplatin, lobaplatin, zeniplatin, iproplatin), triazene compound (e.g., imidazole mustard, CB10-277, mitozolomide, temozolomide, procarbazine, dacarbazine), picoline compound (e.g., penclomedine), or an analogue or derivative thereof. Examples of preferred alkylating agents include but are not limited to cisplatin, dibromodulcitol, fotemustine, ifosfamide (ifosfamid), ranimustine (ranomustine), nedaplatin (latoplatin), bendamustine (bendamustine hydrochloride), eptaplatin, temozolomide (methazolastone), carboplatin, altretamine (hexamethylmelamine), prednimustine, oxaliplatin (oxalaplatinum), carmustine, thiotepa, leusulfon (busulfan), lobaplatin, cyclophosphamide, bisulfan, melphalan, and chlorambucil, or analogues or derivatives thereof.


Intercalating agents can be an anthraquinone compound, bleomycin antibiotic, rebeccamycin analogue, acridine, acridine carboxamide, amonafide, rebeccamycin, anthrapyrazole antibiotic, echinomycin, psoralen, LU 79553, BW A773U, crisnatol mesylate, benzo(a)pyrene-7,8-diol-9,10-epoxide, acodazole, elliptinium, pixantrone, or an analogue or derivative thereof.


DNA adduct forming agents include but are not limited to enediyne antitumor antibiotic (e.g., dynemicin A, esperamicin A1, zinostatin, dynemicin, calicheamicin gamma 1I), platinum compound, carmustine, tamoxifen (e.g., 4-hydroxy-tamoxifen), psoralen, pyrazine diazohydroxide, benzo(a)pyrene-7,8-diol-9,10-epoxide, or an analogue or derivative thereof.


Anti-metabolites include but are not limited to cytosine, arabinoside, floxuridine, fluorouracil, mercaptopurine, Gemcitabine, and methotrexate (MTX).


Ionizing radiation includes but is not limited to x-rays, gamma rays, and electron beams.


5.4.3. Methods of Determining Proteins or Other Molecules that Interact with a DNA Damage Response Gene

Any method suitable for detecting protein-protein interactions may be employed for identifying interaction of DNA damage response protein with another cellular protein. The interaction between DNA damage response gene and other cellular molecules, e.g., interaction between DNA damage response and its regulators, may also be determined using methods known in the art.


Among the traditional methods which may be employed are co-immunoprecipitation, crosslinking and co-purification through gradients or chromatographic columns. Utilizing procedures such as these allows for the identification of cellular proteins which interact with DNA damage response gene products. Once isolated, such a cellular protein can be identified and can, in turn, be used, in conjunction with standard techniques, to identify proteins it interacts with. For example, at least a portion of the amino acid sequence of the cellular protein which interacts with the DNA damage response gene product can be ascertained using techniques well known to those of skill in the art, such as via the Edman degradation technique (see, e.g., Creighton, 1983, “Proteins: Structures and Molecular Principles”, W.H. Freeman & Co., N.Y., pp. 34-49). The amino acid sequence obtained may be used as a guide for the generation of oligonucleotide mixtures that can be used to screen for gene sequences encoding such cellular proteins. Screening may be accomplished, for example, by standard hybridization or PCR techniques. Techniques for the generation of oligonucleotide mixtures and the screening are well-known. (See, e.g., Ausubel, supra., and PCR Protocols: A Guide to Methods and Applications, 1990, Innis, M. et al., eds. Academic Press, Inc., New York).


Additionally, methods may be employed which result in the simultaneous identification of genes which encode the cellular protein interacting with the DNA damage response protein. These methods include, for example, probing expression libraries with labeled DNA damage response protein, using DNA damage response protein in a manner similar to the well known technique of antibody probing of λgt11 libraries.


One method which detects protein interactions in vivo, the two-hybrid system, is described in detail for illustration only and not by way of limitation. One version of this system has been described (Chien et al., 1991, Proc. Natl. Acad. Sci. USA, 88:9578-9582) and is commercially available from Clontech (Palo Alto, Calif.).


Briefly, utilizing such a system, plasmids are constructed that encode two hybrid proteins: one consists of the DNA-binding domain of a transcription activator protein fused to the DNA damage response gene product and the other consists of the transcription activator protein's activation domain fused to an unknown protein that is encoded by a cDNA which has been recombined into this plasmid as part of a cDNA library. The DNA-binding domain fusion plasmid and the cDNA library are transformed into a strain of the yeast Saccharomyces cerevisiae that contains a reporter gene (e.g., HBS or lacZ) whose regulatory region contains the transcription activator's binding site. Either hybrid protein alone cannot activate transcription of the reporter gene: the DNA-binding domain hybrid cannot because it does not provide activation function and the activation domain hybrid cannot because it cannot localize to the activator's binding sites. Interaction of the two hybrid proteins reconstitutes the functional activator protein and results in expression of the reporter gene, which is detected by an assay for the reporter gene product.


The two-hybrid system or related methodology may be used to screen activation domain libraries for proteins that interact with the “bait” gene product. By way of example, and not by way of limitation, DNA damage response gene products may be used as the bait gene product. Total genomic or cDNA sequences are fused to the DNA encoding an activation domain. This library and a plasmid encoding a hybrid of a bait DNA damage response gene product fused to the DNA-binding domain are cotransformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene. For example, and not by way of limitation, a bait DNA damage response gene sequence, such as the coding sequence of a DNA damage response gene can be cloned into a vector such that it is translationally fused to the DNA encoding the DNA-binding domain of the GAL4 protein. These colonies are purified and the library plasmids responsible for reporter gene expression are isolated. DNA sequencing is then used to identify the proteins encoded by the library plasmids.


A cDNA library of the cell line from which proteins that interact with bait DNA damage response gene product are to be detected can be made using methods routinely practiced in the art. According to the particular system described herein, for example, the cDNA fragments can be inserted into a vector such that they are translationally fused to the transcriptional activation domain of GALA. This library can be co-transformed along with the bait DNA damage response gene-GAL4 fusion plasmid into a yeast strain which contains a lacZ gene driven by a promoter which contains GAL4 activation sequence. A cDNA encoded protein, fused to GAL4 transcriptional activation domain, that interacts with bait DNA damage response gene product will reconstitute an active GAL4 protein and thereby drive expression of the HIS3 gene. Colonies which express HIS3 can be detected by their growth on petri dishes containing semi-solid agar based media lacking histidine. The cDNA can then be purified from these strains, and used to produce and isolate the bait DNA damage response gene-interacting protein using techniques routinely practiced in the art.


The interaction between a DNA damage response gene and its regulators may be determined by a standard method known in the art.


5.4.4. Methods of Screening for Agents

The invention provides methods for screening for agents that regulate DNA damage response expression or modulate interaction of DNA damage response with other proteins or molecules.


5.4.4.1. Screening Assays

The following assays are designed to identify compounds that bind to DNA damage response gene or gene products, bind to other cellular proteins that interact with a DNA damage response gene product, bind to cellular constituents, e.g., proteins, that are affected by a DNA damage response gene product, or bind to compounds that interfere with the interaction of the DNA damage response gene or gene product with other cellular proteins and to compounds which modulate the activity of DNA damage response gene (i.e., modulate the level of DNA damage response gene expression and/or modulate the level of DNA damage response gene product activity). Assays may additionally be utilized which identify compounds which bind to DNA damage response gene regulatory sequences (e.g., promoter sequences), see e.g., Platt, K. A., 1994, J. Biol. Chem. 269:28558-28562, which is incorporated herein by reference in its entirety, which may modulate the level of DNA damage response gene expression. Compounds may include, but are not limited to, small organic molecules which are able to affect expression of the DNA damage response gene or some other gene involved in the DNA damage response pathways, or other cellular proteins. Methods for the identification of such cellular proteins are described, above, in Section 5.4.3. Such cellular proteins may be involved in the regulation of the growth inhibitory effect of a DNA damaging agent. Further, among these compounds are compounds which affect the level of DNA damage response gene expression and/or DNA damage response gene product activity and which can be used in the regulation of resistance to the growth inhibitory effect of a DNA damaging agent.


Compounds may include, but are not limited to, peptides such as, for example, soluble peptides, including but not limited to, Ig-tailed fusion peptides, and members of random peptide libraries; (see, e.g., Lam, K. S. et al., 1991, Nature 354:82-84; Houghten, R. et al., 1991, Nature 354:84-86), and combinatorial chemistry-derived molecular library made of D- and/or L-configuration amino acids, phosphopeptides (including, but not limited to members of random or partially degenerate, directed phosphopeptide libraries; see, e.g., Songyang, Z. et al., 1993, Cell 72:767-778), antibodies (including, but not limited to, polyclonal, monoclonal, humanized, anti-idiotypic, chimeric or single chain antibodies, and FAb, F(ab′)2 and FAb expression library fragments, and epitope-binding fragments thereof), and small organic or inorganic molecules.


Compounds identified via assays such as those described herein may be useful, for example, in regulating the biological function of the DNA damage response gene product, and for ameliorating resistance to the growth inhibitory effect of a DNA damaging agent and/or enhancing the growth inhibitory effect of a DNA damaging agent. Assays for testing the effectiveness of compounds are discussed, below, in Section 5.4.4.2.


In vitro systems may be designed to identify compounds capable of binding the DNA damage response gene products of the invention. Compounds identified may be useful, for example, in modulating the activity of wild type and/or mutant DNA damage response gene products, may be useful in elaborating the biological function of the DNA damage response gene product, may be utilized in screens for identifying compounds that disrupt normal DNA damage response gene product interactions, or may in themselves disrupt such interactions.


The principle of the assays used to identify compounds that bind to the DNA damage response gene product involves preparing a reaction mixture of the DNA damage response gene product and the test compound under conditions and for a time sufficient to allow the two components to interact and bind, thus forming a complex which can be removed and/or detected in the reaction mixture. These assays can be conducted in a variety of ways. For example, one method to conduct such an assay would involve anchoring DNA damage response gene product or the test substance onto a solid phase and detecting DNA damage response gene product/test compound complexes anchored on the solid phase at the end of the reaction. In one embodiment of such a method, the DNA damage response gene product may be anchored onto a solid surface, and the test compound, which is not anchored, may be labeled, either directly or indirectly.


In practice, microtiter plates may conveniently be utilized as the solid phase. The anchored component may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished by simply coating the solid surface with a solution of the protein and drying. Alternatively, an immobilized antibody, preferably a monoclonal antibody, specific for the protein to be immobilized may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.


In order to conduct the assay, the nonimmobilized component is added to the coated surface containing the anchored component. After the reaction is complete, unreacted components are removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the previously nonimmobilized component is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the previously nonimmobilized component is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the previously nonimmobilized component (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).


Alternatively, a reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for DNA damage response gene product or the test compound to anchor any complexes formed in solution, and a labeled antibody specific for the other component of the possible complex to detect anchored complexes.


The DNA damage response gene or gene products may interact in vivo with one or more intracellular or extracellular molecules, such as proteins. Such molecules may include, but are not limited to, nucleic acid molecules and those proteins identified via methods such as those described, above, in Section 5.4.3. For purposes of this discussion, such molecules are referred to herein as “binding partners”. Compounds that disrupt DNA damage response gene product binding may be useful in regulating the activity of the DNA damage response gene product. Compounds that disrupt DNA damage response gene binding may be useful in regulating the expression of the DNA damage response gene, such as by regulating the binding of a regulator of DNA damage response gene. Such compounds may include, but are not limited to molecules such as peptides, and the like, as described, for example, in Section 5.4.4.1. above, which would be capable of gaining access to the DNA damage response gene product.


The basic principle of the assay systems used to identify compounds that interfere with the interaction between the DNA damage response gene product and its intracellular or extracellular binding partner or partners involves preparing a reaction mixture containing the DNA damage response gene product, and the binding partner under conditions and for a time sufficient to allow the two to interact and bind, thus forming a complex. In order to test a compound for inhibitory activity, the reaction mixture is prepared in the presence and absence of the test compound. The test compound may be initially included in the reaction mixture, or may be added at a time subsequent to the addition of DNA damage response gene product and its binding partner. Control reaction mixtures are incubated without the test compound or with a placebo. The formation of any complexes between the DNA damage response gene protein and the binding partner is then detected. The formation of a complex in the control reaction, but not in the reaction mixture containing the test compound, indicates that the compound interferes with the interaction of the DNA damage response gene protein and the interactive binding partner. Additionally, complex formation within reaction mixtures containing the test compound and normal DNA damage response gene protein may also be compared to complex formation within reaction mixtures containing the test compound and a mutant DNA damage response gene protein. This comparison may be important in those cases wherein it is desirable to identify compounds that disrupt interactions of mutant but not normal DNA damage response gene proteins.


The assay for compounds that interfere with the interaction of the DNA damage response gene products and binding partners can be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring either the DNA damage response gene product or the binding partner onto a solid phase and detecting complexes anchored on the solid phase at the end of the reaction. In homogeneous assays, the entire reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested. For example, test compounds that interfere with the interaction between the DNA damage response gene products and the binding partners, e.g., by competition, can be identified by conducting the reaction in the presence of the test substance; i.e., by adding the test substance to the reaction mixture prior to or simultaneously with the DNA damage response gene protein and interactive binding partner. Alternatively, test compounds that disrupt preformed complexes, e.g. compounds with higher binding constants that displace one of the components from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed. The various formats are described briefly below.


In a heterogeneous assay system, either the DNA damage response gene product or the interactive binding partner, is anchored onto a solid surface, while the non-anchored species is labeled, either directly or indirectly. In practice, microtiter plates are conveniently utilized. The anchored species may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the DNA damage response gene product or binding partner and drying. Alternatively, an immobilized antibody specific for the species to be anchored may be used to anchor the species to the solid surface. The surfaces may be prepared in advance and stored.


In order to conduct the assay, the partner of the immobilized species is exposed to the coated surface with or without the test compound. After the reaction is complete, unreacted components are removed (e.g., by washing) and any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the non-immobilized species is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the non-immobilized species is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the initially non-immobilized species (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody). Depending upon the order of addition of reaction components, test compounds which inhibit complex formation or which disrupt preformed complexes can be detected.


Alternatively, the reaction can be conducted in a liquid phase in the presence or absence of the test compound, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for one of the binding components to anchor any complexes formed in solution, and a labeled antibody specific for the other partner to detect anchored complexes. Again, depending upon the order of addition of reactants to the liquid phase, test compounds which inhibit complex or which disrupt preformed complexes can be identified.


In an alternate embodiment of the invention, a homogeneous assay can be used. In this approach, a preformed complex of the DNA damage response gene protein and the interactive binding partner is prepared in which either the DNA damage response gene product or its binding partners is labeled, but the signal generated by the label is quenched due to complex formation (see, e.g., U.S. Pat. No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays). The addition of a test substance that competes with and displaces one of the species from the preformed complex will result in the generation of a signal above background. In this way, test substances which disrupt DNA damage response gene protein/binding partner interaction can be identified.


In a particular embodiment, the DNA damage response gene product can be prepared for immobilization using recombinant DNA techniques. For example, the DNA damage response coding region can be fused to a glutathione-5-transferase (GST) gene using a fusion vector, such as pGEX-5X-1, in such a manner that its binding activity is maintained in the resulting fusion protein. The interactive binding partner can be purified and used to raise a monoclonal antibody, using methods routinely practiced in the art. This antibody can be labeled with the radioactive isotope 125I, for example, by methods routinely practiced in the art. In a heterogeneous assay, e.g., the GST-DNA damage response fusion protein can be anchored to glutathione-agarose beads. The interactive binding partner can then be added in the presence or absence of the test compound in a manner that allows interaction and binding to occur. At the end of the reaction period, unbound material can be washed away, and the labeled monoclonal antibody can be added to the system and allowed to bind to the complexed components. The interaction between the DNA damage response gene protein and the interactive binding partner can be detected by measuring the amount of radioactivity that remains associated with the glutathione-agarose beads. A successful inhibition of the interaction by the test compound will result in a decrease in measured radioactivity.


Alternatively, the GST-DNA damage response gene fusion protein and the interactive binding partner can be mixed together in liquid in the absence of the solid glutathione-agarose beads. The test compound can be added either during or after the species are allowed to interact. This mixture can then be added to the glutathione-agarose beads and unbound material is washed away. Again the extent of inhibition of the DNA damage response gene product/binding partner interaction can be detected by adding the labeled antibody and measuring the radioactivity associated with the beads.


In another embodiment of the invention, these same techniques can be employed using peptide fragments that correspond to the binding domains of the DNA damage response protein and/or the interactive binding partner (in cases where the binding partner is a protein), in place of one or both of the full length proteins. Any number of methods routinely practiced in the art can be used to identify and isolate the binding sites. These methods include, but are not limited to, mutagenesis of the gene encoding one of the proteins and screening for disruption of binding in a co-immunoprecipitation assay. Compensating mutations in the gene encoding the second species in the complex can then be selected. Sequence analysis of the genes encoding the respective proteins will reveal the mutations that correspond to the region of the protein involved in interactive binding. Alternatively, one protein can be anchored to a solid surface using methods described in this Section above, and allowed to interact with and bind to its labeled binding partner, which has been treated with a proteolytic enzyme, such as trypsin. After washing, a short, labeled peptide comprising the binding domain may remain associated with the solid material, which can be isolated and identified by amino acid sequencing. Also, once the gene coding for the binding partner is obtained, short gene segments can be engineered to express peptide fragments of the protein, which can then be tested for binding activity and purified or synthesized.


For example, and not by way of limitation, a DNA damage response gene product can be anchored to a solid material as described, above, in this Section by making a GST-DNA damage response fusion protein and allowing it to bind to glutathione agarose beads. The interactive binding partner can be labeled with a radioactive isotope, such as 35S, and cleaved with a proteolytic enzyme such as trypsin. Cleavage products can then be added to the anchored GST-DNA damage response fusion protein and allowed to bind. After washing away unbound peptides, labeled bound material, representing the binding partner binding domain, can be eluted, purified, and analyzed for amino acid sequence by well-known methods. Peptides so identified can be produced synthetically or fused to appropriate facilitative proteins using recombinant DNA technology.


5.4.4.2. Screening Compounds that Regulate and/or Enhance the Growth Inhibitory Effect of a DNA Damaging Agent

Any agents that regulate the expression of DNA damage response gene and/or the interaction of DNA damage response protein with its binding partners, e.g., compounds that are identified in Section 5.4.4.1., antibodies to DNA damage response protein, and so on, can be further screened for its ability to regulate and/or enhance the growth inhibitory effect of a DNA damaging agent in cells. Any suitable proliferation or growth inhibition assays known in the art can be used for this purpose. In one embodiment, a candidate agent and a DNA damaging agent are applied to a cells of a cell line, and a change in growth inhibitory effect is determined. Preferably, changes in growth inhibitory effect are determined using different concentrations of the candidate agent in conjunction with different concentrations of the DNA damaging agent such that one or more combinations of concentrations of the candidate agent and DNA damaging agent which cause 50% inhibition, i.e., the IC50, are determined.


In a preferred embodiment, an MTT proliferation assay (see, e.g., van de Loosdrechet, et al., 1994, J. Immunol. Methods 174: 311-320; Ohno et al., 1991, J. Immunol. Methods 145:199-203; Ferrari et al., 1990, J. Immunol. Methods 131: 165-172; Alley et al., 1988, Cancer Res. 48: 589-601; Carmichael et al., 1987, Cancer Res. 47:936-942; Gerlier et al., 1986, J. Immunol. Methods 65:55-63; Mosmann, 1983, J. Immunological Methods 65:55-63) is used to screen for a candidate agent that can be used in conjunction with a DNA damaging agent to inhibit the growth of cells. The cells are treated with chosen concentrations of the candidate agent and a DNA damaging agent for 4 to 72 hours. The cells are then incubated with a suitable amount of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) for 1-8 hours such that viable cells convert MTT into an intracellular deposit of insoluble formazan. After removing the excess MTT contained in the supernatant, a suitable MTT solvent, e.g., a DMSO solution, is added to dissolved the formazan. The concentration of MTT, which is proportional to the number of viable cells, is then measured by determining the optical density at 570 nm. A plurality of different concentrations of the candidate agent can be assayed to allow the determination of the concentrations of the candidate agent and the DNA damaging agent which causes 50% inhibition.


In another preferred embodiment, an alamarBlue™ Assay for cell proliferation is used to screen for a candidate agent that can be used in conjunction with a DNA damaging agent to inhibit the growth of cells (see, e.g., Page et al., 1993, Int. J. Oncol. 3:473-476). An alamarBlue™ assay measures cellular respiration and uses it as a measure of the number of living cells. The internal environment of proliferating cells is more reduced than that of non-proliferating cells. For example, the ratios of NADPH/NADP, FADH/FAD, FMNH/FMN, and NADH/NAF increase during proliferation. AlamarBlue can be reduced by these metabolic intermediates and, therefore, can be used to monitor cell proliferation. The cell number of a treated sample as measured by alamarBlue can be expressed in percent relative to that of an untreated control sample.


In a specific embodiment, the alamarBlue™ assay is performed to determine whether transfection titration curves of siRNAs targeting DNA damage response genes were changed by the presence of a DNA damaging agent of a chosen concentration, e.g., 6-200 nM of camptothecin. Cells were transfected with an siRNA targeting a DNA damage response gene. 4 hours after siRNA transfection, 100 microliter/well of DMEM/10% fetal bovine serum with or without the DNA damaging agent was added and the plates were incubated at 37° C. and 5% CO2 for 68 hours. The medium was removed from the wells and replaced with 100 microliter/well DMEM/10% Fetal Bovine Serum (Invitrogen) containing 10% (vol/vol) alamarBlue™ reagent (Biosource International Inc., Camarillo, Calif.) and 0.001 volumes of 1M Hepes buffer tissue culture reagent (Invitrogen). The plates were incubated for 2 hours at 37° C. before they were read at 570 and 600 nm wavelengths on a SpectraMax plus plate reader (Molecular Devices, Sunnyvale, Calif.) using Softmax Pro 3.1.2 software (Molecular Devices). The percent reduced for wells transfected with a titration of an siRNA targeting a DNA damage response gene with or without a DNA damaging agent were compared to luciferase siRNA-transfected wells. The number calculated for % Reduced for 0 nM luciferase siRNA-transfected wells without the DNA damaging agent was considered to be 100%.


5.4.4.3. Compounds Identified

The compounds identified in the screen include compounds that demonstrate the ability to selectively modulate the expression of DNA damage response and regulate and/or enhance the growth inhibitory effect of a DNA damaging agent in cells. These compounds include but are not limited to siRNA, antisense, ribozyme, triple helix, antibody, and polypeptide molecules, aptamrs, and small organic or inorganic molecules.


The compounds identified in the screen also include compounds that modulate interaction of DNA damage response with other proteins or molecules. In one embodiment, the compounds identified in the screen are compounds that modulate the interaction of a DNA damage response protein with its interaction partner. In another embodiment, the compounds identified in the screen are compounds that modulate the interaction of DNA damage response gene with a transcription regulator.


5.4.5. Diagnostics

A variety of methods can be employed for the diagnostic and prognostic evaluation of cell or cells for their resistance to the growth inhibitory effect of a DNA damaging agent, e.g., camptothecin, cisplatin or doxorubicin, resulting from defective regulation of DNA damage response, and for the identification of subjects having a predisposition to resistance to the growth inhibitory effect of a DNA damaging agent.


In one embodiment, the method comprises determining an expression level of a DNA damage response gene in the cell, in which an expression level above a predetermined threshold level indicates that the cell is DNA damaging agent resistant. Preferably, the predetermined threshold level is at least 2-fold, 4-fold, 8-fold, or 10-fold of the normal expression level of the DNA damage response gene. In another embodiment, the invention provides a method for evaluating DNA damaging agent resistance in a cell comprising determining a level of abundance of a protein encoded by a DNA damage response gene in the cell, in which a level of abundance of the protein above a predetermined threshold level indicates that the cell is DNA damaging agent resistant. In still another embodiment, the invention provides a method for evaluating DNA damaging agent resistance in a cell comprising determining a level of activity of a protein encoded by the DNA damage response gene in cells of the mammal, in which an activity level above a predetermined threshold level indicates that the cell is DNA damaging agent resistant. Preferably, the predetermined threshold level of abundance or activity is at least 2-fold, 4-fold, 8-fold, or 10-fold of the normal level of abundance or activity of the DNA damage response protein.


Such methods may, for example, utilize reagents such as the DNA damage response gene nucleotide sequences and antibodies directed against DNA damage response gene products, including peptide fragments thereof. Specifically, such reagents may be used, for example, for: (1) the detection of the presence of DNA damage response gene mutations, or the detection of either over- or under-expression of DNA damage response gene mRNA relative to the normal expression level; and (2) the detection of either an over- or an under-abundance of DNA damage response gene product relative to the normal DNA damage response protein level.


The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one specific DNA damage response gene nucleic acid or anti-DNA damage response antibody reagent described herein, which may be conveniently used, e.g., in clinical settings, to diagnose patients exhibiting DNA damage response related disorder or abnormalities.


For the detection of DNA damage response mutations, any nucleated cell can be used as a starting source for genomic nucleic acid. For the detection of DNA damage response gene expression or DNA damage response gene products, any cell type or tissue in which the DNA damage response gene is expressed may be utilized.


Nucleic acid-based detection techniques are described, below, in Section 5.4.5.1. Peptide detection techniques are described, below, in Section 5.4.5.2.


5.4.5.1. Detection of Expression of a DNA Damage Response Gene

The expression of DNA damage response gene in cells or tissues, e.g., the cellular level of DNA damage response transcripts and/or the presence or absence of mutations, can be detected by utilizing a number of techniques. Nucleic acid from any nucleated cell can be used as the starting point for such assay techniques, and may be isolated according to standard nucleic acid preparation procedures which are well known to those of skill in the art. For example, the expression level of the DNA damage response gene can determined by measuring the expression level of the DNA damage response gene using one or more polynucleotide probes, each of which comprises a nucleotide sequence in the DNA damage response gene. In particularly preferred embodiments of the invention, the method is used to diagnose resistance of a cancer to a treatment using DNA damaging agent in a human.


DNA may be used in hybridization or amplification assays of biological samples to detect abnormalities involving DNA damage response gene structure, including point mutations, insertions, deletions and chromosomal rearrangements. Such assays may include, but are not limited to, Southern analyses, single stranded conformational polymorphism analyses (SSCP), DNA microarray analyses, and PCR analyses.


Such diagnostic methods for the detection of DNA damage response gene-specific mutations can involve, for example, contacting and incubating nucleic acids including recombinant DNA molecules, cloned genes or degenerate variants thereof, obtained from a sample, e.g., derived from a patient sample or other appropriate cellular source, with one or more labeled nucleic acid reagents including recombinant DNA molecules, cloned genes or degenerate variants thereof, under conditions favorable for the specific annealing of these reagents to their complementary sequences within the DNA damage response gene. Preferably, the lengths of these nucleic acid reagents are at least 15 to 30 nucleotides. After incubation, all non-annealed nucleic acids are removed from the nucleic acid:DNA damage response molecule hybrid. The presence of nucleic acids which have hybridized, if any such molecules exist, is then detected. Using such a detection scheme, the nucleic acid from the cell type or tissue of interest can be immobilized, for example, to a solid support such as a membrane, or a plastic surface such as that on a microtiter plate or polystyrene beads. In this case, after incubation, non-annealed, labeled nucleic acid reagents are easily removed. Detection of the remaining, annealed, labeled DNA damage response nucleic acid reagents is accomplished using standard techniques well-known to those in the art. The DNA damage response gene sequences to which the nucleic acid reagents have annealed can be compared to the annealing pattern expected from a normal DNA damage response gene sequence in order to determine whether a DNA damage response gene mutation is present.


Alternative diagnostic methods for the detection of DNA damage response gene specific nucleic acid molecules, in patient samples or other appropriate cell sources, may involve their amplification, e.g., by PCR (the experimental embodiment set forth in Mullis, K. B., 1987, U.S. Pat. No. 4,683,202), followed by the detection of the amplified molecules using techniques well known to those of skill in the art. The resulting amplified sequences can be compared to those which would be expected if the nucleic acid being amplified contained only normal copies of the DNA damage response gene in order to determine whether a DNA damage response gene mutation exists.


Among the DNA damage response nucleic acid sequences which are preferred for such hybridization and/or PCR analyses are those which will detect the presence of the DNA damage response gene splice site mutation.


Additionally, well-known genotyping techniques can be performed to identify individuals carrying DNA damage response gene mutations. Such techniques include, for example, the use of restriction fragment length polymorphisms (RFLPs), which involve sequence variations in one of the recognition sites for the specific restriction enzyme used.


Additionally, improved methods for analyzing DNA polymorphisms which can be utilized for the identification of DNA damage response gene mutations have been described which capitalize on the presence of variable numbers of short, tandemly repeated DNA sequences between the restriction enzyme sites. For example, Weber (U.S. Pat. No. 5,075,217, which is incorporated herein by reference in its entirety) describes a DNA marker based on length polymorphisms in blocks of (dC-dA)n-(dG-dT)n short tandem repeats. The average separation of (dC-dA)n-(dG-dT)n blocks is estimated to be 30,000-60,000 bp. Markers which are so closely spaced exhibit a high frequency co-inheritance, and are extremely useful in the identification of genetic mutations, such as, for example, mutations within the DNA damage response gene, and the diagnosis of diseases and disorders related to DNA damage response mutations.


Also, Caskey et al. (U.S. Pat. No. 5,364,759, which is incorporated herein by reference in its entirety) describe a DNA profiling assay for detecting short tri and tetra nucleotide repeat sequences. The process includes extracting the DNA of interest, such as the DNA damage response gene, amplifying the extracted DNA, and labelling the repeat sequences to form a genotypic map of the individual's DNA.


The level of DNA damage response gene expression can also be assayed. For example, RNA from a cell type or tissue known, or suspected, to express the DNA damage response gene, such as a cancer cell type which exhibits DNA damaging agent resistance, may be isolated and tested utilizing hybridization or PCR techniques such as are described, above. The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be a necessary step in the assessment of cells to be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the DNA damage response gene. Such analyses may reveal both quantitative and qualitative aspects of the expression pattern of the DNA damage response gene, including activation or inactivation of DNA damage response gene expression.


In one embodiment of such a detection scheme, a cDNA molecule is synthesized from an RNA molecule of interest (e.g., by reverse transcription of the RNA molecule into cDNA). A sequence within the cDNA is then used as the template for a nucleic acid amplification reaction, such as a PCR amplification reaction, or the like. The nucleic acid reagents used as synthesis initiation reagents (e.g., primers) in the reverse transcription and nucleic acid amplification steps of this method are chosen from among the DNA damage response gene nucleic acid reagents. The preferred lengths of such nucleic acid reagents are at least 9-30 nucleotides. For detection of the amplified product, the nucleic acid amplification may be performed using radioactively or non-radioactively labeled nucleotides. Alternatively, enough amplified product may be made such that the product may be visualized by utilizing any suitable nucleic acid staining method, e.g., by standard ethidium bromide staining.


Additionally, it is possible to perform such DNA damage response gene expression assays “in situ”, i.e., directly upon tissue sections (fixed and/or frozen) of patient tissue obtained from biopsies or resections, such that no nucleic acid purification is necessary. Nucleic acids from a DNA damage response gene may be used as probes and/or primers for such in situ procedures (see, for example, Nuovo, G. J., 1992, “PCR In Situ Hybridization:


Protocols And Applications”, Raven Press, NY).


Alternatively, if a sufficient quantity of the appropriate cells can be obtained, standard Northern analysis can be performed to determine the level of mRNA expression of the DNA damage response gene.


The expression of DNA damage response gene in cells or tissues, e.g., the cellular level of DNA damage response transcripts and/or the presence or absence of mutations, can also be evaluated using DNA microarray technologies. In such technologies, one or more polynucleotide probes each comprising a sequence of the DNA damage response gene are used to monitor the expression of the DNA damage response gene. The present invention therefore provides DNA microarrays comprising polynucleotide probes comprising sequences of the DNA damage response gene.


Any formats of DNA microarray technologies can be used in conjunction with the present invention. In one embodiment, spotted cDNA arrays are prepared by depositing PCR products of cDNA fragments, e.g., full length cDNAs, ESTs, etc., of the DNA damage response gene onto a suitable surface (see, e.g., DeRisi et al., 1996, Nature Genetics 14:457-460; Shalon et al., 1996, Genome Res. 6:689-645; Schena et al., 1995, Proc. Natl. Acad. Sci. U.S.A. 93:10539-11286; and Duggan et al., Nature Genetics Supplement 21:10-14). In another embodiment, high-density oligonucleotide arrays containing oligonucleotides complementary to sequences of DNA damage response gene are synthesized in situ on the surface by photolithographic techniques (see, e.g., Fodor et al., 1991, Science 251:767-773; Pease et al., 1994, Proc. Natl. Acad. Sci. U.S.A. 91:5022-5026; Lockhart et al., 1996, Nature Biotechnology 14:1675; McGall et al., 1996, Proc. Natl. Acad. Sci. U.S.A. 93:13555-13560; U.S. Pat. Nos. 5,578,832; 5,556,752; 5,510,270; 5,858,659; and 6,040,138). This format of microarray technology is particular useful for detection of single nucleotide polymorphisms (SNPs) (see, e.g., Hacia et al., 1999, Nat Genet. 22:164-7; Wang et al., 1998, Science 280:1077-82). In yet another embodiment, high-density oligonucleotide arrays containing oligonucleotides complementary to sequences of DNA damage response gene are synthesized in situ on the surface by inkjet technologies (see, e.g., Blanchard, International Patent Publication WO 98/41531, published Sep. 24, 1998; Blanchard et al., 1996, Biosensors and Bioelectronics 11:687-690; Blanchard, 1998, in Synthetic DNA Arrays in Genetic Engineering, Vol. 20, J. K. Setlow, Ed., Plenum Press, New York at pages 111-123).


In still another embodiment, DNA microarrays that allow electronic stringency control can be used in conjunction with polynucleotide probes comprising sequences of the DNA damage response gene (see, e.g., U.S. Pat. No. 5,849,486).


5.4.5.2. Detection of DNA Damage Response Gene Products

Antibodies directed against wild type or mutant DNA damage response gene products or conserved variants or peptide fragments thereof may be used as diagnostics and prognostics of DNA damaging agent resistance, as described herein. Such diagnostic methods may be used to detect abnormalities in the level of DNA damage response gene expression, or abnormalities in the structure and/or temporal, tissue, cellular, or subcellular location of DNA damage response gene product.


Because evidence disclosed herein indicates that the DNA damage response gene product is an intracellular gene product, the antibodies and immunoassay methods described below have important in vitro applications in assessing the efficacy of treatments for disorders resulting from defective regulation of DNA damage response gene such as proliferative diseases. Antibodies, or fragments of antibodies, such as those described below, may be used to screen potentially therapeutic compounds in vitro to determine their effects on DNA damage response gene expression and DNA damage response peptide production. The compounds which have beneficial effects on disorders related to defective regulation of DNA damage response can be identified, and a therapeutically effective dose determined.


In vitro immunoassays may also be used, for example, to assess the efficacy of cell-based gene therapy for disorders related to defective regulation of DNA damage response. Antibodies directed against DNA damage response peptides may be used in vitro to determine the level of DNA damage response gene expression achieved in cells genetically engineered to produce DNA damage response peptides. Given that evidence disclosed herein indicates that the DNA damage response gene product is an intracellular gene product, such an assessment is, preferably, done using cell lysates or extracts. Such analysis will allow for a determination of the number of transformed cells necessary to achieve therapeutic efficacy in vivo, as well as optimization of the gene replacement protocol.


The tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the DNA damage response gene, such as, a DNA damaging agent resistant cancer cell type. The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York), which is incorporated herein by reference in its entirety. The isolated cells can be derived from cell culture or from a patient. The analysis of cell taken from culture may be used to test the effect of compounds on the expression of the DNA damage response gene.


Preferred diagnostic methods for the detection of DNA damage response gene products or conserved variants or peptide fragments thereof, may involve, for example, immunoassays wherein the DNA damage response gene products or conserved variants or peptide fragments are detected by their interaction with an anti-DNA damage response gene product-specific antibody.


For example, antibodies, or fragments of antibodies, that bind DNA damage response protein, may be used to quantitatively or qualitatively detect the presence of DNA damage response gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody (see below, this Section) coupled with light microscopic, flow cytometric, or fluorimetric detection. Such techniques are especially preferred if such DNA damage response gene products are expressed on the cell surface.


The antibodies (or fragments thereof) useful in the present invention may, additionally, be employed histologically, as in immunofluorescence or immunoelectron microscopy, for in situ detection of DNA damage response gene products or conserved variants or peptide fragments thereof. In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody of the present invention. The antibody (or fragment) is preferably applied by overlaying the labeled antibody (or fragment) onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the DNA damage response gene product, or conserved variants or peptide fragments, but also its distribution in the examined tissue. Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.


Immunoassays for DNA damage response gene products or conserved variants or peptide fragments thereof will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of identifying DNA damage response gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.


The biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled DNA damage response protein specific antibody. The solid phase support may then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on solid support may then be detected by conventional means.


By “solid phase support or carrier” is intended any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention. The support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to an antigen or antibody. Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tub, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc. Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.


The binding activity of a given lot of anti-DNA damage response gene product antibody may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.


One of the ways in which the DNA damage response gene peptide-specific antibody can be detectably labeled is by linking the same to an enzyme and use in an enzyme immunoassay (EIA) (Voller, A., “The Enzyme Linked Immunosorbent Assay (ELISA)”, 1978, Diagnostic Horizons 2:1-7, Microbiological Associates Quarterly Publication, Walkersville, Md.); Voller, A. et al., 1978, J. Clin. Pathol. 31:507-520; Butler, J. E., 1981, Meth. Enzymol. 73:482-523; Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC Press, Boca Raton, Fla.; Ishikawa, E. et al., (eds.), 1981, Enzyme Immunoassay, Kgaku Shoin, Tokyo). The enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. The detection can be accomplished by calorimetric methods which employ a chromogenic substrate for the enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.


Detection may also be accomplished using any of a variety of other immunoassays. For example, by radioactively labeling the antibodies or antibody fragments, it is possible to detect DNA damage response gene peptides through the use of a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March 1986, which is incorporated by reference herein). The radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.


It is also possible to label the antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.


The antibody can also be detectably labeled using fluorescence emitting metals such as 152Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).


The antibody can also be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.


Likewise, a bioluminescent compound may be used to label the antibody of the present invention. Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.


5.4.6. Methods of Regulating Expression of DNA Damage Response Gene

A variety of therapeutic approaches may be used in accordance with the invention to modulate expression of the DNA damage response gene in vivo. For example, siRNA molecules may be engineered and used to silence DNA damage response gene in vivo. Antisense DNA molecules may also be engineered and used to block translation of DNA damage response mRNA in vivo. Alternatively, ribozyme molecules may be designed to cleave and destroy the DNA damage response mRNAs in vivo. In another alternative, oligonucleotides designed to hybridize to the 5′ region of the DNA damage response gene (including the region upstream of the coding sequence) and form triple helix structures may be used to block or reduce transcription of the DNA damage response gene. Oligonucleotides can also be designed to hybridize and form triple helix structures with the binding site of a negative regulator so as to block the binding of the negative regulator and to enhance the transcription of the DNA damage response gene.


In a preferred embodiment, siRNA, antisense, ribozyme, and triple helix nucleotides are designed to inhibit the translation or transcription of one or more of DNA damage response isoforms with minimal effects on the expression of other genes that may share one or more sequence motif with a DNA damage response. To accomplish this, the oligonucleotides used should be designed on the basis of relevant sequences unique to DNA damage response.


For example, and not by way of limitation, the oligonucleotides should not fall within those region where the nucleotide sequence of DNA damage response is most homologous to that of other genes. In the case of antisense molecules, it is preferred that the sequence be chosen from the list above. It is also preferred that the sequence be at least 18 nucleotides in length in order to achieve sufficiently strong annealing to the target mRNA sequence to prevent translation of the sequence. Izant et al., 1984, Cell, 36:1007-1015; Rosenberg et al., 1985, Nature, 313:703-706.


In the case of the “hammerhead” type of ribozymes, it is also preferred that the target sequences of the ribozymes be chosen from the list above. Ribozymes are RNA molecules which possess highly specific endoribonuclease activity. Hammerhead ribozymes comprise a hybridizing region which is complementary in nucleotide sequence to at least part of the target RNA, and a catalytic region which is adapted to cleave the target RNA. The hybridizing region contains nine (9) or more nucleotides. Therefore, the hammerhead ribozymes of the present invention have a hybridizing region which is complementary to the sequences listed above and is at least nine nucleotides in length. The construction and production of such ribozymes is well known in the art and is described more fully in Haseloff et al., 1988, Nature, 334:585-591.


The ribozymes of the present invention also include RNA endoribonucleases (hereinafter “Cech-type ribozymes”) such as the one which occurs naturally in Tetrahymena Thermophila (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224:574-578; Zaug and Cech, 1986, Science, 231:470-475; Zaug, et al., 1986, Nature, 324:429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been et al., 1986, Cell, 47:207-216). The Cech endoribonucleases have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.


In the case of oligonucleotides that hybridize to and form triple helix structures at the 5′ terminus of the DNA damage response gene and can be used to block transcription, it is preferred that they be complementary to those sequences in the 5′ terminus of DNA damage response which are not present in other DNA damage response related genes. It is also preferred that the sequences not include those regions of the DNA damage response promoter which are even slightly homologous to that of other DNA damage response related genes. The foregoing compounds can be administered by a variety of methods which are known in the art including, but not limited to the use of liposomes as a delivery vehicle. Naked DNA or RNA molecules may also be used where they are in a form which is resistant to degradation such as by modification of the ends, by the formation of circular molecules, or by the use of alternate bonds including phosphothionate and thiophosphoryl modified bonds. In addition, the delivery of nucleic acid may be by facilitated transport where the nucleic acid molecules are conjugated to poly-lysine or transferrin. Nucleic acid may also be transported into cells by any of the various viral carriers, including but not limited to, retrovirus, vaccinia, AAV, and adenovirus.


Alternatively, a recombinant nucleic acid molecule which encodes, or is, such antisense, ribozyme, triple helix, or DNA damage response molecule can be constructed. This nucleic acid molecule may be either RNA or DNA. If the nucleic acid encodes an RNA, it is preferred that the sequence be operatively attached to a regulatory element so that sufficient copies of the desired RNA product are produced. The regulatory element may permit either constitutive or regulated transcription of the sequence. In vivo, that is, within the cells or cells of an organism, a transfer vector such as a bacterial plasmid or viral RNA or DNA, encoding one or more of the RNAs, may be transfected into cells e.g. (Llewellyn et al., 1987, J. Mol. Biol., 195:115-123; Hanahan et al. 1983, J. Mol. Biol., 166:557-580). Once inside the cell, the transfer vector may replicate, and be transcribed by cellular polymerases to produce the RNA or it may be integrated into the genome of the host cell. Alternatively, a transfer vector containing sequences encoding one or more of the RNAs may be transfected into cells or introduced into cells by way of micromanipulation techniques such as microinjection, such that the transfer vector or a part thereof becomes integrated into the genome of the host cell.


RNAi can also be used to knock down the expression of DNA damage response. In one embodiment, double-stranded RNA molecules of 21-23 nucleotides which hybridize to a homologous region of mRNAs transcribed from the DNA damage response gene are used to degrade the mRNAs, thereby “silence” the expression of the DNA damage response gene. Preferably, the dsRNAs have a hybridizing region, e.g., a 19-nucleotide double-stranded region, which is complementary to a sequence of the coding sequence of the DNA damage response gene. Any siRNA targeting an appropriate coding sequence of a DNA damage response gene, e.g., a human DNA damage response gene, can be used in the invention. As an exemplary embodiment, 21-nucleotide double-stranded siRNAs targeting the coding regions of DNA damage response gene are designed according to standard selection rules (see, e.g., Elbashir et al., 2002, Methods 26:199-213, which is incorporated herein by reference in its entirety).


Any standard method for introducing nucleic acids into cells can be used. In one embodiment, gene silencing is induced by presenting the cell with the siRNA targeting the DNA damage response gene (see, e.g., Elbashir et al., 2001, Nature 411, 494-498; Elbashir et al., 2001, Genes Dev. 15, 188-200, all of which are incorporated by reference herein in their entirety). The siRNAs can be chemically synthesized, or derived from cleavage of double-stranded RNA by recombinant Dicer. Another method to introduce a double stranded DNA (dsRNA) for silencing of the DNA damage response gene is shRNA, for short hairpin RNA (see, e.g., Paddison et al., 2002, Genes Dev. 16, 948-958; Brummelkamp et al., 2002, Science 296, 550-553; Sui, G. et al. 2002, Proc. Natl. Acad. Sci. USA 99, 5515-5520, all of which are incorporated by reference herein in their entirety). In this method, an siRNA targeting DNA damage response gene is expressed from a plasmid (or virus) as an inverted repeat with an intervening loop sequence to form a hairpin structure. The resulting RNA transcript containing the hairpin is subsequently processed by Dicer to produce siRNAs for silencing. Plasmid-based shRNAs can be expressed stably in cells, allowing long-term gene silencing in cells both in vitro and in vivo (see, McCaffrey et al. 2002, Nature 418, 38-39; Xia et al., 2002, Nat. Biotech. 20, 1006-1010; Lewis et al., 2002, Nat. Genetics 32, 107-108; Rubinson et al., 2003, Nat. Genetics 33, 401-406; Tiscomia et al., 2003, Proc. Natl. Acad. Sci. USA 100, 1844-1848, all of which are incorporated by reference herein in their entirety). SiRNAs targeting the DNA damage response gene can also be delivered to an organ or tissue in a mammal, such a human, in vivo (see, e.g., Song et al. 2003, Nat. Medicine 9, 347-351; Sorensen et al., 2003, J. Mol. Biol. 327, 761-766; Lewis et al., 2002, Nat. Genetics 32, 107-108, all of which are incorporated by reference herein in their entirety). In this method, a solution of siRNA is injected intravenously into the mammal. The siRNA can then reach an organ or tissue of interest and effectively reduce the expression of the target gene in the organ or tissue of the mammal.


5.4.7. Methods of Regulating Activity of a DNA Damage Response Protein and/or Its Pathway

The activity of DNA damage response protein can be regulated by modulating the interaction of DNA damage response protein with its binding partners. In one embodiment, agents, e.g., antibodies, aptamers, small organic or inorganic molecules, can be used to inhibit binding of a DNA damage response binding partner such that DNA damaging agent resistance is regulated. In another embodiment, agents, e.g., antibodies, aptamers, small organic or inorganic molecules, can be used to inhibit the activity of a protein in a DNA damage response protein regulatory pathway such that DNA damaging agent resistance is regulated. In one embodiment, a kinase inhibitor, e.g., Herbimycin, Gleevec, Genistein, Lavendustin, Iressa, is used to regulate the activety of DNA damage response protein kinases.


5.4.8. Cancer Therapy by Targeting a DNA Damage Response Gene and/or Its Product

The methods and/or compositions described above for modulating DNA damage response expression and/or activity may be used to treat patients who have a cancer in conjunction with a DNA damaging agent. In particular, the methods and/or compositions may be used in conjunction with a DNA damaging agent for treatment of a patient having a cancer which exhibits DNA damage response mediated DNA damaging agent resistance. Such therapies may be used to treat cancers, including but not limted to, rhabdomyosarcoma, neuroblastoma and glioblastoma, small cell lung cancer, osteoscarcoma, pancreatic cancer, breast and prostate cancer, murine melanoma and leukemia, and B-cell lymphoma.


In preferred embodiments, the methods and/or compositions of the invention are used in conjunction with a DNA damaging agent for treatment of a patient having a cancer which exhibits DNA damage response mediated DNA damaging agent resistance. In such embodiments, the expression and/or activity of DNA damage response are modulated to confer cancer cells sensitivity to a DNA damaging agent, thereby conferring or enhancing the efficacy of DNA damaging agent therapy.


In a combination therapy, one or more compositions of the present invention can be administered before, at the same time of, or after the administration of a DNA damaging agent. In one embodiment, the compositions of the invention are administered before the administration a DNA damaging agent. The time intervals between the administration of the compositions of the invention and a DNA damaging agent can be determined by routine experiments that are familiar to one skilled person in the art. In one embodiment, a DNA damaging agent is given after the DNA damage response protein level reaches a desirable threshold. The level of DNA damage response protein can be determined by using any techniques described supra.


In another embodiment, the compositions of the invention are administered at the same time with the DNA damaging agent.


In still another embodiment, one or more of the compositions of the invention are also administered after the administration of a DNA damaging agent. Such administration can be beneficial especially when the DNA damaging agent has a longer half life than that of the one or more of the compositions of the invention used in the treatment.


It will be apparent to one skilled person in the art that any combination of different timing of the administration of the compositions of the invention and a DNA damaging agent can be used. For example, when the DNA damaging agent has a longer half life than that of the composition of the invention, it is preferable to administer the compositions of the invention before and after the administration of the DNA damaging agent.


The frequency or intervals of administration of the compositions of the invention depends on the desired DNA damage response level, which can be determined by any of the techniques described supra. The administration frequency of the compositions of the invention can be increased or decreased when the DNA damage response protein level changes either higher or lower from the desired level.


The effects or benefits of administration of the compositions of the invention alone or in conjunction with a DNA damaging agent can be evaluated by any methods known in the art, e.g., by methods that are based on measuring the survival rate, side effects, dosage requirement of the DNA damaging agent, or any combinations thereof. If the administration of the compositions of the invention achieves any one or more of the benefits in a patient, such as increasing the survival rate, decreasing side effects, lowing the dosage requirement for the DNA damaging agent, the compositions of the invention are said to have augmented the DNA damaging agent therapy, and the method is said to have efficacy.


5.5. Pharmaceutical Formulations and Routes of Administration

The compounds that are determined to affect STK6 gene expression or gene product activity can be administered to a patient at therapeutically effective doses to treat or ameliorate disorders related to defective regulation of STK6. A therapeutically effective dose refers to that amount of the compound sufficient to result in amelioration of KSPi resistance and/or enhancement of the growth inhibitory effect of a KSP inhibitor in cells.


5.5.1. Effective Dose

Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.


The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.


5.5.2. Formulations and Use

Pharmaceutical compositions for use in accordance with the present invention may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients.


Thus, the compounds and their physiologically acceptable salts and solvates may be formulated for administration by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.


For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.


Preparations for oral administration may be suitably formulated to give controlled release of the active compound.


For buccal administration the compositions may take the form of tablets or lozenges formulated in conventional manner.


For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.


The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.


The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.


In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.


The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.


5.5.3. Routes of Administration

Suitable routes of administration may, for example, include oral, rectal, transmucosal, transdermal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.


Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into an affected area, often in a depot or sustained release formulation.


Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with an antibody specific for affected cells. The liposomes will be targeted to and taken up selectively by the cells.


5.5.4. Packaging

The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions indicated on the label may include treatment of a disease such as one characterized by aberrant or excessive STK6 or a DNA damage response gene expression or activity.


6. Examples

The following examples are presented by way of illustration of the present invention, and are not intended to limit the present invention in any way.


6.1. Example 1
STK6 and TPX2 Interacts with KSP

This Example illustrates screening of an siRNA library for genes that interact with inhibitors of KSP gene. CIN8 is the S. cerevisiae homolog of KSP. Deletion mutants of CIN8 are viable and many genes have been identified that are essential in the absence (but not the presence) of CIN8 (Geiser et al., 1997, Mol Biol Cell. 8:1035-1050). By analogy, it was reasoned that disruption of human homologues of these genes might be more disruptive to tumor cell growth in the presence than in the absence of suboptimal concentrations of a KSPi. An siRNA library containing siRNAs to homologues of 11 genes reported to be synthetic lethal with CIN8: CDC20, ROCK2, TTK, FZR1, BUB1, BUB3, BUB1B, MAD1L1, MAD2L1, DNCH1 and STK6 was screened for genes that modulates the effect of a KSP inhibitor, (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine, (EC50˜80 nM). The sequences of siRNAs targeting the 11 genes are listed in Table I. These siRNAs were transfected into HeLa cells in the presence or absence of an <EC10 concentration (25 nM) of (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine. Table I also lists the sequences of siRNAs that target respectively luciferase, PTEN, and KSP.


siRNA transfection was carried out as follows: one day prior to transfection, 100 microliters of a chosen cells, e.g., cervical cancer HeLa cells (ATCC, Cat. No. CCL-2), grown in DMEM/10% fetal bovine serum (Invitrogen, Carlsbad, Calif.) to approximately 90% confluency were seeded in a 96-well tissue culture plate (Corning, Corning, N.Y.) at 1500 cells/well. For each transfection 85 microliters of OptiMEM (Invitrogen) was mixed with 5 microliter of serially diluted siRNA (Dharmacon, Denver) from a 20 micro molar stock. For each transfection 5 microliter of OptiMEM was mixed with 5 microliter of Oligofectamine reagent (Invitrogen) and incubated 5 minutes at room temperature. The 10-microliter OptiMEM/Oligofectamine mixture was dispensed into each tube with the OptiMEM/siRNA mixture, mixed and incubated 15-20 minutes at room temperature. 10 microliter of the transfection mixture was aliquoted into each well of the 96-well plate and incubated for 4 hours at 37° C. and 5% CO2.


After 4 hours, 100 microliter/well of DMEM/10% fetal bovine serum with or without 25 nM (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine was added and the plates were incubated at 37° C. and 5% CO2 for 68 hours. The alamarBlue Assay was used for measurement of cell growth (see, Section 5.2). The alamarBlue assay measures cellular respiration and uses the meausrement as a measure of the number of living cells. The internal environment of the proliferating cell is more reduced than that of non-proliferating cells. Specifically, the ratios of NADPH/NADP, FADH/FAD, FMNH/FMN, and NADH/NAF increase during proliferation. AlamarBlue can be reduced by these metabolic intermediates and, therefore, can be used to monitor cell proliferation. In this Example, the alamarBlue assay was performed to determine whether STK6 siRNA transfection titration curves were changed by the presence of 25 nM of the KSP inhibitor (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine as follows: 72 hours after transfection the medium was removed from the wells and replaced with 100 microliter/well DMEM/10% Fetal Bovine Serum (Invitrogen) containing 10% (vouvol) alamarBlue reagent (Biosource International Inc., Camarillo, Calif.) and 0.001 volumes of 1M Hepes buffer tissue culture reagent (Invitrogen). The plates were incubated 2 hours at 37° C. and the plate was read at 570 and 600 nm wavelengths on a SpectraMax plus plate reader (Molecular Devices, Sunnyvale, Calif.) using Softmax Pro 3.1.2 software (Molecular Devices). The % Reduced of wells containing samples was determined according to Eq. 1. The % Reduced of the wells containing no cell was subtracted from the % Reduced of the wells containing samples to determine the % Reduced above the background level. The % Reduced for wells transfected with a titration of STK6 siRNA with or without 25 nM (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine were compared to that of wells transfected with an siRNA targeting luciferase. The number calculated for % Reduced for 0 nM luciferase siRNA-transfected wells without (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine was considered to be 100%.


Three siRNAs targeting STK6 (STK6-1, STK6-2, and STK6-3) showed inhibition of tumor cell growth in the presence of (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine. Among the three, STK6-1 showed the strongest growth inhibitory activity in the initial screens. To investigate whether this growth inhibitory activity was due to on or off-target activity of the siRNA, three additional siRNAs targeting STK6 were used and the abilities of all six siRNAs to induce STK6 silencing and growth inhibition were investigated. There was a good correlation between the level of STK6 silencing and growth inhibition (FIG. 1). This correlation suggested that growth inhibition was due to on target activity (i.e., STK6 disruption). Next, STK6-1 and control siRNAs to luciferase (negative control) were titrated in the presence or absence of (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine (FIG. 2). The addition of the KSPi shifted the STK6-1 dose response curve ˜5-10-fold to the left. This concentration of the KSPi did not augment effects on cell growth caused by a luciferase siRNA. In contrast, the dose response curve to an siRNA targeting PTEN (Table I) with similar effects on cell growth as STK6-1 was not shifted by the KSPi. Other siRNAs to STK6 also enhanced effects of KSPi on cell growth. Thus, disruption of KSP enhances the effects of STK6 siRNAs on cell growth. Further support for this was obtained by studies using combinations of siRNAs to STK6 and KSP, which showed greater growth inhibitory activity than either siRNAs alone. Because the concentrations of KSPi used in these experiments did not affect cell growth on its own, the effects of KSPi on STK6 siRNA activity appeared synergistic rather than additive.


The interaction between human STK6 and KSP is consistent with evidence of physiological interactions between these genes in Xenopus (Giet et al., 1999, J Biol. Chem. 274:15005-5013). In particular, the Xenopus homologues of STK6 and KSP co-localize at the mitotic spindle poles and the proteins show molecular association by immunoprecipitation. Furthermore, KSP is a substrate for STK6.


The growth inhibition by STK6 siRNAs suggests that this gene is essential for tumor cell growth and supports investigation of STK6 as an anti-tumor target. The data showing synthetic lethal interactions between inhibitors of STK6 and KSPi suggest that combination therapy with these compounds might be more effective than therapy with either compounds alone. STK6 is frequently over-expressed in human tumors, including breast cancers with poor prognosis (van 't Veer et al., 2002, Nature. 2002 415:530-536). Amplification of STK6 has been implicated in resistance to Taxol (Anand et al., 2003, Cancer Cell. 3:51-62). Since both KSPi and Taxol affect the same target (mitotic spindle), over-expression of STK6 may likewise reduces the effectiveness of KSPi. This possibility is consistent with the results showing interactions between inhibitors of KSPi and STK6, and should be investigated during the clinical development of KSPi. For instance, a KSPi may not be optimally effective in breast cancer patients with poor prognosis because of the tendency of these tumors to over-express STK6.



FIG. 17 shows results of screens for genes that sensitize to KSPi. The results demonstrate that TPX2 also interacts with KSP. The siRNA sequences used in silencing TPX2 are also listed in Table I.

TABLE IList of siRNAsSTK6-1GCACAAAAGCUUGUCUCCATT(SEQ ID NO:1)STK6-2UUGCAGAUUUUGGGUGGUCTT(SEQ ID NO:2)STK6-3ACAGUCUUAGGAAUCGUGCTT(SEQ ID NO:3)STK6-4CCUCCCUAUUCAGAAAGCUTT(SEQ ID NO:4)STK6-5GACUUUGAAAUUGGUCGCCTT(SEQ ID NO:5)STK6-6CACCCAAAAGAGCAAGCAGTT(SEQ ID NO:6)ROCK2-1AACCAGUCUAUUAGACGGCTT(SEQ ID NO:7)ROCK2-2GUGACUCUCCAUCUUGUAGTT(SEQ ID NO:8)ROCK2-3GUGGCCUCAAAGGCACUUATT(SEQ ID NO:9)CDC20-1CCCAUCACCUCAGUUGUUUTT(SEQ ID NO:10)CDC20-2GACCUGCCGUUACAUUCCUTT(SEQ ID NO:11)CDC20-3GGAGAACCAGUCUGAAAACTT(SEQ ID NO:12)TTK-1AUGCUGGAAAUUGCCCUGCTT(SEQ ID NO:13)TTK-2ACAACCCAGAGGACUGGUUTT(SEQ ID NO:14)TTK-3UAUGUUCUGGGCCAACUUGTT(SEQ ID NO:15)FZR1-1CCAGAUCCUUGUCUGGAAGTT(SEQ ID NO:16)FZR1-2CGACAACAAGCUGCUGGUCTT(SEQ ID NO:17)FZR1-3GAAGCUGUCCAUGUUGGAGTT(SEQ ID NO:18)BUB1-1CUGUAUGGGGUAUUCGCUGTT(SEQ ID NO:19)BUB1-2ACCCAUUUGCCAGCUCAAGTT(SEQ ID NO:20)BUB1-3CAGACUCCAUGUUUGCAGUTT(SEQ ID NO:21)BUB3-1UACAUUUGCCACAGGUGGUTT(SEQ ID NO:22)BUB3-2CAAUUCGUACUCCCCAAUGTT(SEQ ID NO:23)BUB3-3AGCUGCUUCAGACUGCUUCTT(SEQ ID NO:24)MAD1L1-1GACCUUUCCAGAUUCGUGGTT(SEQ ID NO:25)MAD1L1-2AGAGCAGAGCAGAUCCGUUTT(SEQ ID NO:26)MAD1L1-3CCAGCGGCUCAAGGAGGUUTT(SEQ ID NO:27)MAD2L2-1CCAUGACGUCGGACAUUUUTT(SEQ ID NO:28)MAD2L2-2GUGCUCUUAUCGCCUCUGUTT(SEQ ID NO:29)MAD2L2-3ACGCAAGAAGUACAACGUGTT(SEQ ID NO:30)DNCH1-1GCAAGUUGAGCUCUACCGCTT(SEQ ID NO:31)DNCH1-2UGGCCAGCGCUUACUGGAATT(SEQ ID NO:32)DNCH1-3GGCCAAGGAGGCGCUGGAATT(SEQ ID NO:33)BUB1B-1AUGACCCUCUGGAUGUUUGTT(SEQ ID NO:34)BUB1B-2UGCCAAUGAUGAGGCCACATT(SEQ ID NO:35)BUB1B-3GAAAGAACAGGUGAUCAGCTT(SEQ ID NO:36)LuciferaseCGUACGCGGAAUACUUCGATT(SEQ ID NO:37)KSP-1CUGGAUCGUAAGAAGGCAGTT(SEQ ID NO:38)KSP-2GGACAACUGCAGCUACUCUTT(SEQ ID NO:39)PTEN-1UGGAGGGGAAUGCUCAGAATT(SEQ ID NO:40)PTEN-2UAAAGAUGGCACUUUCCCGTT(SEQ ID NO:41)PTEN-3AAGGCAGCUAAAGGAAGUGTT(SEQ ID NO:42)TPX2UACUUGAAGGUGGGCCCAUTT(SEQ ID NO:1237)TPX2GAAAUCAGUUGCUGAGGGCTT(SEQ ID NO:1238)TPX2ACCUAGGACCGUCUUGCUUTT(SEQ ID NO:1239)


6.2. Example 2
Synthetic Lethal Screen Using shRNA and siRNA

This Example illustrates that simultaneous RNAi-mediated silencing of CHEK1 and TP53 leads to synthetic lethality in human tumor cells.


Two problems have limited the potential for synthetic lethal screening using RNAi approaches. First, the demonstration of synthetic lethality requires that a lethal phenotype induced by a defined gene disruption be observed in cells predisposed by a first hit gene loss or mutation but not in cells containing only wild-type alleles or protein. Thus for highly controlled experimentation, it is desirable to assay for synthetic lethality with matched cell line pairs that are isogenic except for the first hit gene disruption. For most of the available tumor cell lines, such matched cell line pairs have not been available. Second, attempts at creating two gene disruptions in cells by use of dual siRNA transfection has been hampered by the observation that siRNAs targeting distinct mRNAs compete with each other, effectively decreasing the efficacy of one or both of the siRNAs used. It is shown in this example that dual RNAi screens can be achieved through the use of stable in vivo delivery of an shRNA disrupting the first hit gene and supertransfection of an siRNA targeting a second gene. This approach provided matched (isogenic) cell line pairs (plus or minus the shRNA) and did not result in competition between the shRNA and siRNA. In this example, clonal cell lines with a primary gene target silenced by stable expression of short hairpin RNAs (shRNAs) were established. Transient transfection (supertransfection) of these clones with siRNAs targeting other genes did not appreciably affect primary target silencing by the shRNA, nor was target silencing by siRNAs affected by shRNAs. This approach was employed to demonstrate synthetic lethality between TP53 (p53), and the checkpoint kinase, CHEK1, in the presence of low concentrations of the DNA-damaging agent doxorubicin.


RNA interference can be achieved by delivery of synthetic double-stranded small interfering RNAs (siRNAs) via transient transfection or by expression within the cell of short hairpin RNAs (shRNAs) from recombinant vectors introduced either transiently or stably integrated into the genome (see, e.g., Paddison et al., 2002, Genes Dev 16:948-958; Sui et al., 2002, Proc Natl Acad Sci USA 99:5515-5520; Yu et al., 2002, Proc Natl Acad Sci USA 99:6047-6052; Miyagishi et al., 2002, Nat Biotechnol 20:497-500; Paul et al., 2002, Nat Biotechnol 20:505-508; Kwak et al., 2003, J Pharmacol Sci 93:214-217; Brummelkamp et al., 2002, Science 296:550-553; Boden et al., 2003, Nucleic Acids Res 31:5033-5038; Kawasaki et al., 2003, Nucleic Acids Res 31:700-707). The pRETRO-SUPER (pRS) vector which encodes a puromycin-resistance marker and drives shRNA expression from an H1 (RNA Pol III) promoter was used. The pRS-TP53 1026 shRNA plasmid was deconvoluted from the NKI library plasmid pool for TP53 by transforming bacteria with the pool and looking for clones containing only the plasmid of interest. The sequences used are as follows: pRS-p53 1026 19mer sequence: GACTCCAGTGGTAATCTAC (SEQ ID NO:43); primers for sequence specific PCR: Forward: GTAGATTACCACTGGAGTC (SEQ ID NO:44), Reverse: CCCTTGAACCTCCTCGTTCGACC (SEQ ID NO:45). Plasmids were identified by sequence specific PCR, and confirmed by sequencing. Stables were generated by transfecting HCT116 cells using FuGENE 6 (Roche) with the pRS-TP53 1026 plasmid. Cells were split into 10 cm dishes plus 1 ug/ml puromycin 48 hours later, and maintained until colonies were evident (5-7 days). Clones were picked into a 96 well plate, maintained in 1 ug/ml puromycin, and tested for knockdown by TaqMan using the TP53 and hGUS Pre-Developed. Assay Reagent (Applied Biosystems). To measure transient knockdown by the pRS-TP53 1026 plasmid, HCT116 cells were transfected using Lipofectamine 2000 (Invitrogen), and RNA harvested 24 hours later. TP53 levels were assessed by TaqMan.


Analysis of multiple puromycin-resistant TP53 shRNA clones (pRS-p53) derived from the colon tumor line HCT116 showed varying levels of target silencing (50% to 96%). FIG. 3 shows the level of TP53 expression in clones A5 and A11, which exhibited the highest levels of silencing. TP53 silencing achieved in these clones exceeded that observed 24 hr after delivery of pRS-p53 into HCT116 cells by transient transfection (FIG. 3). It is possible that transfection efficiency limits the effectiveness of TP53 shRNA in transient assays. Alternatively, cells having greater levels of TP53 silencing gain a growth advantage during clonal growth. With an shRNA that targets STK6 (pRS-STK6: pRS-STK6 2178 19mer sequence: CATTGGAGTCATAGCATGT (SEQ ID NO:46)), a range of silencing in stable clones was also observed. These clones, however, did not achieve as high a degree of silencing observed in the TP53 lines, nor was silencing greater than that achieved in transient assays. This may indicate selection against high level of STK6 silencing because STK6 is an essential gene for tumor cell growth in culture.


To test whether TP53 silencing in HCT116 clone A11 was subject to competition with siRNAs, cells of this clone were supertransfected with a pool of CHEK1-specific siRNAs. CHK1 pool contains the following three siRNAs: CUGAAGAAGCAGUCGCAGUTT (SEQ ID NO:99); AUCGAUUCUGCUCCUCUAGTT (SEQ ID NO:98); and UGCCUGAAAGAGACUUGUGTT (SEQ ID NO:100). This siRNA pool had been found to competitively reduce silencing activity of a TP53 targeted siRNA. siRNAs were transfected using Oligofectamine (Invitrogen) at 10 nM or 100 nM where indicated. For the CHK1 pool, three siRNAs were transfected simultaneously at 33.3 nM each for a total delivery of 100 nM. RNA was harvested 24 hours post transfection and knockdown was assessed by TaqMan analysis using the CHK1 or TP53 and hGUS Pre-Developed Assay Reagent (Applied Biosystems). As shown in FIG. 4A, the shRNA and the siRNA pool did not competitively inhibit silencing of each other's targets. Inhibition by known competitive siRNAs of either a transiently transfected siRNA or a stably expressed shRNA of the same sequence was then assayed. As shown in FIG. 4B, three individual siRNAs targeting KNSL1 (KNSLI 210: GACCUGUGCCUUUUAGAGATT (SEQ ID NO:47); KNSLI 211: GACUUCAUUGACAGUGGCCTT (SEQ ID NO:48); KNSLI 212: AAAGGACAACUGCAGCUACTT (SEQ ID NO:49)) competitively inhibited the silencing achieved by co-transfected siRNA targeting STK6 (left bars). In contrast, silencing by the homologous STK6 shRNA in stably transfected lines was unaffected by supertransfection of the KNSL1 siRNAs, even when the competitor siRNAs were added at ten fold higher concentrations (middle and right bars). These experiments suggested that there was little competition between stably expressed shRNAs and transiently transfected siRNAs. pRS and pRS-p53 HCT116 cells were transiently transfected with siRNA pools for ˜800 genes (see Example 3, infra) and measured effects on cellular growth by Alamar Blue assay. Nearly identical responses to the ˜800 siRNA pools in pRS cells and in pRS-p53 cells with no suggestion of competitive inhibition of silencing were observed.


Next, supertransfection of the CHEK1 siRNA pool into cells stably expressing TP53 shRNAs was evaluated to determine if it could be used to investigate genetic interactions (SL) between these molecules. This interaction has been speculated previously, but definitive demonstration of it has been hampered by lack of reagents or genetic knockouts with adequate specificity to rule out off-target effects. Matched cell lines +/−TP53 expression were generated by selecting stable clones of A549 lung cancer cell lines containing either empty pRS vector or pRS-p53. The latter cells showed >90% silencing of TP53 mRNA. Both cell lines were then supertransfected with either control luciferase siRNA (luc, 100 nM) or the CHEK1 siRNA pool (100 nM total; 33 nM each of 3 siRNAs) and their cell cycle profiles examined with or without exposure to the DNA damaging agent, doxorubicin (Dox, FIG. 5). Cell cycle profiles of pRS-p53 cells were not appreciably different from those of pRS cells in the absence of Dox. Transient transfection of CHEK1 siRNAs also did not affect cell cycle profiles in the absence of Dox. In the presence of Dox, however, pRS-transfected cells exhibited G1 and G2/M arrest as is expected of cells expressing functional TP53. Supertransfection of CHEK1 siRNAs resulted in an override of the G2 checkpoint and an increase in the number of cells blocked at G1. Because the cells retained TP53 function, they stopped in G1 and did not proceed back into S phase.


In contrast, pRS-p53 cells lost the ability to arrest at G1 and arrested primarily at G2 in response Dox treatment, consistent with the role of TP53 in the G1 checkpoint. The cell cycle profile of pRS-p53 cells was unchanged by supertransfection of luc siRNA (FIG. 5). The failure of luc siRNA to cause even partial restoration of the TP53 response (and a corresponding increase in the G1 peak) suggests that there was little competitive inhibition of TP53 silencing and phenotype by this siRNA. Therefore, competitive inhibition of TP53 silencing by the CHEK1 siRNA pool was not expected to exist. Indeed, in response to Dox treatment, pRS-p53 cells transiently transfected with CHEK1 showed profound alterations in their cell cycle profile with large increases in the fraction of cells in S and with sub-G1 (dead cells) amounts of DNA. Similar findings were also observed in pRS and pRS-p53 stably transfected HCT116 cells. Thus, simultaneous disruption of the G1 checkpoint mediated by TP53 and the G2 checkpoint mediated by CHEK1 is lethal to TP53− but not TP53+ tumor cells.


The finding that transfected siRNAs did not competitively inhibit silencing by stably expressed shRNAs was unexpected. It is presently unclear why siRNAs competitively cross inhibit silencing whereas shRNAs and siRNAs do not. It may suggest that these two types of RNAs enter the RNAi pathway at biochemically distinct steps.


FIGS. 15A-C shows results of CHEK1 silencing on the sensitivity of cells to DNA damage. 15A CHEK1 silencing/inhibition sensitizes HeLa cells to DNA damage. 15B CHEK1 silencing/inhibition sensitizes p53-A549 cells. 15C CHEK1 silencing does not sensitize HREP cells to Doxorubicin.


6.3. Example 3
Genes that Enhance or Reduces Cell Killing by DNA Damaging Agents

This Example illustrates a semi-automated siRNA screens for identification of genes that enhance or reduces cell killing by DNA damaging agents. The semi-automated platform enables loss-of-function RNAi screens using small interfering RNAs (siRNA's). A library of siRNAs targeting ˜800 human genes was used to identify enhancers of DNA damaging agents, Doxorubicin (Dox), Camptothecin (Campto), and Cisplatin (Cis). In each of the screens, many genes (“hits”) whose disruption sensitized cells to cell killing by the chemotherapeutic agent were identified (see Table IIIA-C). Some of these hits (e.g. WEE1) suggest new targets to enhance the activity of common chemotherapeutics; other hits (BRCA1, BRCA2) suggest new therapies for genetically determined cancers caused by mutations in these genes.


The library of siRNA duplexes was assembled for genetic screens in human cells. One version of the library targets ˜800 genes with 3 siRNAs per gene. This library was expended to target ˜2,000 genes, with further expansion to target >7,000 genes (2-3 siRNAs/gene). The library comprises siRNAs that target genes from the “druggable genome” (i.e., genes or gene families that have previously been drugged using small molecules). The library also comprises siRNAs that target genes from the “membraneome” (membrane proteins) to facilitate identification of potential targets for therapeutic antibodies. Tables IIIA-C list the sequences of portions of the siRNAs used in this Example. To facilitate large-scale siRNA screens using the library, a semi-automated platform was developed. Three different siRNAs targeting the same gene were pooled before transfection (100 nM total siRNA concentration). An entire library can be transfected into cells in duplicate by one person in less than 4 hrs. Each siRNA pool was typically tested 2-4 times in a single experiment and each experiment is generally repeated at least twice, usually by different individuals. Excellent reproducibility between screens done on different days or by different persons was achieved.


The goal of the screens was to identify targets that sensitize cells to commonly used cancer chemotherapeutics Dox, Campto, and Cis. Dox (adriamycin) inhibits the activity of topoisomerase II (TopoII). TopoII functions primarily at the G2 and M phases of the cell cycle and is important for resolving DNA structures to allow the proper packing and segregation of chromosomes. Campto inhibits topoisomerase I (TopoI). TopoI functions in S phase to relieve torsional stress of the advancing DNA polymerase complex. The addition of Campto to replicating cells results in stalled replication forks and DNA strand breaks. Cis causes DNA adducts and strand cross-linking. Both Cis and Campto treatments lead to replication fork arrest and possibly fork breakage, leading to dsDNA breaks and cell death.


The primary screen with each agent was performed in HeLa cells, which are TP53 deficient. HeLa cells were transfected with siRNA pools, and the drugs were added 4 hrs later. Preliminary experiments were performed to determine the concentration of each drug used; typically this was the amount required to give 10%-20% growth inhibition (EC10 or EC20). The growth of cells +/−drug was assessed at 72 hrs post-transfection.


The results of a screen with Cis are shown in FIG. 6. Table IIA shows fold sensitization by cisplatin averaged over cis concentrations of 400 ng/ml and 500 ng/ml. The graph shows the percent growth (log scale) for cells transfected with the siRNA pool in the absence of drug (X axis) versus the percent growth in the presence of drug (Y axis). Genes whose knockdown sensitizes to drug treatment fall below the diagonal whereas genes whose knockdown mediates resistance to the drug fall above the diagonal. The siRNA pool targeting BRCA2 caused >10-fold sensitization to Cis. The siRNA pool to BRCA1 caused >3-fold sensitization. siRNAs targeting kinases WEE1 and EPHB3 also caused >3-fold sensitization to Cis. A total of 15 genes caused >2-fold sensitization. In similar screens, ˜50 genes were identified in each of the Dox and Campto screens that caused >2-fold sensitization to drug (see Table IIB-C). The overlap between the different gene sets is discussed below.


It is important to point out that this screen was designed to reveal enhancers of drug activity. Since the drug concentrations used caused very little effect on cell growth, suppressors of drug activity would also cause very little effect on cell growth. Thus, as expected, we observed very few genes whose disruption suppressed drug activity. The one notable exception was that siRNAs targeting polo-like kinase, PLK, were less active in the presence of Cis. This probably reflects the fact that both DNA damage and PLK disruption cause cell cycle arrest. When cell cycle arrest is induced by the former treatment, the latter treatment is less effective.


To visualize the overlap between genes causing sensitization to the different drugs, we compared the ratios of cell growth −/+drug (fold sensitization) for the different agents (FIG. 7). Comparison of genes causing sensitization to Dox vs. Cis (FIG. 7, left) revealed that siRNAs to some genes, such as WEE1 kinase, sensitized cells to killing by both agents. In contrast, strong sensitization of cells to killing by Cis (>10 fold) was only observed with siRNAs targeting breast cancer susceptibility gene BRCA2. Comparison of genes causing sensitization to Campto vs. Cis (FIG. 2, right) revealed the same top-scoring genes with both treatments (BRCA2, BRCA1, EPHB3, WEE1, and ELK1).


The observation that WEE1 disruption causes sensitization to all three agents suggests that this kinase regulates a DNA damage response common to all agents. Biochemically, human WEE1 coordinates the transition between DNA replication and mitosis by protecting the nucleus from cytoplasmically activated CDC2 kinase (Heald et al., 1993, Cell 74: 463-474). Other studies suggest that WEE1 is a component of a DNA repair checkpoint functioning during the G2 phase of the cell cycle. Since most human tumors are TP53-deficient, they lack the TP53-regulated checkpoint functioning primarily in G1 and thus are more dependant on other checkpoints than normal tissues that express TP53 (i.e., that have normal checkpoint redundancy). Taken together, available data suggest that WEE1 offer a therapeutic target for treatment of TP53-deficient tumors whose survival is dependent on G2 checkpoint integrity. Indeed, a small molecule inhibitor of WEE1 was reported to act as a radiosensitizer to TP53-deficient cells (i.e., sensitized cells to radiation-induced cell death), although the degree of sensitization conferred by this compound was modest (Wang et al., 2001, Cancer Res. 61:8211-7). The “hits” from these screens in tumor cell checkpoint function are been tested for their ability to sensitize cell killing in other contexts: for example, by use of other DNA damaging agents, in other tumor types, and in matched cells +/−TP53 function.


The overlap in genes sensitizing to Cis and Campto is consistent with the mechanism of action of these drugs. Both target S phase and ultimately stall the progression of replication forks, leading to the formation of dsDNA breaks. In contrast, Dox functions primarily at the G2/M phases of the cell cycle. Thus, sensitization to Campto and Cis by BRCA1 and BRCA2 likely represents an S phase-specific mechanism-based sensitization. These results are consistent with emerging data on the role of BRCA1 and BRCA2 in DNA damage pathways (D'Andrea et al., 2003, Nat Rev Cancer 3:23-34). Indeed, both of these genes are now known to function in the DNA-repair pathway mediated by genes associated with Fanconi anemia; BRCA2 is identical to one of these genes, FANCD1. Cells that harbor defects in the BRCA pathway have an increased sensitivity to Cis (Taniguchi et al., 2003, Nat Med. 9:568-74). At present, cancer patients with BRCA mutations do not receive therapy that targets their genetic defects, although efforts are underway to test platinum drugs in these patients (Couzin, 2003, Science 302:592).


Taken together, these data suggest that the siRNA screens have identified a potential “responder” population for certain DNA damaging agents (i.e., BRCA pathway-deficient tumors). Until recently, it was thought that only a small fraction of breast and ovarian tumors were caused by germline mutations in BRCA genes, as sporadic tumors generally do not manifest alterations in these genes. However, recent data indicate that gene inactivation of other members of the BRCA pathway may be more widespread within sporadic tumors than alterations in the BRCA genes themselves (Marsit et al., 2004, Oncogene 23:1000-4). Future siRNA screens using larger libraries may help identify other genes whose disruption in tumors is diagnostic of sensitivity t6 DNA damaging agents. Indeed, many known and predicted DNA repair genes are represented in the expanded siRNA library (e.g., including other Fanconi anemia genes in the BRCA pathway). Appropriately designed screens may also identify other molecular targets that could benefit patients having BRCA pathway gene disruptions in their tumors.


The primary screens were carried out as follows: the siRNA library containing siRNAs to approximately 800 genes was screened for genes that modulate the effect of Cisplatin (cis-Diaminedichloroplatinum). The library was screened using pools of siRNAs (pool of 3 siRNA per gene) at 100 nM (each siRNA at 33 nM). These siRNAs were transfected into HeLa cells in the presence or absence of an <EC25 concentration (400 ng/ml) of Cisplatin.


siRNA transfection was carried out as follows: one day prior to transfection, 50 microliters of a chosen cell line, e.g., cervical cancer HeLa cells (ATCC, Cat. No. CCL-2), grown in DMEM/10% fetal bovine serum (Invitrogen, Carlsbad, Calif.) to approximately 90% confluency were seeded in a 384-well tissue culture plate at 450 cells/well. For each transfection 20 microliters of OptiMEM (Invitrogen) was mixed with 2 microliter of siRNA (Dharmacon, Lafayette, Colo.) from a 10 micromolar stock. For each transfection, a ratio of 20 microliter of OptiMEM was mixed with 1 microliter of Oligofectamine reagent (Invitrogen) and incubated 5 minutes at room temperature. Then 20-microliter OptiMEM/Oligofectamine mixture was dispensed into each well of the 96 well plate with the OptiMEM/siRNA mixture, mixed and incubated 15-20 minutes at room temperature. 5 microliter of the transfection mixture was aliquoted into each well of the 384-well plate and incubated for 4 hours at 37° C. and 5% CO2. Four different 96 well plates containing different siRNA pools were distributed at one plate per quadrant of a 384 well plate. All liquid transfers were performed using a BioMek FX liquid handler with a 96 well dispense head.


After 4 hours, 5 microliter/well of DMEM/10% fetal bovine serum with or without 2400 ng/ml of Cisplatin was added and the plates were incubated at 37° C. and 5% CO2 for 68 hours. The alamarBlue Assay was used for measurement of cell growth (see, Section 5.4.2.2). The alamarBlue assay measures cellular respiration and uses the meausrement as a measure of the number of living cells. The internal environment of the proliferating cell is more reduced than that of non-proliferating cells. Specifically, the ratios of NADPH/NADP, FADH/FAD, FMNH/FMN, and NADH/NAF increase during proliferation. AlamarBlue can be reduced by these metabolic intermediates and, therefore, can be used to monitor cell proliferation. At 72 hours after transfection the medium was removed from the wells and replaced with 50 microliter/well DMEM/10% Fetal Bovine Serum (Invitrogen) containing 10% (vol/vol) alamarBlue reagent (Biosource International Inc., Camarillo, Calif.) and 0.001 volumes of 1M Hepes buffer tissue culture reagent (Invitrogen). The plates were incubated 2 hours at 37° C. and the plate was read by fluorescence with excitation at 545 nm and emission at 590 on a Gemini EM microplate reader (Molecular Devices, Sunnyvale, Calif.) using Softmax Pro 3.1.2 software (Molecular Devices). The relative fluorescence units of the wells containing no cells were subtracted from the relative fluorescence units of the wells transfected with different siRNA pools to determine the relative fluorescence units above the background level. The relative fluorescence units for wells transfected with a siRNA pools with or without Cisplatin were compared to that of wells transfected with an siRNA targeting luciferase. The relative fluorescence units for luciferase siRNA-transfected wells with or without Cisplatin were considered to be 100%. A compare plot was generated by plotting the % growth relative to luciferase in the absence of drug on the X axis versus the the % growth relative to luciferase in the presence of drug on the Y axis.


The secondary screening was carried out using HeLa cells, A549-pRS cells and A549-C7 cells. The cells were transfected using pools of siRNAs (pool of 3 siRNA per gene) at 100 nM (each siRNA at 33 nM), or with single siRNA at 100 nM. These siRNAs were transfected into HeLa cells in the presence or absence of varying concentrations of DNA damaging agents. The concentration for each agent is as following: for HeLa cells, Doxorubicin (10 nM), Camptothecin (6 nM), Cisplatin (500 ng/ml); for the other cell lines, Doxorubicin (200 nM), Camptothecin (200 nM), Cisplatin (4 ug/ml).


The following siRNAs were employed: WEE1 pool, EPHB3 pool, CHUK pool, BRCA1 pool, BRCA2 pool, and STK6. The sequences of the siRNAs used are listed in Table IIIA.


siRNA transfection was carried out as follows: one day prior to transfection, 2000 microliters of a chosen cell line, e.g., cervical cancer HeLa cells (ATCC, Cat. No. CCL-2), grown in DMEM/10% fetal bovine serum (Invitrogen, Carlsbad, Calif.) to approximately 90% confluency were seeded in a 6-well tissue culture plate at 45,000 cells/well. For each transfection 70 microliters of OptiMEM (Invitrogen) was mixed with 5 microliter of siRNA (Dharmacon, Lafayette, Colo.) from a 20 micromolar stock. For each transfection, a ratio of 20 microliter of OptiMEM was mixed with 1 microliter of Oligofectamine reagent (Invitrogen) and incubated 5 minutes at room temperature. Then 25-microliter OptiMEM/Oligofectamine mixture was mixed with the 75-microliter of OptiMEM/siRNA mixture, and incubated 15-20 minutes at room temperature. 100 microliter of the transfection mixture was aliquoted into each well of the 6-well plate and incubated for 4 hours at 37° C. and 5% CO2.


After 4 hours, 100 microliter/well of DMEM/10% fetal bovine serum with or without DNA damage agents was added to each well to reach the final concentration of each agents as described above. The plates were incubated at 37° C. and 5% CO2 for another 44 or 68 hours. Samples from the two time points (48 hr or 72 hr post-transfection) were then analyzed for cell cycle profiles.


For cell cycle analysis, the supernatant from each well was combined with the cells that were harvested by trypsinization. The mixture was then centrifuged at 1200 rpm for 5 minutes. The cells were then fixed with ice cold 70% ethanol for ˜30 minutes. Fixed cells were washed once with PBS and resuspended in 0.5 ml of PBS containing Propidium Iodide (10 microgram/ml) and RNase A(1 mg/ml), and incubated at 37° C. for 30 min. Flow cytometric analysis was carried out using a FACSCalibur flow cytometer (Becton Dickinson) and the data was analyzed using FlowJo software (Tree Star, Inc). The Sub-G1 cell population was used to measure cell death. The siRNAs are said to sensitize cells to DNA damage if the summation of the Sub-G1 population from the (siRNA+DMSO) sample and (Luc+drug) sample is larger than the Sub-G1 population of (siRNA+drug) sample.



FIGS. 9-14 show the results of the secondary screens. FIGS. 9A-9C show that silencing of WEE1 sensitizes HeLa cells to DNA damage induced by Dox, Campto, and Cis. FIGS. 9D-9I show that silencing of WEE1 sensitizes p53− A549 cells to DNA damage induced by Dox, Campto, and Cis, but does not sensitize p53+ A549 cells to such DNA damage. FIGS. 10A-10C show that silencing of EPHB3 sensitizes HeLa cells and p53− A549 C7, and to a lesser extent p53+ A549 pRS cells, to DNA damage induced by Dox, Campto, and Cis. FIGS. 11A-11C show that silencing of STK6 sensitizes HeLa cells and p53− A549 C7, and to a lesser extent p53+ A549 pRS cells to DNA damage induced by Dox, Campto, and Cis. FIGS. 12A-12C show that silencing of BRCA1 sensitizes HeLa cells and p53− A549 C7 cells to DNA damage induced by Dox, Campto, and Cis. Silencing of BRCA also sensitizes p53+ A549 pRS cells to DNA damage induced by Cis to a lesser extent, but does not sensitize p53+ A549 pRS cells to DNA damage induced by Dox and Campto. FIGS. 13A-13B show that silencing of BRCA2 sensitizes HeLa cells and p53− A549 C7 cells to DNA damage induced by Dox, Campto, and Cis. FIG. 13C shows that silencing of BRCA2 sensitize p53+ A549 pRS cells to DNA damage induced by Cis to a lesser extent, but not dox and Campto. FIGS. 14A-14B show that silencing of CHUK sensitizes HeLa cells to DNA damage induced by Dox, Campto, and Cis. FIG. 14C shows that silencing of CHUK sensitizes p53− A549 C7 cells to DNA damage induced by Campto, and Cis. FIG. 14D shows that silencing of CHUK does not sensitize p53+ A549 pRS cells to DNA damage induced by Campto and Cis.

TABLE IIAAverage fold sensitization by cisplatinave foldGene IDGene Namesensitizationstd dev12514PLK0.3029875530.12244223099PLK0.3444426340.15722133433PLK0.4156186170.14288843266PLK0.4712585340.27341953006PLK0.5730263770.29502263534PLK0.5801353730.40306973806C10orf30.5816782840.12209883322CCNA20.6036152990.02789993805C20orf10.6180838360.081029103423NM_0061010.6400548780.131981113464INSR0.671845410.043498123722TLK20.6802016670.164793133731CSNK1E0.709719280.169767143261ERBB20.7218049970.095466153093PIK3CG0.7305176350.16341163391PLK0.735668720.438713173813ANLN0.7422866860.076826183687CAMK40.7637851820.078326193838PRKAA20.7681284770.09846120270227020.774220780.032982213435FLT30.7860696410.033061223740STK350.7862518340.241352233826NM_0156940.786686190.158833243113CNK0.7897510970.074976253648CLK10.7959624860.119858263397BUB30.7988973090.041819272982CDC2L20.8032902640.28261282975NEK40.8049729260.092313293003PER0.8067612290.283308303776NOTCH20.8076269740.090463313600RRM2B0.8077911390.116058323303CDKN2D0.8082360380.106543333536PIK3C30.8116238710.072924343491PRKCE0.8185543140.081903353181ST50.8202278770.105561363812CDCA80.8251941750.149709373525NOTCH40.8260758240.135465383182MYCN0.8269977540.074996392992PRKR0.830264110.107682402972KSR0.8407370730.220722413359TUBA10.8416562880.176344423183NM_0052000.8437550020.126232432961PIM10.8468143160.1791443814HMMR0.8485845650.089675453326CCT70.8508059080.139648463819TACC30.8510512240.151449473495FGFR20.8516580580.169414482952PRKG10.8530837440.103483493680CLK30.8531114210.029348503650NM_0251950.8557693330.097938513635STAT10.8567328190.045221523487MAP2K30.8586096430.046727533831CLSPN0.8653004470.043122543416IKBKE0.8687706940.033925553693NEK90.8718651150.272749563686MAP3K80.8723216060.276021573677HCK0.8742428620.099478583509KIF21A0.8761523480.070276593666PAK60.8773471390.070142603563RAB3A0.8773924520.07511612993SRMS0.8779144290.052743623658STK180.8844097160.022945633153RB10.8848020120.066909643000BMX0.887909350.05788653784MAPK80.8884444340.124134663503EGR10.88881580.172111673578RREB10.8894063560.126074683085KIF5C0.8897478740.062749693431NM_0184540.8930828930.124062702954ROCK20.8939337980.055935712922NM_0047830.894875870.052019723631WISP20.8957992220.04132733752CCNB30.8959030640.014712743808CKAP20.8974295320.077036753399HSPCB0.8985881230.283379763251ABL10.8997471730.09061773695PRKAA10.8999261910.099839783319CCND10.9013425960.14162793786FRAP10.9014815860.064389802964RIPK20.9016580940.057156813179PDGFB0.9023584540.054703822987RNASEL0.904859080.109916833086KIF110.9059254730.044166843610LEF10.9060264450.269465853798ACTR20.90861660.162743863088KIF13B0.9121593460.09222873332CDC5L0.9126259360.141471883711LIMK10.9128916210.150911893775NOTCH10.9146493140.049686903743RAGE0.9158754340.062887913410RPS270.9166113220.14842923403AURKC0.9171628450.112884933197ARHB0.9175496710.07581943145C20orf230.9185174480.040236952980RIPK10.9186932410.035801963646NM_0057810.9196291840.074213973256CDC2L10.9203118610.161437983171VHL0.9211971390.154964993661FGR0.9219038630.0627181002978AB0674700.9227131350.0581261012983GUCY2C0.9228910010.1324991023557JUND0.9233862310.2125161033573NM_0168480.9242555090.0257471043783KRAS20.9243358690.0319751053833ATR0.9251517960.0364591063762MCC0.9267667970.0632151072934IRAK20.9271375420.0900481083311CDK100.9274874930.1973031093230MAP2K10.9295282920.0878661103461KIT0.9298646070.0651051113581RASGRP10.9300463340.0859361123782SOS10.930782760.0869571133348DCK0.9329345790.1409271143518NFKB10.9335380420.2547761153692AB0079410.9340314790.1228911162936SGKL0.9352688560.128691173788PRKCE0.9358254590.1004371183791NM_0052000.9373731510.1245511193827NM_0181230.9387526870.1208851203343CENPJ0.9392763610.150641213413KIF230.9407192230.2244761223540PPP2CB0.9408255490.077861233559RAP1GDS10.9411860980.0923181242943DYRK20.9417515870.0797681253090KIF3C0.9429947130.0431871263306CDC14A0.9431592120.1053141273572RASA30.9437563860.0449241283822GTSE10.9447555560.23321293351ESR10.9449203780.1536221303258MOS0.94603370.0902051313601POLE0.947082410.1267311322960LYN0.9473228770.191481333828KIF20A0.9507735580.1839381343778VHL0.9519388610.2324811353196ARHI0.9528422480.0586811363566JUN0.952940250.1272851373240MAPK120.9535647170.0715861383184TSG1010.9540021380.048231393714NM_0133550.9541978850.124881403364HPRT10.9544143940.2717711413685LTK0.9544433020.2853981423751BCR0.9544674510.1210041433434DDX60.9547908430.0829731443298CCNE10.9551132810.0801491453449TBK10.9553016320.0189691463795NR4A20.9555572770.0966861473739NM_0178860.9555816370.1037711483471MAPK100.9567055190.0687651493139XM_0958270.9569936280.2173271503545IRS20.9578611160.0586381512985MKNK10.9587842740.027551523618DVL20.9588604280.1459171533726MAPKAPK20.959228530.0712821543678PFTK10.9607094640.0434351553821ASPM0.9612209450.1290441563163THRA0.962043760.1380311573101MAPK140.9621949670.0897721583561FOS0.962200970.0383941593133XM_1680690.9623555450.0751191603443EPS80.9626705090.0802841613117ATM0.9634481580.176841623401HDAC10.9635949210.0530871633799ACTR30.963851530.1062811643733MYLK20.963905860.0719561653801PSEN10.964323090.1333991663716ULK10.9647143740.1727561672977RIPK30.9653214880.2880061683571VAV10.9660857910.0406961692946NM_0177190.9667268540.0704161703459EGFR0.9684751970.039891713835CHEK20.9684924790.0773941723125NM_0312170.9687058780.1588151733308CDKN2B0.9704546970.0303161743458ARAF10.9721265140.1503831753162MADH20.972287490.0772511762949MYO3B0.9736366180.069161773664STK17A0.9753433120.068111783488AURKB0.9757421320.1783211793112KNSL70.9766651030.1579111803485DHX80.9780535960.0732621813809CDCA30.9790022650.2311811823161WT10.9792216930.1148381833513ROS10.9792715770.1215891843185VCAM10.9794382570.0697591853553CKS1B0.9794654690.055551863763NM_0162310.9809905740.1235551873245AXL0.9810227830.0787241883334CUL4B0.9814858930.0484621893193FGF30.9815150570.0759821903335CDK5R20.9831881370.0955351913455MAP2K40.9843832990.0959211922925FYN0.9845975350.1776111933215MAD2L10.9846742890.1668171943519NTRK10.9856255260.225903195254125410.9856585290.029341963109KIF1C0.9858915360.1625831973792ARHGEF10.9859863940.1505031983374POLR2A0.9868753980.1746751993362NR3C10.987113750.092492003231ILK0.9875001240.0689422013166PMS10.9875934760.0400162023703AK0245040.9882389470.0783142033707TXK0.989004850.1381862043323CDK5R10.9895956040.1763762053180CD440.9901210580.0904132063630WISP30.9902256270.0716312073576GRAP0.9905053460.1209592083800CHFR0.993696920.1174292093142KIF250.9939323420.0440872103160TACSTD10.9944472650.1285132113497EPHA80.994467710.0152062123757CLK40.9956832840.1668592133645CASK0.9963957270.079592143357PRIM2A0.9980923710.1172472153594RAP2A0.998148420.1428182163796ARHGEF60.9985773670.0914132173767FZD30.999211320.0963952183715CDC42BPA0.9995248480.1963892192938ALS2CR70.999666530.0077182203419RFC40.9997564760.07934222163M15077102223672SYK1.0000943060.0283162233832ATM1.000190020.0915462243627CTNNA11.0002914590.2154532252984EPHB61.0006039480.0980442263200REL1.0006165850.1044642273492PRKCQ1.0007850850.1031812283478EPHA21.0017569950.1014442293539PLCG21.0020080860.0723052303378NM_0060091.0029128610.0198862313381POLR2B1.0036530730.0215422323452JAK11.004100170.2469162332926AF1722641.00436010.1952912343641TYRO31.0050629540.131442353750CAMK2A1.0058795190.1979812363595FEN11.0061077130.15592373375AHCY1.0068473570.0989142383367DHFR1.0076974090.0484762393555RASA11.0079073570.0601072403246RPS6KB11.0082957050.0981992413551STAT31.0086977760.0695592423708RPS6KC11.0087380040.1585392433820NM_0184101.0088034410.004232443548RAC11.0090006640.109052453527DTX21.009403990.0827672463339CCNB21.0096253250.3214342473226RBX11.010291590.2359692483473DAPK11.0103353940.0652662493469AAK11.0116530850.1538192503517MYC1.0118557570.0880322513005MERTK1.0119102660.1391122523294CCNF1.013554020.1512172533392BIRC51.0140185750.1472922543533HES71.0168689540.2092442553524NOTCH31.0172854720.0688772563587VAV31.0181291730.0627372573425DLG71.0182648270.0373252583674CSNK1D1.0186506990.0875212593380TUBG21.0192484320.0276972603486RPS6KA31.0199855270.0500312613746HUNK1.0207799180.0823722623535SKP21.0211429530.1000642633797ARHGEF91.0216355620.1377832642969NM_0149161.0218878110.0804672653460CSK1.0220853660.1358052663132KIF231.0238067820.1294962672963MAP3K111.02428730.0652232683702MAP3K131.0242948740.0830142693382TUBB1.0259156080.0499372703237CDC71.0259946030.0964092713592SOS21.0262355130.1789952723365PRIM11.026538550.1047982733570RALGDS1.0274606970.0848732743224FBXO51.0291555840.1545452753585GAB11.0294815260.060772763414HDAC7A1.0304248950.1395872773514HRAS1.0304816710.092812783597SHMT21.0312079970.1808272793657PCTK11.0318391280.0678282803257IGF1R1.031927290.102642813773WNT21.0323097310.1740042823625CTBP21.0325380090.1590782833302CDK81.0327605450.0777712843409TTK1.0331135170.0893832853465EPHA11.0335164870.1278092863705NM_0121191.0337513640.1079482872966NM_0332661.0350123350.0976412882999FES1.0355585820.127252893474CSNK2A11.0361512180.0850572903824MAPRE31.0361978380.0927062913094KIF3A1.0364649420.1199212923769PLAU1.0373902110.0648932933213NM_0162381.0377459090.1387862942950NEK61.0382918540.1497762953815MAPRE21.0383059470.2177092963543PDK21.0383112210.1976792973437FGFR11.03832690.2836432983542PPP2CA1.0393576710.1943522993511XM_1680691.0393709130.1552623003002CRKL1.0399839710.1011293013398HDAC111.0416639340.0994063023675ADRBK11.0417414590.0844193033623CTNND11.0422102380.1059783043268CDC25C1.0427623570.027263053633CTBP11.0428185690.1431713063804NM_0243221.0428955730.052663073526HES61.0431467870.0592443082947NM_0070641.0434566890.0803053092979PAK21.0435377930.1151883102959PIM21.0439420640.0503523113602MCM31.0440711080.238653123665PAK41.0442465230.0529213133421ORC6L1.0448254230.2417263143745CAMKK21.0449660320.0328443153736PTK71.0450087770.1189653163119CDKN1B1.0451547490.0268033173643DDR21.0457964260.1237483183603POLS1.0467962830.0902123193346CCNK1.0477374420.1481523203438DTR1.049750540.1396193212942TTN1.0509443860.1345753222937NM_0250521.0515934480.0521183233577RAB2L1.0519772480.0739923243203ITGA51.0521970110.1094433253599DTYMK1.0522068960.1470413263373TOP2A1.0539469260.0610713273222PTTG11.0549344650.0597343283154MADH41.0553671420.3922853293829KIF2C1.0560174380.1876843303652PDGFRA1.0560200560.0845373312944MARK11.0564915680.1612323323656PRKCN1.0567558780.1779433333626DVL31.0587112690.196473343802NOTCH31.0590319180.1174953353127MAPK11.0594412610.0704493363549PIK3R21.0594954930.1786973372935MAPK61.0605337090.0750313383307CDC61.0608582360.0939333393260STK111.0618484450.1207623403766S100A21.0628320730.1745763413457BAD1.0639447910.076373423347TOP11.066144810.1697483433450MAP3K21.0666389710.1668693443164MYCL11.0666669640.1985323453412KIF251.0685361130.2028873463317CCNI1.0689664640.1261883473550PLCG11.0690528940.1230643483668DAPK31.0691202780.166973493454FLT41.069851220.1298073503394HDAC61.0701687650.0506173513122ATSV1.0712918710.1266753523169NME11.0713533820.0629213533342CCNT11.072082870.0306243543523NOTCH21.0722358010.0968083553591RALB1.0726371910.1312853562970AATK1.0734606820.0791163573593VAV21.0736492350.0873723583489SRC1.0746210490.0963473593363GART1.0763808910.079193603097KIF20A1.0777416280.0651923613494MAPK41.0779228950.0725493623114PIK3CD1.0780957520.1188453632976NEK71.0781082860.5431363643352NR3C21.0785247450.2007143653115MDM21.0794081630.1091663663108KIF221.0803268140.0896863672973NEK11.0805465270.2103343683219CENPC11.0806377030.2115863693583JUNB1.0808286820.0611823703476PRKCD1.0814219320.0637053713717NTRK21.081845510.1793593723760CDKL51.0820319570.1228573733744PRKWNK41.0828216950.0410893743147CDKN2A1.0831747680.1425563753170BLM1.0833967070.0871033763390NM_0809251.0838140730.1873063773691NM_0240461.0840079510.4552023783682DYRK1A1.0853830770.131643793338CUL4A1.0856961660.1137523803445BMPR1A1.086530480.2173883813639STAT61.0871722410.2407113823683NM_0031381.0877656270.1074823833694STK381.0889257690.153093843228CDC271.0895464610.2304383852923ERN11.0900526820.1605033863366TYMS1.0907849890.1578413873816NM_0177691.0908760670.1706193883107KIF21.0913008750.0821853893262LATS11.091489380.0589193903188PMS21.0920502130.1407273913498CSNK1A11.0927069430.0599833923293CDC25A1.0929864020.0992273933721ANKRD31.0931274670.1141013943793MAPRE11.0934144580.1075173953305CDC2L51.0950699910.0589693963647YES11.0952201180.4391753973324CUL51.0952537580.1094643982965NM_0147201.0958614280.2958523993300CDC14B1.0959008120.0532764003296CDKN2C1.0967285870.060434013724EPHA71.0967799370.208144023165FGF21.0992048650.0524424032928IRAK11.0995448460.117054043502PRKCH1.0997958020.0764934053728TIE1.1004080420.0597594063424EZH21.1004144290.1379944073756CDK5RAP21.101487940.1691724082920EIF2AK31.1018746790.1935174093556RAP1A1.1026033530.2166294103214CENPF1.1026660550.2295654113102CKS21.1034900840.2761094122974NEK111.1035757210.386624133297CCT21.1039745290.0753864143393HDAC21.1044728610.0747074153568PLD11.1048123110.0438744163470RPS6KA11.1049272240.1215094173496EIF4EBP21.1050813320.0260614183432PRC11.1050878330.1095144193446PRKCG1.1053758170.113564203512TGFBR11.1069701970.083514213749NM_1390211.1071766070.0609564223807SPAG51.1072001520.1903754233579PDZGEF21.1083844920.1063744243422SMC4L11.1089673430.1684624253830NM_0132961.1096202310.1242874263537EIF4EBP11.1108339690.0900694273684STK38L1.1108354420.1275174283681SRPK11.111260950.1383194292990NM_0151121.1115409670.2900524303605FZD41.1116057050.1100014313477FGFR41.1118987610.0650074323490ERBB31.1136056540.0882784333575LATS21.1138699570.1213254343755CDKL31.1143629340.2390224353205NM_1392861.1146499420.2439354363105BUB11.1147279350.211324373389NM_0529631.1148303380.0921644383110KIF13A1.1161955090.0730394393608MAP3K7IP11.1173245130.1932664402957TYK21.117880430.1203234412996MAPK31.1179726890.3071634423628CTNNBL11.1185484290.0922344433624CTNNB11.1186091660.1709844443159RET1.1188677670.0291284453120PIK3CB1.1191353160.2226044463742RHOK1.1192967160.1666134473136XM_0666491.1194631010.1306164483328CCNC1.1194896730.0672014493199NF21.1197656370.0708054503309CCND21.1213334310.1469374513143NM_0175961.1216233680.079954523208ZW101.1219022850.1442794533753CDK51.1234276290.1308214543001PRKY1.1254569420.1649374553729RYK1.1256231620.1965784563156MSH21.1259918190.1286434573253PRKCA1.1263525970.0976874583607TLE11.1263888770.2555054593818AI3384511.1264472430.0853074603530NOTCH11.1279395590.1281554613141NM_1457541.1294792670.0263464623768ARAF11.1297052880.0869724633596SHMT11.1298185140.0461774643653NPR21.1298533770.1847094653640STAT5B1.1325896350.2996674662924STK251.132703960.0836954673356TUBG11.1336237410.2483714683008SGK21.1353730860.095694693499GRB21.1354044570.1745834703506XM_0958271.1358236020.0584834713770TGFBR21.1360617750.2830984723441PRKCI1.1377124940.1749464733609FZD31.1380826850.1808034743370AR1.1393366440.1143554753126KIF3B1.1395885480.0949144763508KIF251.1405737180.1587384773233ROCK11.1405845590.2363834782941DYRK31.1420525490.1389364793336CDC371.1421739190.1327654803741RPS6KB21.1422530820.1148894813546INPP5D1.1426462820.177324823350ADA1.142705220.2020274833759NM_0066221.1435284360.0532714843149TP531.1441169680.0286644853310CDC341.1450012460.1247534863267CCNH1.1452031210.0817784873638STAT5A1.1452840220.1820154883564RALBP11.1453027660.1877264893360RRM21.1453717510.1062324903662LCK1.1456387470.0911844913223NM_0162631.1456676140.1606734923408PIN11.1465393590.1009544932986ACVR21.1466618130.105124943304CCNE21.1467956540.1027694952997MST1R1.1472218660.2831634963194RARB1.147779130.3304334973669NTRK31.1482229270.0375664983616FZD11.1484739230.2428764993255CDK71.1485535870.169515003238MAP3K31.14898970.0671235013613DVL11.149017780.0826475023614CTNND21.149370050.1879885033318CUL21.1502677830.0780135043644EPHB11.150718960.1232575053567SHC11.1515879890.1242275063116KIF5A1.1520390390.2804225073148LIG11.1521831280.1908955083765CREBBP1.1545894090.1287125093232KDR1.1565810970.111535103748NM_0165071.1571877620.1875515113428ECT21.1573831050.1711415123649CAMK2B1.1574155030.0514725133426TK11.1580485590.1614585143250CHEK21.1584732010.0997375153636STAT21.1584955670.1618755163187WNT7B1.1585905590.0242175173505STK61.1591465770.0584385183341APLP21.1608012390.1961695193606CREBBP1.1613264050.0646955203263CDC21.1615708490.0956065212939TLK11.1630527190.1107795223123AKT31.1631458740.3068155233615FZD21.163224220.1693395243688GUCY2D1.1643216630.1528015253379NM_0325251.164469750.0748025263710GPRK2L1.1649001420.1124465273611CTNNAL11.1662579310.0373355283521MET1.1680139180.2329235293659NM_0159781.1695236830.0563925303582GRAP21.1705734920.0551185313562RASD11.1712296990.1011955323723NM_0184011.17185120.2397475333130FRAP11.1720729280.0297795343772RPS6KB11.1728239340.0665185353333CCNT21.1742356420.2147325363501RPS6KA21.1757815340.1930385373803MPHOSPH11.1760119710.1288645383248JAK21.1760209770.1763455393538NFKB21.1761298030.0523535403732CSNK2A21.1775216110.2312675413730TESK11.1779042680.2125265422989ACVR1B1.1785781610.2174925433327CDC45L1.1802041580.2993575443301CCNB11.1808641240.1629925453092KIF121.1819376050.0887085463239CDK61.1821579040.0610445473190WNT41.1826976760.0726445483811NM_1525241.1831917010.141875492940DCAMKL11.1844919380.1246985503761WT11.1845477960.161295513439EGR21.1856711360.1052845523295CDK2AP11.1870455360.2068565533817NM_0190131.1877807430.0821165543754CDKL21.1881230770.1228775553663ALS2CR21.1882464040.1407775563718PTK61.1887845860.0677815573236PTK2B1.1898185320.3523995583475EPHB41.1898884770.1054065593211BUB1B1.1898968240.2928865603411HIF1A1.190696660.2458835612927MAPK131.1907109160.1296335623264CDK31.1910422670.1003355633207MAD1L11.1912325460.0922665643372TUBA81.1915405930.0583855653349IMPDH11.1919679830.2255055663353PGR1.1923603990.0195295673252NEK21.1926016350.2824455683515PDGFRB1.1926408730.0576785693216CDC201.1930401810.0771435702971DAPK21.193066260.0853665713552PDK11.1942182910.0646735723823NM_0177791.1948610640.1383965733528TCF31.1958511970.123895743201RARA1.197395210.0879825752945CDC42BPB1.197531470.0875665763634BTRC1.2010763390.1753565773377NM_0060881.2027200910.0964115783781SRC1.2029318140.3311395793516ARHA1.2031092560.1593425803700AB0377821.2043297710.4027065813699NM_0328441.2076232660.2487035822931MAP4K31.2118546330.1496735833189MYB1.2120035690.1176065843586RASA21.2121661420.2107115853836TP531.2121838990.1691525863206ANAPC51.2135457460.0792565873701STK101.2144127530.231195883210NM_0133661.214505990.3079135893472MAP3K51.2150425610.1281345903371NM_0060871.2160594570.108045913825NM_1525621.2161089370.1533455923106KIF91.2171617370.2774455933249MAP2K61.2173826490.234085943186ETS11.2184288950.1258095953541PKD21.2203743840.2523015963654VRK21.2212660950.1801335973151MLH11.2219771950.1005295983325CDKN1C1.2225738950.1835555993774CDC45L1.2233734960.1703356003354RRM11.2241555020.1632186013225NM_0133671.2263600750.3772686023837PRKAA11.2268673110.2600996032930ITK1.2274380860.1341026043118NM_0325591.228256480.0375646053316CCNA11.2286670930.2039356063651VRK11.2290291590.1552086073368TOP3A1.2290324230.141346083376AGA1.2313631350.1280586093735PRKACB1.2315348490.0924366103007MAP3K141.2342316750.175516113420NM_0141091.2348909890.3705286123131KIF1B1.2351859890.2420876133444NEK31.235205170.3583856142919OSR11.2370862360.1065626153128AKT21.2424076110.1243946163810AI2786331.2431267350.1651376173337CDKN1A1.2446280230.0187976183091KIFC31.2447577330.1536246193191WNT21.2448173110.1872826203146KIF21A1.245725790.0412676213220ANAPC111.2461979870.179886223785GRB21.2489715570.1084916233195CDH11.2502598590.1861526243500SGK1.2519147880.0592996253103PIK3CA1.2522486060.0680436263507NM_1457541.2526897210.2229516273565RAB21.2535869020.1865526283462TGFBR21.2564599960.1360166293229PRKCL11.2566498760.1534196303790ERBB31.2603536330.1045866313704ACVR2B1.2618574330.0759946323340CENPH1.2627863260.1312156333598PCNA1.2656677790.1160326342967NM_0166531.2669231950.2327716353725EPHA41.2674389930.190566362932MAPKAPK31.2686439450.0253326373167S100A21.2708599990.0692966382994MATK1.2711547350.1280186393315CCT41.2723551920.3090656403344CDKL11.2725363830.2731556413689BLK1.273878950.2183066423104CDK41.2765784460.1617166433604TK21.2779129470.1018016443209MAD2L21.2780381140.2539766453554PIK3R31.2802843140.2283536463218CDC231.2804839470.3343816473670MAP3K101.2806497540.1291666483532NM_0190891.2808723310.1381316493558RALA1.2823432130.1931646503440FGFR31.2832779490.2789466513779CTNNA11.2850660690.058536523312CUL31.2860866630.0951716533111KIF5B1.2861559630.0454546543320CCND31.2864276990.0497816553493MAPK91.2867085550.2042546563463TEC1.2867313530.1163466573198ICAM11.2870872110.1051646582933MAP4K51.2888487140.195886592995PTK21.2897812270.1220066603637STAT41.2914780710.1267656613089KIFC11.2922966310.1041856623330CDK91.2931899890.2003326633588RHEB1.2957869150.1139226643589SOS11.3008349590.0288466653418CENPA1.3008513560.2246486663314CCNG11.3021320750.1670186673697CAMK2G1.3055212880.1415826683620AXIN21.3068812350.1757256692921RPS6KA51.3078957880.1169766703157NF11.3123640050.219796713172PLAU1.3142197650.2213956723221TOP3B1.3167677240.1537326733529DTX11.3171041310.1000426743520NRAS1.3181623790.3377986753138KIF171.3190213320.0472396763466JAK31.3245908570.3419236773447PRKCM1.3258527820.0901646783396HDAC101.3270360670.0954216793405HDAC81.3289863830.1374566802956PRKCL21.3297599870.2775446813771PIK3CA1.3309278540.3186056823100GSK3B1.3326618430.1720856833140XM_0890061.3346346880.2101996843417HDAC31.3347391360.2137356852912MPHOSPH11.3356068170.1922466863453MAP2K21.3371781840.2311336873777ABL11.3379463550.133816882991NPR11.3396685280.2137196893234CDK21.3413786320.3276036903617CTNNBIP11.3441299690.1721196913217NM_0148851.3466421040.375786923632WISP11.347986210.2675846933404PPARG1.3506082260.3287996943834CHEK11.3536828070.1773326953244PRKCZ1.3545064470.4235696963242PRKCB11.3561101770.1778566972998MAPK71.3579150270.3209186983227NUMA11.3580335670.3362066993676MAP4K11.3606242020.356657003087PTEN1.3610430770.2218037013734BMPR1B1.3627517450.196637023569RASGRP21.3628527130.0837467032953MAPK111.3674175830.3354117043355GUK11.3688548880.1213287053713PRKG21.3707627530.0962817063415HSPCA1.3731023910.3116377073212NM_0226621.3738452060.36437083789ELK11.3782932970.1192767093395HDAC51.3809185090.3161187103448NM_0162311.3829816390.2503467113737NM_0164571.3843930780.350167123456FLT11.3870010710.1175737133696NM_0162811.3925132470.1799227143124KIF4A1.3927744730.3959317153451MAP3K41.393596150.2153287163738PRKWNK31.3951970420.1169477173719BMPR21.3956769780.0839417183429MCM61.3996240470.4224757193243NM_0042031.4012786630.3036757203660DMPK1.4026047450.2030117213084KIF141.4056674440.0229097223574SH3KBP11.4080960570.0800557233137KIF26A1.4103972980.2092717243671STK41.4104821570.3066997253202MCC1.4107757730.2858787263134XM_1707831.4154827220.2269177273204CDC161.4157802910.2219627283121PIK3C2A1.4159500120.1523567293321CDKN31.4161767250.038267302951AJ3117981.4167699380.1772397313504PIK3CB1.4175929550.1746327322955PAK11.4183417220.073627333612TCF11.4212152060.0996377343655CAMK2D1.4229939880.2270427353135XM_0640501.427774710.2050427363400BCL21.4323420010.203887373794WASL1.4326659960.0937567383667NM_0165421.4342499050.1744787393407HDAC91.4375400110.1948917403430STMN11.4379432270.3130777413698ADRBK21.4409322230.2482177423547FOXO1A1.4611021510.1135687433265RAF11.4633158460.1911027443690PRKAA21.4706967950.2338277453510CDK41.4872898180.0893187463254CSF1R1.4879460960.4803797473622FZD91.495264230.0865997483544IRS11.4956622110.0405127492948MYO3A1.49708510.0812597503467MAP2K71.4979282870.2812537513096AKT11.4982690530.1140847522968STK17B1.5043463660.4134987533402HDAC41.5081197620.2130897543764NOTCH41.5105511970.0815867553621CTNNA21.5110562950.1116497563168DCC1.5134095820.192599757270127011.513482110.0833917583629AXIN11.5207341180.1741787593361IMPDH21.5236310110.0678737603129STK61.5271034370.1345047613679CLK21.536261190.447387623709X954251.5378273870.4376767632962MAP4K21.5478931130.3290217643442ERBB41.5510089530.1468037653247NM_0184921.5528028460.1370337663720AB0023011.5532586330.3138917673584RASAL21.5634056080.1373367683299CUL11.5899136590.1699097693522KRAS21.5906610170.068417703590ARHGEF21.5979509270.2525267713406TERT1.6001691720.0940967723259MAPK81.6019900570.3338837733369NM_0070271.6050900710.1621727743787FZD41.6214978810.0536397752929CHUK1.6460576790.1117167763468ABL21.6520076020.1808027772988FRK1.6531528710.2988827783758RAD51L11.6624232930.1356757793531NM_0211701.6669891540.0942067803155ATR1.6805717150.3886877813747GSK3A1.6886377130.389537823144KIF4B1.6958738910.4672587833235CHEK11.6979108250.3562247843313CCNG21.7036511140.2162667853004MAP3K11.7214922220.3764387863619FRAT11.7614469150.2920317873192WNT11.7650377480.3940637883673DDR11.7700539780.23387893358TOP2B1.8002937020.1957547902981ALK1.843489010.2083387912958PRKACA1.8891429340.4947737923152APC1.8946940060.1913587933712RPS6KA61.9571450810.4212927943436BRAF1.9998257371.1732087953727GPRK62.0446057436.2568067963780MCM32.0628931910.1870387973329CDC422.1316936290.4833927983095KIF2C2.1638344670.2896857993098CENPE2.1704565590.1200258003331CDC25B2.1993407510.4847168013706C20orf972.3778228090.6783298023580ELK12.4561957890.4340438033241WEE12.667552350.6252318043642EPHB32.7580931540.5652568053158BRCA12.8780716850.4183588063150BRCA211.616336981.101248









TABLE IIB










Average fold sensitization by camptothecin















fold



Gene ID
BIOID
GENE
sensitization
















1
63
M15077
1



2
2514
PLK
0.029197



3
2540
2540
#DIV/0!



4
2541
2541
0.860453



5
2701
2701
0.034091



6
2702
2702
0.432441



7
3391
PLK
0.052632



8
3534
PLK
0.083815



9
3099
PLK
0.090142



10
3006
PLK
0.09146



11
3266
PLK
0.096774



12
3433
PLK
0.13



13
3322
CCNA2
0.264029



14
3154
MADH4
0.361653



15
3518
NFKB1
0.372726



16
3600
RRM2B
0.381056



17
3184
TSG101
0.432287



18
3348
DCK
0.446467



19
3332
CDC5L
0.451264



20
3812
CDCA8
0.453177



21
3423
NM_006101
0.478261



22
3464
INSR
0.480578



23
2961
PIM1
0.51581



24
3661
FGR
0.517647



25
3171
VHL
0.524194



26
3809
CDCA3
0.529046



27
3525
NOTCH4
0.534058



28
3093
PIK3CG
0.557692



29
3740
STK35
0.55782



30
3435
FLT3
0.56



31
3805
C20orf1
0.564035



32
3219
CENPC1
0.575465



33
3003
FER
0.579832



34
3183
NM_005200
0.580153



35
3374
POLR2A
0.583796



36
3601
POLE
0.588331



37
3112
KNSL7
0.597685



38
3489
SRC
0.606833



39
3478
EPHA2
0.608258



40
3422
SMC4L1
0.608696



41
3357
PRIM2A
0.611218



42
3262
LATS1
0.613321



43
2987
RNASEL
0.617089



44
3123
AKT3
0.618574



45
3687
CAMK4
0.61913



46
3303
CDKN2D
0.625741



47
2966
NM_033266
0.627321



48
3226
RBX1
0.632166



49
3509
KIF21A
0.634426



50
2999
FES
0.634596



51
3517
MYC
0.637624



52
3592
SOS2
0.640343



53
3139
XM_095827
0.642535



54
3105
BUB1
0.643861



55
3397
BUB3
0.644077



56
3267
CCNH
0.64503



57
2975
NEK4
0.645485



58
3766
S100A2
0.646739



59
2936
SGKL
0.64695



60
3524
NOTCH3
0.647109



61
3806
C10orf3
0.64878



62
3448
NM_016231
0.654135



63
3461
KIT
0.662033



64
3501
RPS6KA2
0.667039



65
3494
MAPK4
0.668898



66
3251
ABL1
0.675159



67
3103
PIK3CA
0.679543



68
3572
RASA3
0.681105



69
3246
RPS6KB1
0.681548



70
3230
MAP2K1
0.683985



71
3733
MYLK2
0.684534



72
3491
PRKCE
0.685882



73
2982
CDC2L2
0.687831



74
3542
PPP2CA
0.690237



75
3350
ADA
0.692046



76
3651
VRK1
0.692308



77
2937
NM_025052
0.693089



78
3007
MAP3K14
0.694169



79
3751
BCR
0.694278



80
3410
RPS27
0.695238



81
3240
MAPK12
0.696498



82
2949
MYO3B
0.698039



83
3413
KIF23
0.701413



84
3773
WNT2
0.702997



85
3762
MCC
0.706533



86
3731
CSNK1E
0.707275



87
3778
VHL
0.707386



88
3476
PRKCD
0.708251



89
3754
CDKL2
0.712665



90
3741
RPS6KB2
0.715261



91
3744
PRKWNK4
0.716089



92
3148
LIG1
0.71816



93
2964
RIPK2
0.71873



94
3486
RPS6KA3
0.71875



95
3772
RPS6KB1
0.722315



96
3193
FGF3
0.723179



97
3363
GART
0.723732



98
3438
DTR
0.725061



99
3351
ESR1
0.725395



100
3416
IKBKE
0.726044



101
2972
KSR
0.727171



102
3326
CCT7
0.727769



103
3648
CLK1
0.728232



104
3401
HDAC1
0.728268



105
3498
CSNK1A1
0.728714



106
2976
NEK7
0.729805



107
3347
TOP1
0.731826



108
3236
PTK2B
0.736339



109
3256
CDC2L1
0.738272



110
3606
CREBBP
0.738487



111
3657
PCTK1
0.739866



112
3452
JAK1
0.745847



113
3250
CHEK2
0.745919



114
3200
REL
0.746919



115
3403
AURKC
0.747841



116
3663
ALS2CR2
0.749671



117
3208
ZW10
0.75



118
3647
YES1
0.750637



119
3466
JAK3
0.750708



120
3196
ARHI
0.75402



121
3757
CLK4
0.757793



122
3434
DDX6
0.758671



123
3460
CSK
0.759454



124
3722
TLK2
0.761568



125
3306
CDC14A
0.761859



126
3412
KIF25
0.761959



127
2926
AF172264
0.762856



128
3382
TUBB
0.763294



129
2965
NM_014720
0.763727



130
3625
CTBP2
0.763827



131
3702
MAP3K13
0.764125



132
3650
NM_025195
0.764957



133
3323
CDK5R1
0.765293



134
3653
NPR2
0.765609



135
2997
MST1R
0.767068



136
3658
STK18
0.768411



137
3739
NM_017886
0.768662



138
2993
SRMS
0.768678



139
3166
PMS1
0.769717



140
3775
NOTCH1
0.770983



141
3469
AAK1
0.772082



142
3833
ATR
0.772423



143
3211
BUB1B
0.773389



144
3557
JUND
0.773496



145
3179
PDGFB
0.777522



146
3674
CSNK1D
0.779923



147
3566
JUN
0.780371



148
3341
APLP2
0.781888



149
3188
PMS2
0.785359



150
3633
CTBP1
0.786631



151
2923
ERN1
0.787194



152
3086
KIF11
0.787201



153
3688
GUCY2D
0.787284



154
3605
FZD4
0.787879



155
3640
STAT5B
0.789018



156
2974
NEK11
0.791024



157
3473
DAPK1
0.791285



158
3376
AGA
0.791586



159
3263
CDC2
0.792593



160
3475
EPHB4
0.797463



161
3346
CCNK
0.79871



162
3298
CCNE1
0.800418



163
3359
TUBA1
0.801205



164
3609
FZD3
0.806613



165
3201
RARA
0.808157



166
3394
HDAC6
0.810106



167
3770
TGFBR2
0.810897



168
3258
MOS
0.811566



169
3541
PKD2
0.811594



170
3822
GTSE1
0.814495



171
3450
MAP3K2
0.81592



172
3577
RAB2L
0.816



173
3203
ITGA5
0.817391



174
3838
PRKAA2
0.821543



175
3085
KIF5C
0.82316



176
3477
FGFR4
0.824427



177
3573
NM_016848
0.824468



178
3836
TP53
0.825022



179
3782
SOS1
0.825161



180
3366
TYMS
0.828914



181
3381
POLR2B
0.828921



182
3710
GPRK2L
0.830756



183
2934
IRAK2
0.830809



184
3364
HPRT1
0.831103



185
3182
MYCN
0.831349



186
3783
KRAS2
0.831863



187
3113
CNK
0.834672



188
3835
CHEK2
0.836402



189
3680
CLK3
0.836728



190
3131
KIF1B
0.83697



191
3088
KIF13B
0.838299



192
3581
RASGRP1
0.839735



193
3829
KIF2C
0.840215



194
3380
TUBG2
0.840866



195
3334
CUL4B
0.842773



196
3746
HUNK
0.84279



197
2921
RPS6KA5
0.845122



198
3769
PLAU
0.845466



199
2984
EPHB6
0.847067



200
3814
HMMR
0.850166



201
3623
CTNND1
0.850309



202
3444
NEK3
0.851852



203
2935
MAPK6
0.852713



204
2996
MAPK3
0.853188



205
2969
NM_014916
0.856081



206
3120
PIK3CB
0.856195



207
3107
KIF2
0.856252



208
3502
PRKCH
0.856893



209
3763
NM_016231
0.858333



210
3419
RFC4
0.858657



211
3639
STAT6
0.858685



212
2930
ITK
0.860156



213
3124
KIF4A
0.860439



214
3209
MAD2L2
0.860811



215
3832
ATM
0.861555



216
3774
CDC45L
0.862319



217
3342
CCNT1
0.86272



218
3430
STMN1
0.864508



219
3802
NOTCH3
0.865116



220
3309
CCND2
0.865741



221
3411
HIF1A
0.867769



222
3717
NTRK2
0.867864



223
3465
EPHA1
0.867876



224
3795
NR4A2
0.867991



225
3659
NM_015978
0.868205



226
3643
DDR2
0.868618



227
3392
BIRC5
0.869293



228
3786
FRAP1
0.870607



229
3297
CCT2
0.872024



230
2991
NPR1
0.872727



231
3318
CUL2
0.87438



232
3293
CDC25A
0.875



233
3421
ORC6L
0.875341



234
3454
FLT4
0.875663



235
2950
NEK6
0.876961



236
3815
MAPRE2
0.877732



237
3831
CLSPN
0.878064



238
3232
KDR
0.878378



239
3709
X95425
0.879358



240
2929
CHUK
0.881491



241
3378
NM_006009
0.882129



242
2952
PRKG1
0.883408



243
3776
NOTCH2
0.88366



244
3356
TUBG1
0.884709



245
3308
CDKN2B
0.885077



246
2967
NM_016653
0.886094



247
3591
RALB
0.889362



248
3635
STAT1
0.889881



249
3530
NOTCH1
0.890111



250
3750
CAMK2A
0.891525



251
3523
NOTCH2
0.893064



252
2980
RIPK1
0.894417



253
3249
MAP2K6
0.895216



254
3589
SOS1
0.895558



255
3587
VAV3
0.896552



256
2968
STK17B
0.899438



257
3505
STK6
0.899549



258
3526
HES6
0.899892



259
3261
ERBB2
0.904662



260
3252
NEK2
0.904873



261
3426
TK1
0.906569



262
3328
CCNC
0.909091



263
3470
RPS6KA1
0.909627



264
3798
ACTR2
0.910468



265
3595
FEN1
0.910569



266
3597
SHMT2
0.911368



267
3362
NR3C1
0.911404



268
3257
IGF1R
0.911442



269
3665
PAK4
0.913313



270
3678
PFTK1
0.913673



271
3344
CDKL1
0.913907



272
3302
CDK8
0.913988



273
3536
PIK3C3
0.916089



274
3685
LTK
0.917246



275
3749
NM_139021
0.917681



276
3268
CDC25C
0.919654



277
3743
RAGE
0.922602



278
3414
HDAC7A
0.925495



279
3162
MADH2
0.927277



280
3429
MCM6
0.928214



281
3682
DYRK1A
0.928261



282
3585
GAB1
0.928513



283
3549
PIK3R2
0.930054



284
3233
ROCK1
0.930818



285
3315
CCT4
0.931751



286
2990
NM_015112
0.933149



287
3409
TTK
0.934641



288
3237
CDC7
0.938429



289
2960
LYN
0.938849



290
3664
STK17A
0.93923



291
2931
MAP4K3
0.939649



292
3693
NEK9
0.939894



293
3694
STK38
0.941537



294
3000
BMX
0.94164



295
3445
BMPR1A
0.944154



296
3207
MAD1L1
0.945191



297
3714
NM_013355
0.947122



298
3652
PDGFRA
0.947533



299
3631
WISP2
0.948783



300
3799
ACTR3
0.949315



301
2912
MPHOSPH1
0.95053



302
3142
KIF25
0.950655



303
3755
CDKL3
0.950839



304
3231
ILK
0.951



305
3155
ATR
0.951613



306
3646
NM_005781
0.952566



307
2979
PAK2
0.953323



308
3296
CDKN2C
0.954784



309
2983
GUCY2C
0.956701



310
3497
EPHA8
0.958146



311
3163
THRA
0.959354



312
3471
MAPK10
0.960665



313
2940
DCAMKL1
0.963487



314
3593
VAV2
0.963865



315
3398
HDAC11
0.964784



316
3752
CCNB3
0.964824



317
3641
TYRO3
0.965291



318
3195
CDH1
0.96542



319
3552
PDK1
0.96695



320
3132
KIF23
0.970496



321
2951
AJ311798
0.971591



322
3214
CENPF
0.974453



323
3436
BRAF
0.97619



324
3104
CDK4
0.976337



325
2959
PIM2
0.977075



326
3228
CDC27
0.978723



327
3570
RALGDS
0.979927



328
3826
NM_015694
0.981405



329
3788
PRKCE
0.983254



330
2954
ROCK2
0.983541



331
3539
PLCG2
0.985447



332
3732
CSNK2A2
0.985667



333
3759
NM_006622
0.98881



334
2998
MAPK7
0.993015



335
3352
NR3C2
0.996058



336
3488
AURKB
0.996508



337
3130
FRAP1
0.996898



338
3691
NM_024046
0.997251



339
3683
NM_003138
0.998179



340
3569
RASGRP2
1.000543



341
2920
EIF2AK3
1.005703



342
3365
PRIM1
1.006112



343
3462
TGFBR2
1.00726



344
3513
ROS1
1.009016



345
3102
CKS2
1.013052



346
2945
CDC42BPB
1.01398



347
3656
PRKCN
1.016229



348
3726
MAPKAPK2
1.016458



349
3002
CRKL
1.01909



350
3670
MAP3K10
1.01919



351
3767
FZD3
1.019811



352
3645
CASK
1.020319



353
3707
TXK
1.022666



354
3455
MAP2K4
1.023218



355
3372
TUBA8
1.025814



356
3540
PPP2CB
1.027826



357
3690
PRKAA2
1.029902



358
3307
CDC6
1.03012



359
3495
FGFR2
1.032093



360
3485
DHX8
1.032492



361
3696
NM_016281
1.038149



362
3716
ULK1
1.039167



363
3265
RAF1
1.039442



364
3161
WT1
1.039655



365
3215
MAD2L1
1.039783



366
3415
HSPCA
1.040186



367
3127
MAPK1
1.040277



368
3686
MAP3K8
1.040303



369
3490
ERBB3
1.040323



370
3441
PRKCI
1.042373



371
3115
MDM2
1.04276



372
3264
CDK3
1.044285



373
3147
CDKN2A
1.045872



374
3568
PLD1
1.048696



375
3559
RAP1GDS1
1.05



376
2928
IRAK1
1.050577



377
3197
ARHB
1.052064



378
3785
GRB2
1.052525



379
3248
JAK2
1.053539



380
3199
NF2
1.053654



381
2992
PRKR
1.055468



382
3516
ARHA
1.058051



383
3449
TBK1
1.059537



384
2953
MAPK11
1.059656



385
3164
MYCL1
1.060646



386
3745
CAMKK2
1.061685



387
3324
CUL5
1.062571



388
3243
NM_004203
1.062998



389
3187
WNT7B
1.063935



390
3459
EGFR
1.066553



391
3239
CDK6
1.067257



392
3170
BLM
1.068402



393
2943
DYRK2
1.06862



394
3320
CCND3
1.070018



395
3369
NM_007027
1.071887



396
3624
CTNNB1
1.072588



397
3500
SGK
1.074011



398
3101
MAPK14
1.074871



399
3408
PIN1
1.075614



400
2924
STK25
1.076046



401
3548
RAC1
1.07851



402
3676
MAP4K1
1.079121



403
3698
ADRBK2
1.079569



404
3301
CCNB1
1.080243



405
2925
FYN
1.081081



406
3565
RAB2
1.081968



407
2977
RIPK3
1.082037



408
3810
AI278633
1.084388



409
3796
ARHGEF6
1.084848



410
3116
KIF5A
1.086755



411
3590
ARHGEF2
1.088083



412
3679
CLK2
1.088737



413
3119
CDKN1B
1.089067



414
3367
DHFR
1.092319



415
3797
ARHGEF9
1.092391



416
3405
HDAC8
1.096856



417
2957
TYK2
1.099156



418
3091
KIFC3
1.10008



419
3546
INPP5D
1.102828



420
3227
NUMA1
1.104478



421
3181
ST5
1.104782



422
3807
SPAG5
1.105317



423
3090
KIF3C
1.10597



424
3343
CENPJ
1.107383



425
3245
AXL
1.108766



426
3097
KIF20A
1.108842



427
3360
RRM2
1.109827



428
3349
IMPDH1
1.111043



429
3474
CSNK2A1
1.111842



430
3616
FZD1
1.11295



431
3620
AXIN2
1.113386



432
2995
PTK2
1.115385



433
3634
BTRC
1.117674



434
3504
PIK3CB
1.118194



435
3561
FOS
1.118649



436
3618
DVL2
1.12



437
3537
EIF4EBP1
1.121316



438
3550
PLCG1
1.121971



439
3443
EPS8
1.122744



440
3370
AR
1.123767



441
3543
PDK2
1.12548



442
3122
ATSV
1.127371



443
3167
S100A2
1.127907



444
3596
SHMT1
1.128114



445
3811
NM_152524
1.129555



446
3779
CTNNA1
1.129565



447
3312
CUL3
1.133047



448
2963
MAP3K11
1.133758



449
2942
TTN
1.133889



450
3790
ERBB3
1.135274



451
3094
KIF3A
1.13729



452
3545
IRS2
1.139283



453
3305
CDC2L5
1.140753



454
3748
NM_016507
1.140961



455
3614
CTNND2
1.141748



456
3437
FGFR1
1.143284



457
3389
NM_052963
1.145845



458
3213
NM_016238
1.145939



459
3533
HES7
1.148773



460
3321
CDKN3
1.152745



461
3711
LIMK1
1.153559



462
3503
EGR1
1.156344



463
3701
STK10
1.160598



464
3608
MAP3K7IP1
1.161191



465
3730
TESK1
1.162946



466
3156
MSH2
1.163507



467
3571
VAV1
1.164063



468
3668
DAPK3
1.165365



469
3677
HCK
1.166105



470
3708
RPS6KC1
1.166667



471
3110
KIF13A
1.167294



472
3185
VCAM1
1.170254



473
3837
PRKAA1
1.171443



474
3514
HRAS
1.171476



475
3371
NM_006087
1.175311



476
3420
NM_014109
1.176378



477
3669
NTRK3
1.17801



478
2939
TLK1
1.179137



479
3654
VRK2
1.180868



480
3636
STAT2
1.181562



481
3506
XM_095827
1.18376



482
3728
TIE
1.184901



483
3496
EIF4EBP2
1.188138



484
2994
MATK
1.188439



485
3353
PGR
1.188925



486
3771
PIK3CA
1.191131



487
3111
KIF5B
1.191167



488
3396
HDAC10
1.192015



489
3330
CDK9
1.194303



490
3705
NM_012119
1.195395



491
3339
CCNB2
1.195402



492
3005
MERTK
1.196303



493
3220
ANAPC11
1.1994



494
3507
NM_145754
1.199485



495
3418
CENPA
1.199564



496
3492
PRKCQ
1.199597



497
3499
GRB2
1.204124



498
3667
NM_016542
1.204923



499
3084
KIF14
1.207333



500
3317
CCNI
1.208734



501
3457
BAD
1.208929



502
3819
TACC3
1.209677



503
3377
NM_006088
1.210588



504
3472
MAP3K5
1.210677



505
2922
NM_004783
1.211356



506
3453
MAP2K2
1.212321



507
3724
EPHA7
1.213738



508
3260
STK11
1.214815



509
3675
ADRBK1
1.215503



510
3379
NM_032525
1.223512



511
2956
PRKCL2
1.223938



512
3666
PAK6
1.229403



513
3216
CDC20
1.231173



514
3672
SYK
1.231714



515
3555
RASA1
1.236402



516
3354
RRM1
1.237695



517
3153
RB1
1.237825



518
3253
PRKCA
1.239404



519
3146
KIF21A
1.240245



520
3756
CDK5RAP2
1.242775



521
3721
ANKRD3
1.245185



522
3224
FBXO5
1.24973



523
3607
TLE1
1.250329



524
2981
ALK
1.252514



525
2978
AB067470
1.252713



526
3440
FGFR3
1.253731



527
3578
RREB1
1.256567



528
3393
HDAC2
1.258824



529
3520
NRAS
1.263715



530
3190
WNT4
1.265328



531
3463
TEC
1.265973



532
3621
CTNNA2
1.26658



533
3425
DLG7
1.267399



534
3311
CDK10
1.269347



535
3567
SHC1
1.270057



536
3753
CDK5
1.276163



537
2989
ACVR1B
1.276215



538
3692
AB007941
1.27931



539
3244
PRKCZ
1.279368



540
3092
KIF12
1.279896



541
3487
MAP2K3
1.280835



542
3813
ANLN
1.282313



543
3198
ICAM1
1.285429



544
3697
CAMK2G
1.286036



545
3735
PRKACB
1.286694



546
3100
GSK3B
1.289078



547
3431
NM_018454
1.289806



548
3615
FZD2
1.292222



549
2947
NM_007064
1.29381



550
3340
CENPH
1.293935



551
3172
PLAU
1.297571



552
3160
TACSTD1
1.297585



553
3212
NM_022662
1.301215



554
3098
CENPE
1.305802



555
3626
DVL3
1.306682



556
3830
NM_013296
1.307494



557
3713
PRKG2
1.307933



558
3768
ARAF1
1.308011



559
3493
MAPK9
1.308449



560
3108
KIF22
1.308726



561
3169
NME1
1.310985



562
3125
NM_031217
1.311267



563
3375
AHCY
1.311852



564
3583
JUNB
1.31241



565
3458
ARAF1
1.315519



566
3612
TCF1
1.316285



567
3294
CCNF
1.317748



568
3338
CUL4A
1.318527



569
3649
CAMK2B
1.322337



570
3576
GRAP
1.322985



571
3527
DTX2
1.33023



572
3145
C20orf23
1.334687



573
3180
CD44
1.335574



574
3758
RAD51L1
1.335901



575
3165
FGF2
1.336082



576
3828
KIF20A
1.337004



577
3553
CKS1B
1.339383



578
3089
KIFC1
1.341566



579
3442
ERBB4
1.345118



580
3554
PIK3R3
1.347147



581
3613
DVL1
1.347505



582
2985
MKNK1
1.347934



583
3117
ATM
1.348967



584
3424
EZH2
1.352941



585
3695
PRKAA1
1.355145



586
3446
PRKCG
1.355556



587
3194
RARB
1.359932



588
3644
EPHB1
1.36061



589
3700
AB037782
1.361005



590
3599
DTYMK
1.361789



591
3729
RYK
1.361997



592
3114
PIK3CD
1.362808



593
3821
ASPM
1.363705



594
3373
TOP2A
1.363708



595
3563
RAB3A
1.365615



596
3764
NOTCH4
1.36911



597
3628
CTNNBL1
1.3702



598
3823
NM_017779
1.372126



599
3715
CDC42BPA
1.372256



600
3562
RASD1
1.372563



601
3784
MAPK8
1.376577



602
3574
SH3KBP1
1.384674



603
3594
RAP2A
1.393939



604
3662
LCK
1.3981



605
3787
FZD4
1.399749



606
3316
CCNA1
1.404295



607
3684
STK38L
1.406161



608
3610
LEF1
1.407463



609
3390
NM_080925
1.407563



610
3152
APC
1.414678



611
3149
TP53
1.420044



612
3238
MAP3K3
1.420428



613
3109
KIF1C
1.420608



614
3325
CDKN1C
1.42522



615
3314
CCNG1
1.426516



616
3825
NM_152562
1.428805



617
3588
RHEB
1.435039



618
3736
PTK7
1.440171



619
3118
NM_032559
1.440252



620
3521
MET
1.440418



621
3096
AKT1
1.440951



622
3361
IMPDH2
1.442308



623
3582
GRAP2
1.444349



624
3584
RASAL2
1.450119



625
3801
PSEN1
1.466292



626
3803
MPHOSPH1
1.470276



627
2938
ALS2CR7
1.471357



628
3106
KIF9
1.47493



629
3313
CCNG2
1.48267



630
3792
ARHGEF1
1.48329



631
3210
NM_013366
1.48366



632
3820
NM_018410
1.483709



633
2932
MAPKAPK3
1.488



634
3747
GSK3A
1.491773



635
2962
MAP4K2
1.495448



636
3699
NM_032844
1.502812



637
3189
MYB
1.504618



638
3629
AXIN1
1.505556



639
2941
DYRK3
1.505717



640
3818
AI338451
1.511194



641
2919
OSR1
1.512906



642
3140
XM_089006
1.518548



643
3229
PRKCL1
1.525203



644
3510
CDK4
1.529837



645
3319
CCND1
1.531034



646
3159
RET
1.536506



647
3242
PRKCB1
1.540024



648
3519
NTRK1
1.547773



649
3808
CKAP2
1.554545



650
2988
FRK
1.557214



651
2944
MARK1
1.557763



652
2971
DAPK2
1.55938



653
3299
CUL1
1.560841



654
3660
DMPK
1.5625



655
3515
PDGFRB
1.562977



656
3522
KRAS2
1.564353



657
3004
MAP3K1
1.570175



658
3395
HDAC5
1.571159



659
3468
ABL2
1.571225



660
3529
DTX1
1.57276



661
3329
CDC42
1.580386



662
3704
ACVR2B
1.58046



663
3827
NM_018123
1.581315



664
3456
FLT1
1.583826



665
3310
CDC34
1.585818



666
3331
CDC25B
1.585938



667
3368
TOP3A
1.58728



668
3126
KIF3B
1.588728



669
3780
MCM3
1.590296



670
3128
AKT2
1.592696



671
3598
PCNA
1.59319



672
3535
SKP2
1.593333



673
2955
PAK1
1.59552



674
3234
CDK2
1.596033



675
3138
KIF17
1.604846



676
3632
WISP1
1.607319



677
3611
CTNNAL1
1.611386



678
3300
CDC14B
1.611486



679
3511
XM_168069
1.614698



680
3144
KIF4B
1.619674



681
3627
CTNNA1
1.620915



682
3337
CDKN1A
1.626582



683
3202
MCC
1.627957



684
3143
NM_017596
1.628521



685
3186
ETS1
1.635593



686
3432
PRC1
1.637647



687
3556
RAP1A
1.638173



688
3335
CDK5R2
1.656172



689
2933
MAP4K5
1.656522



690
2927
MAPK13
1.659401



691
2973
NEK1
1.664311



692
3538
NFKB2
1.667808



693
3602
MCM3
1.678819



694
3603
POLS
1.678937



695
3630
WISP3
1.679045



696
3447
PRKCM
1.680152



697
3402
HDAC4
1.68123



698
3133
XM_168069
1.681935



699
3428
ECT2
1.690096



700
3720
AB002301
1.691718



701
3793
MAPRE1
1.693966



702
3681
SRPK1
1.700611



703
3817
NM_019013
1.702326



704
3136
XM_066649
1.708388



705
3355
GUK1
1.710938



706
3087
PTEN
1.716866



707
3579
PDZGEF2
1.717714



708
3168
DCC
1.719083



709
3151
MLH1
1.72077



710
3217
NM_014885
1.722045



711
3191
WNT2
1.728016



712
3765
CREBBP
1.72973



713
3655
CAMK2D
1.733773



714
3407
HDAC9
1.748784



715
3255
CDK7
1.75



716
3295
CDK2AP1
1.75



717
3192
WNT1
1.751208



718
3333
CCNT2
1.761104



719
3703
AK024504
1.764425



720
3760
CDKL5
1.769444



721
2948
MYO3A
1.769759



722
3800
CHFR
1.772809



723
3544
IRS1
1.776668



724
3235
CHEK1
1.776886



725
3137
KIF26A
1.782366



726
3673
DDR1
1.792507



727
3336
CDC37
1.807985



728
3725
EPHA4
1.820076



729
3404
PPARG
1.822581



730
3604
TK2
1.82846



731
3738
PRKWNK3
1.836245



732
3141
NM_145754
1.843889



733
3451
MAP3K4
1.855556



734
3417
HDAC3
1.857143



735
3508
KIF25
1.871592



736
3575
LATS2
1.879574



737
3761
WT1
1.88089



738
3723
NM_018401
1.88722



739
3719
BMPR2
1.890545



740
3204
CDC16
1.892826



741
3467
MAP2K7
1.894459



742
2986
ACVR2
1.896882



743
3218
CDC23
1.904255



744
3791
NM_005200
1.913043



745
3804
NM_024322
1.920139



746
3558
RALA
1.92029



747
3824
MAPRE3
1.940871



748
3622
FZD9
1.988166



749
3205
NM_139286
1.997054



750
3221
TOP3B
1.997534



751
3794
WASL
1.998403



752
3637
STAT4
2.005199



753
3834
CHEK1
2.01625



754
3400
BCL2
2.045028



755
3223
NM_016263
2.045139



756
3358
TOP2B
2.050562



757
3512
TGFBR1
2.062016



758
3259
MAPK8
2.064081



759
3742
RHOK
2.075949



760
2946
NM_017719
2.078131



761
3406
TERT
2.10274



762
3206
ANAPC5
2.159615



763
3531
NM_021170
2.163086



764
3008
SGK2
2.1766



765
3706
C20orf97
2.1875



766
3254
CSF1R
2.196822



767
3439
EGR2
2.213333



768
2970
AATK
2.235211



769
3528
TCF3
2.273649



770
3327
CDC45L
2.288265



771
3551
STAT3
2.29125



772
3001
PRKY
2.313131



773
3734
BMPR1B
2.330839



774
3095
KIF2C
2.336785



775
3222
PTTG1
2.347826



776
3532
NM_019089
2.352437



777
3547
FOXO1A
2.352444



778
3671
STK4
2.362408



779
3781
SRC
2.37859



780
3789
ELK1
2.394828



781
3247
NM_018492
2.480851



782
3586
RASA2
2.506796



783
3727
GPRK6
2.553987



784
3689
BLK
2.584588



785
3777
ABL1
2.615226



786
3399
HSPCB
2.632207



787
2958
PRKACA
2.635514



788
3304
CCNE2
2.677656



789
3617
CTNNBIP1
2.698292



790
3225
NM_013367
2.714286



791
3619
FRAT1
2.728111



792
3121
PIK3C2A
2.828125



793
3816
NM_017769
2.847273



794
3134
XM_170783
2.923286



795
3737
NM_016457
2.940451



796
3135
XM_064050
3.063002



797
3129
STK6
3.146434



798
3564
RALBP1
3.170605



799
3580
ELK1
3.356401



800
3157
NF1
3.402273



801
3638
STAT5A
3.754386



802
3241
WEE1
3.801887



803
3718
PTK6
4.317857



804
3712
RPS6KA6
5.356624



805
3158
BRCA1
5.821429



806
3642
EPHB3
6.43



807
3150
BRCA2
14.13136

















TABLE IIC










Average fold sensitization by doxorubicin















ave of 3



Gene ID
BioID
Gene
screens
















1
2514
PLK
0.094489



2
3099
PLK
0.195626



3
3099
PLK
0.211482



4
3099
PLK
0.211747



5
3099
PLK
0.219626



6
3099
PLK
0.227603



7
3099
PLK
0.235482



8
3099
PLK
0.235683



9
3099
PLK
0.235747



10
3099
PLK
0.251539



11
3099
PLK
0.251603



12
3099
PLK
0.259683



13
3099
PLK
0.275539



14
3099
PLK
0.282503



15
3099
PLK
0.298359



16
3099
PLK
0.298624



17
3099
PLK
0.31448



18
3099
PLK
0.32256



19
3534
PLK
0.330807



20
3099
PLK
0.338416



21
3099
PLK
0.395491



22
3099
PLK
0.411612



23
3006
PLK
0.415454



24
3099
PLK
0.419491



25
3099
PLK
0.435548



26
3099
PLK
0.435612



27
3433
PLK
0.435845



28
3391
PLK
0.440842



29
3099
PLK
0.459548



30
3099
PLK
0.482368



31
3099
PLK
0.498489



32
3322
CCNA2
0.512614



33
3099
PLK
0.522425



34
3805
C20orf1
0.562328



35
3423

0.613084



36
3600
RRM2B
0.659243



37
3305
CDC2L5
0.68014



38
3542
PPP2CA
0.695506



39
3266
PLK
0.696721



40
3228
CDC27
0.70157



41
3464
INSR
0.70706



42
3326
CCT7
0.724986



43
3740
STK35
0.754807



44
3731
CSNK1E
0.765738



45
3416
IKBKE
0.773235



46
3293
CDC25A
0.77957



47
3309
CCND2
0.791487



48
3350
ADA
0.800034



49
3812
CDCA8
0.815766



50
3354
RRM1
0.817751



51
3446
PRKCG
0.822809



52
3648
CLK1
0.824307



53
3509
KIF21A
0.826427



54
3526
HES6
0.826991



55
3250
CHEK2
0.828202



56
3262
LATS1
0.82944



57
3359
TUBA1
0.839308



58
3344
CDKL1
0.840425



59
2984
EPHB6
0.846685



60
3702
MAP3K13
0.84685



61
3838
PRKAA2
0.853115



62
3422
SMC4L1
0.854651



63
3332
CDC5L
0.85491



64
3750
CAMK2A
0.857171



65
3686
MAP3K8
0.8599



66
3226
RBX1
0.862335



67
3438
DTR
0.863218



68
3318
CUL2
0.863485



69
3454
FLT4
0.864511



70
3366
TYMS
0.866092



71
3444
NEK3
0.866318



72
3397
BUB3
0.867363



73
3007
MAP3K14
0.86906



74
3373
TOP2A
0.875387



75
2934
IRAK2
0.875671



76
3188
PMS2
0.876644



77
3461
KIT
0.876727



78
3398
HDAC11
0.878587



79
3665
PAK4
0.879213



80
3494
MAPK4
0.879947



81
3303
CDKN2D
0.88429



82
2925
FYN
0.885569



83
3437
FGFR1
0.889075



84
3219
CENPC1
0.889832



85
3491
PRKCE
0.891708



86
3105
BUB1
0.892262



87
3609
FZD3
0.89297



88
3421
ORC6L
0.893859



89
3414
HDAC7A
0.894925



90
3342
CCNT1
0.89645



91
3193
FGF3
0.897275



92
3203
ITGA5
0.89915



93
3679
CLK2
0.899792



94
3656
PRKCN
0.903305



95
3677
HCK
0.903727



96
3172
PLAU
0.904045



97
2999
FES
0.904351



98
3161
WT1
0.907863



99
3230
MAP2K1
0.908157



100
2937

0.910875



101
3502
PRKCH
0.913184



102
3317
CCNI
0.913695



103
3086
KIF11
0.914508



104
3412
KIF25
0.915671



105
3710
GPRK2L
0.917359



106
3585
GAB1
0.91762



107
3807
SPAG5
0.918025



108
3815
MAPRE2
0.919461



109
3646

0.920311



110
3000
BMX
0.920926



111
3365
PRIM1
0.922943



112
3574
SH3KBP1
0.924261



113
3485
DHX8
0.924589



114
3527
DTX2
0.92511



115
3378

0.927814



116
3799
ACTR3
0.929286



117
3822
GTSE1
0.929871



118
3100
GSK3B
0.932676



119
3206
ANAPC5
0.932816



120
3351
ESR1
0.932858



121
3623
CTNND1
0.932974



122
3601
POLE
0.935664



123
3097
KIF20A
0.939338



124
2991
NPR1
0.941392



125
2926

0.943073



126
3717
NTRK2
0.94323



127
3162
MADH2
0.953335



128
3783
KRAS2
0.954957



129
3660
DMPK
0.955308



130
3236
PTK2B
0.955874



131
3088
KIF13B
0.960206



132
3774
CDC45L
0.961565



133
3540
PPP2CB
0.96255



134
3251
ABL1
0.96267



135
3498
CSNK1A1
0.963185



136
3307
CDC6
0.963749



137
3830

0.96419



138
3374
POLR2A
0.964327



139
3413
KIF23
0.967774



140
3296
CDKN2C
0.967818



141
3132
KIF23
0.96794



142
3708
RPS6KC1
0.969675



143
3445
BMPR1A
0.970178



144
3694
STK38
0.970842



145
3566
JUN
0.971389



146
3140

0.97186



147
3571
VAV1
0.972374



148
2993
SRMS
0.972957



149
3268
CDC25C
0.973198



150
3835
CHEK2
0.973353



151
3557
JUND
0.973868



152
3195
CDH1
0.973895



153
3375
AHCY
0.974215



154
3163
THRA
0.976052



155
3164
MYCL1
0.979364



156
3798
ACTR2
0.980521



157
3392
BIRC5
0.980792



158
3196
ARHI
0.980973



159
3536
PIK3C3
0.981403



160
2950
NEK6
0.981709



161
3773
WNT2
0.982648



162
3776
NOTCH2
0.983584



163
3814
HMMR
0.983597



164
3234
CDK2
0.983724



165
2982
CDC2L2
0.984121



166
3826

0.985249



167
2953
MAPK11
0.987788



168
3403
AURKC
0.988679



169
3586
RASA2
0.989648



170
3503
EGR1
0.991443



171
3166
PMS1
0.99314



172
3394
HDAC6
0.994139



173
3652
PDGFRA
0.994658



174
3625
CTBP2
0.994928



175
3294
CCNF
0.995133



176
3260
STK11
0.998488



177
2968
STK17B
0.998826



178
3703

0.999818



179
3577
RAB2L
1.00021



180
3184
TSG101
1.00109



181
2927
MAPK13
1.001159



182
3116
KIF5A
1.002239



183
3496
EIF4EBP2
1.005451



184
3741
RPS6KB2
1.00589



185
3298
CCNE1
1.005922



186
2990

1.006496



187
3142
KIF25
1.006522



188
3218
CDC23
1.009586



189
3517
MYC
1.010689



190
2997
MST1R
1.011122



191
3003
FER
1.012506



192
3700

1.013542



193
3470
RPS6KA1
1.013802



194
3439
EGR2
1.013847



195
3429
MCM6
1.014653



196
3372
TUBA8
1.017048



197
3556
RAP1A
1.017133



198
3155
ATR
1.017435



199
3649
CAMK2B
1.017461



200
3501
RPS6KA2
1.018616



201
3336
CDC37
1.019161



202
2928
IRAK1
1.021732



203
3733
MYLK2
1.021742



204
2960
LYN
1.022112



205
3301
CCNB1
1.022891



206
3743
RAGE
1.023372



207
3525
NOTCH4
1.02341



208
3767
FZD3
1.023646



209
2954
ROCK2
1.02397



210
3475
EPHB4
1.024709



211
3635
STAT1
1.026128



212
3746
HUNK
1.026176



213
2977
RIPK3
1.0272



214
3573

1.028343



215
3751
BCR
1.028418



216
3112
KNSL7
1.029109



217
3488
AURKB
1.029885



218
3356
TUBG1
1.029908



219
3364
HPRT1
1.030247



220
3465
EPHA1
1.032043



221
3828
KIF20A
1.032108



222
3434
DDX6
1.03425



223
3143

1.03439



224
3212

1.034473



225
3725
EPHA4
1.034871



226
3473
DAPK1
1.035466



227
3581
RASGRP1
1.036407



228
3357
PRIM2A
1.036773



229
3469
AAK1
1.037538



230
3171
VHL
1.038422



231
3123
AKT3
1.039278



232
3572
RASA3
1.04084



233
3615
FZD2
1.042378



234
3658
STK18
1.043083



235
3261
ERBB2
1.044345



236
3220
ANAPC11
1.0449



237
3639
STAT6
1.045395



238
2959
PIM2
1.048207



239
2935
MAPK6
1.050943



240
3752
CCNB3
1.051148



241
3431

1.05315



242
3101
MAPK14
1.054104



243
3462
TGFBR2
1.056272



244
3319
CCND1
1.057299



245
3592
SOS2
1.058842



246
3655
CAMK2D
1.061571



247
3513
ROS1
1.062804



248
3297
CCT2
1.064889



249
3549
PIK3R2
1.066314



250
2998
MAPK7
1.066798



251
3334
CUL4B
1.066807



252
3381
POLR2B
1.068615



253
3633
CTBP1
1.069269



254
3678
PFTK1
1.07042



255
2987
RNASEL
1.072118



256
3256
CDC2L1
1.073967



257
3558
RALA
1.074961



258
3749

1.075156



259
3252
NEK2
1.075822



260
2919
OSR1
1.077885



261
3393
HDAC2
1.077906



262
3747
GSK3A
1.078401



263
3410
RPS27
1.078517



264
3107
KIF2
1.078686



265
3654
VRK2
1.081195



266
3533
HES7
1.081287



267
2983
GUCY2C
1.083605



268
3555
RASA1
1.084083



269
3258
MOS
1.084874



270
3180
CD44
1.085294



271
3124
KIF4A
1.086165



272
3179
PDGFB
1.086599



273
3209
MAD2L2
1.088835



274
3295
CDK2AP1
1.089703



275
3726
MAPKAPK2
1.09032



276
3674
CSNK1D
1.090974



277
3616
FZD1
1.091935



278
3452
JAK1
1.092015



279
3823

1.092187



280
3745
CAMKK2
1.092307



281
3149
TP53
1.092755



282
3561
FOS
1.092859



283
3836
TP53
1.093641



284
3170
BLM
1.094952



285
2930
ITK
1.095322



286
3744
PRKWNK4
1.095854



287
3401
HDAC1
1.096531



288
3300
CDC14B
1.096651



289
3348
DCK
1.096689



290
3405
HDAC8
1.096956



291
3239
CDK6
1.097696



292
3640
STAT5B
1.098035



293
2992
PRKR
1.098133



294
3548
RAC1
1.09835



295
3306
CDC14A
1.098874



296
2943
DYRK2
1.099617



297
3127
MAPK1
1.102044



298
3716
ULK1
1.104258



299
2922

1.106102



300
3160
TACSTD1
1.107397



301
2964
RIPK2
1.109482



302
3634
BTRC
1.110007



303
3576
GRAP
1.110227



304
3833
ATR
1.110614



305
3837
PRKAA1
1.111073



306
2939
TLK1
1.111429



307
3125

1.11143



308
3299
CUL1
1.111864



309
3813
ANLN
1.112297



310
3756
CDK5RAP2
1.112508



311
2976
NEK7
1.112602



312
2965

1.11309



313
3784
MAPK8
1.114132



314
3653
NPR2
1.115282



315
3302
CDK8
1.115429



316
3628
CTNNBL1
1.115905



317
3664
STK17A
1.115938



318
3504
PIK3CB
1.117357



319
3395
HDAC5
1.118952



320
3369

1.119457



321
3243

1.119572



322
3715
CDC42BPA
1.119975



323
2924
STK25
1.122952



324
3568
PLD1
1.123878



325
3676
MAP4K1
1.124218



326
3343
CENPJ
1.127564



327
3238
MAP3K3
1.127647



328
3424
EZH2
1.127778



329
3418
CENPA
1.128399



330
3829
KIF2C
1.128457



331
3476
PRKCD
1.128572



332
3407
HDAC9
1.129023



333
2951

1.13065



334
3685
LTK
1.130723



335
2942
TTN
1.131132



336
3085
KIF5C
1.133235



337
3367
DHFR
1.133721



338
3362
NR3C1
1.134725



339
3400
BCL2
1.134785



340
3800
CHFR
1.134967



341
3103
PIK3CA
1.135082



342
3711
LIMK1
1.135687



343
3165
FGF2
1.136323



344
3213

1.137328



345
3370
AR
1.137843



346
3772
RPS6KB1
1.138023



347
3189
MYB
1.138695



348
3631
WISP2
1.138989



349
2945
CDC42BPB
1.140434



350
3593
VAV2
1.141048



351
3338
CUL4A
1.141509



352
3092
KIF12
1.14183



353
3782
SOS1
1.14272



354
2989
ACVR1B
1.143948



355
3808
CKAP2
1.144074



356
3310
CDC34
1.14429



357
3760
CDKL5
1.144621



358
3159
RET
1.144761



359
3508
KIF25
1.144865



360
3788
PRKCE
1.145993



361
3231
ILK
1.146387



362
3471
MAPK10
1.146497



363
3668
DAPK3
1.14781



364
3595
FEN1
1.14853



365
3775
NOTCH1
1.150372



366
3145
C20orf23
1.151785



367
3570
RALGDS
1.152146



368
2972
KSR
1.152379



369
3441
PRKCI
1.152901



370
3737

1.153373



371
3463
TEC
1.154814



372
3748

1.155977



373
3816

1.156751



374
3582
GRAP2
1.158058



375
3360
RRM2
1.158514



376
3516
ARHA
1.15962



377
3312
CUL3
1.160258



378
3005
MERTK
1.160604



379
3456
FLT1
1.160651



380
3567
SHC1
1.161312



381
3647
YES1
1.161861



382
3447
PRKCM
1.162427



383
3739

1.163543



384
3181
ST5
1.163581



385
3466
JAK3
1.164099



386
3311
CDK10
1.1651



387
3486
RPS6KA3
1.165517



388
3779
CTNNA1
1.165697



389
3148
LIG1
1.166358



390
3683

1.167226



391
3544
IRS1
1.167527



392
3335
CDK5R2
1.167989



393
3821
ASPM
1.167998



394
3108
KIF22
1.168525



395
3168
DCC
1.170395



396
3182
MYCN
1.172038



397
3119
CDKN1B
1.172505



398
3692

1.173629



399
3687
CAMK4
1.17436



400
3420

1.175153



401
3762
MCC
1.175576



402
3519
NTRK1
1.175989



403
3257
IGF1R
1.176551



404
3769
PLAU
1.176774



405
3339
CCNB2
1.177549



406
3682
DYRK1A
1.178203



407
3240
MAPK12
1.178713



408
3156
MSH2
1.17907



409
2936
SGKL
1.17989



410
2920
EIF2AK3
1.179969



411
3670
MAP3K10
1.180357



412
3207
MAD1L1
1.181963



413
3630
WISP3
1.182009



414
3153
RB1
1.183084



415
3632
WISP1
1.183165



416
3824
MAPRE3
1.183387



417
3624
CTNNB1
1.18419



418
3151
MLH1
1.185254



419
3495
FGFR2
1.185537



420
3349
IMPDH1
1.185827



421
2932
MAPKAPK3
1.186058



422
3130
FRAP1
1.186158



423
3714

1.188036



424
3467
MAP2K7
1.188179



425
3727
GPRK6
1.188457



426
3500
SGK
1.189014



427
3638
STAT5A
1.189492



428
3242
PRKCB1
1.191673



429
3588
RHEB
1.194214



430
2940
DCAMKL1
1.194443



431
3222
PTTG1
1.194583



432
3411
HIF1A
1.194933



433
2952
PRKG1
1.197336



434
3539
PLCG2
1.198326



435
3797
ARHGEF9
1.20036



436
2969

1.201116



437
3194
RARB
1.201145



438
3490
ERBB3
1.202371



439
3197
ARHB
1.2033



440
3347
TOP1
1.203483



441
2966

1.203678



442
3089
KIFC1
1.204418



443
3232
KDR
1.205127



444
3090
KIF3C
1.205488



445
3599
DTYMK
1.205645



446
3139

1.206674



447
3695
PRKAA1
1.20878



448
3425
DLG7
1.209098



449
3535
SKP2
1.20949



450
3327
CDC45L
1.209854



451
3651
VRK1
1.210029



452
3569
RASGRP2
1.210373



453
3246
RPS6KB1
1.210471



454
3131
KIF1B
1.21146



455
3671
STK4
1.212033



456
3757
CLK4
1.212447



457
2985
MKNK1
1.212627



458
2988
FRK
1.213049



459
3432
PRC1
1.2136



460
3699

1.214212



461
3008
SGK2
1.21451



462
2996
MAPK3
1.217258



463
3399
HSPCB
1.217278



464
3610
LEF1
1.219128



465
2980
RIPK1
1.220712



466
3675
ADRBK1
1.22227



467
3663
ALS2CR2
1.223782



468
3468
ABL2
1.223785



469
2961
PIM1
1.223874



470
3804

1.224331



471
3594
RAP2A
1.226644



472
3377

1.227759



473
3341
APLP2
1.229895



474
3524
NOTCH3
1.230078



475
3253
PRKCA
1.233866



476
3518
NFKB1
1.23456



477
3328
CCNC
1.236473



478
3563
RAB3A
1.237081



479
3765
CREBBP
1.23722



480
2979
PAK2
1.237809



481
3235
CHEK1
1.239472



482
3146
KIF21A
1.239694



483
3340
CENPH
1.239979



484
3215
MAD2L1
1.24524



485
3379

1.245359



486
3662
LCK
1.245594



487
3754
CDKL2
1.247567



488
3187
WNT7B
1.247786



489
3552
PDK1
1.248615



490
3618
DVL2
1.249105



491
3602
MCM3
1.249136



492
3564
RALBP1
1.249919



493
3404
PPARG
1.252208



494
3248
JAK2
1.252557



495
3147
CDKN2A
1.252718



496
3358
TOP2B
1.253573



497
3459
EGFR
1.255853



498
3249
MAP2K6
1.256087



499
3254
CSF1R
1.258457



500
2949
MYO3B
1.259934



501
3157
NF1
1.260606



502
3680
CLK3
1.262403



503
3113
CNK
1.262742



504
3825

1.263089



505
3667

1.26407



506
3753
CDK5
1.264077



507
3553
CKS1B
1.265301



508
2933
MAP4K5
1.265655



509
3796
ARHGEF6
1.265751



510
3419
RFC4
1.266922



511
3460
CSK
1.266969



512
3094
KIF3A
1.26728



513
3736
PTK7
1.267303



514
3707
TXK
1.268516



515
3791

1.26868



516
3523
NOTCH2
1.26965



517
3755
CDKL3
1.271644



518
3204
CDC16
1.271646



519
3353
PGR
1.271733



520
3115
MDM2
1.274517



521
3126
KIF3B
1.274522



522
3095
KIF2C
1.274859



523
2947

1.277515



524
3408
PIN1
1.278984



525
3657
PCTK1
1.279578



526
3211
BUB1B
1.282741



527
3643
DDR2
1.28316



528
3449
TBK1
1.285291



529
3669
NTRK3
1.285519



530
3200
REL
1.285524



531
3729
RYK
1.291213



532
3691

1.291243



533
3214
CENPF
1.291507



534
3801
PSEN1
1.291634



535
2978

1.293122



536
3141

1.294102



537
3792
ARHGEF1
1.294579



538
3477
FGFR4
1.29633



539
3169
NME1
1.298008



540
3693
NEK9
1.299989



541
3583
JUNB
1.300395



542
3768
ARAF1
1.302137



543
2975
NEK4
1.302558



544
3221
TOP3B
1.30285



545
3478
EPHA2
1.30329



546
3666
PAK6
1.303653



547
2963
MAP3K11
1.306508



548
3199
NF2
1.307337



549
3724
EPHA7
1.308035



550
3457
BAD
1.308937



551
3185
VCAM1
1.309542



552
3244
PRKCZ
1.310965



553
3587
VAV3
1.31294



554
3712
RPS6KA6
1.313627



555
3216
CDC20
1.315701



556
3551
STAT3
1.316414



557
3590
ARHGEF2
1.316547



558
3659

1.317441



559
3831
CLSPN
1.318145



560
3109
KIF1C
1.318847



561
3455
MAP2K4
1.319428



562
3118

1.319892



563
3709

1.320996



564
3122
ATSV
1.321439



565
3809
CDCA3
1.329173



566
3237
CDC7
1.330145



567
3650

1.335065



568
3382
TUBB
1.336093



569
3190
WNT4
1.336703



570
3591
RALB
1.338466



571
3091
KIFC3
1.339318



572
3761
WT1
1.340453



573
3832
ATM
1.343275



574
3154
MADH4
1.343448



575
3002
CRKL
1.345404



576
2946

1.346714



577
3389

1.347114



578
3645
CASK
1.34749



579
3315
CCT4
1.348613



580
3150
BRCA2
1.34964



581
3474
CSNK2A1
1.350654



582
3458
ARAF1
1.351534



583
3528
TCF3
1.354214



584
3529
DTX1
1.357117



585
3559
RAP1GDS1
1.359432



586
3721
ANKRD3
1.361311



587
3819
TACC3
1.367136



588
3578
RREB1
1.367845



589
3245
AXL
1.367989



590
3543
PDK2
1.368231



591
3352
NR3C2
1.368397



592
3493
MAPK9
1.368899



593
2958
PRKACA
1.371294



594
3435
FLT3
1.371521



595
3316
CCNA1
1.37217



596
3263
CDC2
1.373177



597
3224
FBXO5
1.374389



598
2701

1.378002



599
3497
EPHA8
1.378119



600
3255
CDK7
1.379045



601
3766
S100A2
1.379101



602
3690
PRKAA2
1.383537



603
3152
APC
1.384859



604
3201
RARA
1.387549



605
3396
HDAC10
1.391302



606
3363
GART
1.392568



607
2957
TYK2
1.392639



608
3323
CDK5R1
1.394769



609
3380
TUBG2
1.401159



610
3233
ROCK1
1.404806



611
3806
C10orf3
1.405976



612
3614
CTNND2
1.409777



613
3093
PIK3CG
1.41077



614
3763

1.412316



615
3487
MAP2K3
1.412348



616
3732
CSNK2A2
1.414035



617
3110
KIF13A
1.414042



618
3789
ELK1
1.414448



619
3786
FRAP1
1.416676



620
3554
PIK3R3
1.418107



621
3167
S100A2
1.418532



622
3084
KIF14
1.41956



623
3661
FGR
1.423887



624
3617
CTNNBIP1
1.425161



625
2974
NEK11
1.426945



626
3330
CDK9
1.428872



627
3227
NUMA1
1.432118



628
3734
BMPR1B
1.437299



629
3138
KIF17
1.441566



630
3186
ETS1
1.442612



631
3673
DDR1
1.444582



632
3450
MAP3K2
1.446117



633
3133

1.450561



634
3598
PCNA
1.451899



635
3106
KIF9
1.45407



636
3608
MAP3K7IP1
1.455728



637
3376
AGA
1.457466



638
3443
EPS8
1.460769



639
3102
CKS2
1.464872



640
3409
TTK
1.465445



641
3346
CCNK
1.465948



642
3604
TK2
1.466617



643
2921
RPS6KA5
1.467418



644
3597
SHMT2
1.468236



645
3499
GRB2
1.469395



646
3406
TERT
1.482496



647
3158
BRCA1
1.485158



648
3114
PIK3CD
1.485825



649
3575
LATS2
1.487058



650
3390

1.487135



651
3596
SHMT1
1.487573



652
3514
HRAS
1.488051



653
3730
TESK1
1.489848



654
3620
AXIN2
1.491167



655
3619
FRAT1
1.491691



656
3644
EPHB1
1.492026



657
3117
ATM
1.498897



658
3541
PKD2
1.5005



659
3607
TLE1
1.501123



660
3229
PRKCL1
1.502059



661
3104
CDK4
1.502301



662
3684
STK38L
1.5024



663
3626
DVL3
1.504253



664
2986
ACVR2
1.510627



665
2971
DAPK2
1.516585



666
3759

1.517994



667
3636
STAT2
1.519082



668
3611
CTNNAL1
1.523973



669
3794
WASL
1.529001



670
2944
MARK1
1.53037



671
3713
PRKG2
1.535337



672
3087
PTEN
1.540121



673
3506

1.54045



674
3191
WNT2
1.553178



675
3202
MCC
1.554866



676
3210

1.555868



677
3738
PRKWNK3
1.559877



678
3096
AKT1
1.560781



679
3308
CDKN2B
1.566367



680
3606
CREBBP
1.570055



681
3641
TYRO3
1.574144



682
3758
RAD51L1
1.575115



683
3192
WNT1
1.579448



684
3705

1.591723



685
3565
RAB2
1.597556



686
3770
TGFBR2
1.602887



687
3771
PIK3CA
1.605624



688
3314
CCNG1
1.606354



689
3579
PDZGEF2
1.609017



690
3603
POLS
1.609696



691
3589
SOS1
1.610658



692
2938
ALS2CR7
1.613751



693
3621
CTNNA2
1.62379



694
3265
RAF1
1.625904



695
3698
ADRBK2
1.626836



696
3622
FZD9
1.630823



697
3111
KIF5B
1.633474



698
3688
GUCY2D
1.63373



699
3489
SRC
1.633931



700
3320
CCND3
1.636023



701
2970
AATK
1.636349



702
3562
RASD1
1.636677



703
3728
TIE
1.637314



704
3827

1.638302



705
3778
VHL
1.639913



706
3448

1.6515



707
3426
TK1
1.654609



708
2701

1.655539



709
3331
CDC25B
1.661891



710
3371

1.664564



711
2923
ERN1
1.665407



712
3550
PLCG1
1.667241



713
3803
MPHOSPH1
1.668632



714
3333
CCNT2
1.669848



715
3520
NRAS
1.670763



716
3121
PIK3C2A
1.675061



717
3264
CDK3
1.681459



718
3785
GRB2
1.681539



719
3205

1.682938



720
3811

1.685119



721
3507

1.688535



722
2955
PAK1
1.688691



723
3440
FGFR3
1.695183



724
2994
MATK
1.696094



725
2967

1.703715



726
3325
CDKN1C
1.703926



727
3545
IRS2
1.705996



728
3492
PRKCQ
1.706638



729
3547
FOXO1A
1.710389



730
3530
NOTCH1
1.711344



731
3810

1.723959



732
3321
CDKN3
1.724044



733
3453
MAP2K2
1.737418



734
3793
MAPRE1
1.738248



735
2941
DYRK3
1.741034



736
3217

1.745349



737
3451
MAP3K4
1.753145



738
3442
ERBB4
1.760041



739
3696

1.760172



740
3701
STK10
1.767448



741
3817

1.768117



742
2912
MPHOSPH1
1.771582



743
3001
PRKY
1.772697



744
3128
AKT2
1.773864



745
2981
ALK
1.781796



746
3337
CDKN1A
1.781903



747
3802
NOTCH3
1.787122



748
3735
PRKACB
1.790032



749
3183

1.793085



750
3430
STMN1
1.798292



751
3531

1.800094



752
3515
PDGFRB
1.80459



753
3324
CUL5
1.820969



754
3511

1.832738



755
3472
MAP3K5
1.834487



756
3428
ECT2
1.84097



757
3642
EPHB3
1.84828



758
3208
ZW10
1.858453



759
3820

1.861643



760
3225

1.868827



761
3834
CHEK1
1.869085



762
3510
CDK4
1.869212



763
3795
NR4A2
1.870845



764
3247

1.875796



765
3521
MET
1.887521



766
3538
NFKB2
1.892227



767
3818

1.900799



768
3719
BMPR2
1.919267



769
3144
KIF4B
1.924148



770
3355
GUK1
1.925235



771
2956
PRKCL2
1.929173



772
3198
ICAM1
1.937953



773
3361
IMPDH2
1.938577



774
3672
SYK
1.945812



775
3697
CAMK2G
1.946161



776
3415
HSPCA
1.94686



777
3505
STK6
1.949702



778
3368
TOP3A
1.959095



779
3681
SRPK1
1.963919



780
3613
DVL1
1.984151



781
3720

1.996699



782
2995
PTK2
2.015836



783
3522
KRAS2
2.026984



784
3436
BRAF
2.036457



785
3787
FZD4
2.049799



786
3584
RASAL2
2.089191



787
3098
CENPE
2.090255



788
3267
CCNH
2.096356



789
2931
MAP4K3
2.11675



790
2962
MAP4K2
2.12521



791
3790
ERBB3
2.13688



792
3742
RHOK
2.142917



793
2948
MYO3A
2.173575



794
3629
AXIN1
2.184253



795
3546
INPP5D
2.197591



796
3723

2.212338



797
2973
NEK1
2.222767



798
3512
TGFBR1
2.223853



799
3135

2.223901



800
3637
STAT4
2.227212



801
3004
MAP3K1
2.235803



802
3304
CCNE2
2.239326



803
3129
STK6
2.248154



804
3402
HDAC4
2.253527



805
3627
CTNNA1
2.28197



806
3537
EIF4EBP1
2.322458



807
3704
ACVR2B
2.322634



808
3329
CDC42
2.333632



809
3259
MAPK8
2.334959



810
3689
BLK
2.340679



811
3241
WEE1
2.35419



812
3137
KIF26A
2.359341



813
3612
TCF1
2.413867



814
3532

2.468626



815
3764
NOTCH4
2.482525



816
3417
HDAC3
2.485246



817
3120
PIK3CB
2.528659



818
3313
CCNG2
2.568855



819
3722
TLK2
2.571781



820
3136

2.916125



821
3780
MCM3
2.988111



822
3580
ELK1
3.0307



823
3718
PTK6
3.090027



824
3777
ABL1
3.099871



825
3605
FZD4
3.155698



826
3134

3.263194



827
2929
CHUK
3.298485



828
3781
SRC
3.433423



829
3223

3.587036



830
3706
C20orf97
4.288466


















TABLE IIIA










siRNA sequences used in screens of DNA damaging



agents: cisplatin screen












SEQUENCE





GENE NAME
ID
SENSE SEQ
SEQ ID NO














CHUK
NM_001278
AAAGGCUGCUCACAAGUUCTT
50






CHUK
NM_001278
AGCUGCUCAACAAACCAGATT
51





CHUK
NM_001278
AUGAGGAACAGGGCAAUAGTT
52





PRKACA
NM_002730
GAAUGGGGUCAACGAUAUCTT
53





PRKACA
NM_002730
GGACGAGACUUCCUCUUGATT
54





PRKACA
NM_002730
GUGUGGCAAGGAGUUUUCUTT
55





MAP4K2
NM_004579
GAAUCCUAAGAAGAGGCCGTT
56





MAP4K2
NM_004579
GAGGAGGUCUUUCAUUGGGTT
57





MAP4K2
NM_004579
GAUAGUCAAGCUAGACCCATT
58





STK17B
NM_004226
AUCCUCCUGUAAUGGAACCTT
59





STK17B
NM_004226
GAAGAGGACAGGAUUGUCGTT
60





STK17B
NM_004226
GACCAACAGCAGAGAUAUGTT
61





ALK
NM_004304
ACCAGAGACCAAAUGUCACTT
62





ALK
NM_004304
AUAAGCCCACCAGCUUGUGTT
63





ALK
NM_004304
UCAACACCGCUUUGCCGAUTT
64





FRK
NM_002031
ACUAUAGACUUCCGCAACCTT
65





FRK
NM_002031
CAGUAGAUUGCUGUGGCCUTT
66





FRK
NM_002031
CUCCAUACAGCUUCUGAAGTT
67





MAP3K1
AF042838
UCACUUAGCAGCUGAGUCUTT
68





MAP3K1
AF042838
UUGACAGCACUGGUCAGAGTT
69





MAP3K1
AF042838
UUGGCAAGAACUUCUUGGCTT
70





KIF2C
NM_006845
ACAAAAACGGAGAUCCGUCTT
71





KIF2C
NM_006845
AUAAGCAGCAAGAAACGGCTT
72





KIF2C
NM_006845
GAAUUUCGGGCUACUUUGGTT
73





CENPE
NM_001813
GAAAAUGAAGCUUUGCGGGTT
74





CENPE
NM_001813
GAAGAGAUCCCAGUGCUUCTT
75





CENPE
NM_001813
UCUGAAAGUGACCAGCUCATT
76





STK6
NM 003600
ACAGUCUUAGGAAUCGUGCTT
3





STK6
NM_003600
GCACAAAAGCUUGUCUCCATT
1





STK6
NM_003600
UUGCAGAUUUUGGGUGGUCTT
2





KIF4B
AF241316
CCUGCAGCAACUGAUUACCTT
77





KIF4B
AF241316
GAACUUGAGAAGAUGCGAGTT
78





KIF4B
AF241316
GAAGAGGCCCACUGAAGUUTT
79





BRCA2
NM_000059
CAAAUGGGCAGGACUCUUATT
80





BRCA2
NM_000059
CUGUUCAGCCCAGUUUGAATT
81





BRCA2
NM_000059
UCUCCAAGGAAGUUGUACCTT
82





APC
NM_000038
ACCAAGUAUCCGCAAAAGGTT
83





APC
NM_000038
AGACCUGUAUUAGUACGCCTT
84





APC
NM_000038
CAAGCUUUACCCAGCCUGUTT
85





ATR
NM_001184
GAAACUGCAGCUAUCUUCCTT
86





ATR
NM_001184
GUUACAAUGAGGCUGAUGCTT
87





ATR
NM_001184
UCACGACUCGCUGAACUGUTT
88





BRCA1
NM_007296
ACUUAGGUGAAGCAGCAUCTT
89





BRCA1
NM_007296
GGGCAGUGAAGACUUGAUUTT
90





BRCA1
NM_007296
UGAAGUGGGCUCCAGUAUUTT
91





DCC
NM_005215
ACAUCGUGGUGCGAGGUUATT
92





DCC
NM_005215
AUGAGCCGCCAAUUGGACATT
93





DCC
NM_005215
AUGGCAAGUUUGGAAGGACTT
94





WNT1
NM_005430
ACGGCGUUUAUCUUCGCUATT
95





WNT1
NM_005430
CCCUCUUGCCAUCCUGAUGTT
96





WNT1
NM_005430
CUAUUUAUUGUGCUGGGUCTT
97





CHEK1
NM_001274
AUCGAUUCUGCUCCUCUAGTT
98





CHEK1
NM_001274
CUGAAGAAGCAGUCGCAGUTT
99





CHEK1
NM_001274
UGCCUGAAAGAGACUUGUGTT
100





WEE1
NM_003390
AUCGGCUCUGGAGAAUUUGTT
101





WEE1
NM_003390
CAAGGAUCUCCAGUCCACATT
102





WEE1
NM_003390
UGUACCUGUGUGUCCAUCUTT
103






NM_018492
AGGACACUUUGGGUACCAGTT
104






NM_018492
GACCCUAAAGAUCGUCCUUTT
105






NM_018492
GCUGAGGAGAAUAUGCCUCTT
106





MAPK8
NM_139049
CACCCGUACAUCAAUGUCUTT
107





MAPK8
NM_139049
GGAAUAGUAUGCGCAGCUUTT
108





MAPK8
NM_139049
GUGAUUCAGAUGGAGCUAGTT
109





CUL1
NM_003592
GACCGCAAACUACUGAUUCTT
110





CUL1
NM_003592
GCCAGCAUGAUCUCCAAGUTT
111





CUL1
NM_003592
UAGACAUUGGGUUCGCCGUTT
112





CCNG2
NM_004354
CCUCGAGAAAAAGGGCUGATT
113





CCNG2
NM_004354
GCUCAGCUGAAAGCUUGCATT
114





CCNG2
NM_004354
UGCCUAGCCGAGUAUUCUUTT
115





CDC42
NM_044472
ACCUUAUGGAAAAGGGGUGTT
116





CDC42
NM_044472
CCAUCCUGUUUGAAAGCCUTT
117





CDC42
NM_044472
CCCAAAAGGAAGUGCUGUATT
118





CDC25B
NM_021874
AGGAUGAUGAUGCAGUUCCTT
119





CDC25B
NM_021874
GACAAGGAGAAUGUGCGCUTT
120





CDC25B
NM_021874
GAGCCCAGUCUGUUGAGUUTT
121





TOP2B
NM_001068
ACAUUCCCUGGAGUGUACATT
122





TOP2B
NM_001068
GAGGAUUUAGCGGCAUUUGTT
123





TOP2B
NM_001068
GCUGCUGGACUGCAUAAAGTT
124





IMPDH2
NM_000884
AGAGGGAAGACUUGGUGGUTT
125





IMPDH2
NM_000884
CACUCAUGCCAGGACAUUGTT
126





IMPDH2
NM_000884
GAAGAAUCGGGACUACCCATT
127






NM_007027
ACUCACAGAAAAACCGUCGTT
128






NM_007027
AUGAUGGGCGGACGAGUAUTT
129






NM_007027
GAGUCAGCACCAUCAAAUGTT
130





HDAC4
NM_006037
AGAGGACGUUUUCUACGGCTT
131





HDAC4
NM_006037
AUCUGUUUGCAAGGGGAAGTT
132





HDAC4
NM_006037
CAAGAUCAUCCCCAAGCCATT
133





TERT
NM_003219
CACCAAGAAGUUCAUCUCCTT
134





TERT
NM_003219
GAGUGUCUGGAGCAAGUUGTT
135





TERT
NM_003219
GUUUGGAAGAACCCCACAUTT
136





BRAF
NM_004333
ACACUUGGUAGACGGGACUTT
137





BRAF
NM_004333
GUCAAUCAUCCACAGAGACTT
138





BRAF
NM_004333
UUGCAUGUGGAAGUGUUGGTT
139





ERBB4
NM_005235
GAGUACUCUAUAGUGGCCUTT
140





ERBB4
NM_005235
GCUUCCCAGUCCAAAUGACTT
141





ERBB4
NM_005235
UGACAGUGGAGCAUGUGUUTT
142





ABL2
NM_007314
AUCAGUGAUGUGGUGCAGATT
143





ABL2
NM_007314
GAGUCGGACACUGAAGAAATT
144





ABL2
NM_007314
UGGCACAGCAGGUACUAAATT
145





KRAS2
NM_033360
GAAAAGACUCCUGGCUGUGTT
146





KRAS2
NM_033360
GGACUCUGAAGAUGUACCUTT
147





KRAS2
NM_033360
GGCAUACUAGUACAAGUGGTT
148






NM_021170
AUCCUGGAGAUGACCGUGATT
149






NM_021170
GCCGGUCAUGGAGAAGCGGTT
150






NM_021170
UGGCCCUGAGACUGCAUCGTT
151





ELK1
NM_005229
GCCAUUCCUUUGUCUGCCATT
152





ELK1
NM_005229
GUGAAAGUAGAAGGGCCCATT
153





ELK1
NM_005229
UUCAAGCUGGUGGAUGCAGTT
154





RASAL2
NM_004841
AGUACCAGGAUUCUUCAGCTT
155





RASAL2
NM_004841
CUUAGUUCUGGGCCAUGUATT
156





RASAL2
NM_004841
GACCCCACUGACAGUGAUU1T
157





ARHGEF2
NM_004723
AGCUACACCACAGAUGCCATT
158





ARHGEF2
NM_004723
GGACUUUGCAGCUGACUCUTT
159





ARHGEF2
NM_004723
UAAAGGUUGGGGUGGCCAUTT
160





FRAT1
NM_005479
AAGCUAAUGACGAGGAACCTT
161





FRAT1
NM_005479
CCAUGGUGAAGUGCUUGGATT
162





FRAT1
NM_005479
UAACAGCUGCAAUUCCCUGTT
163





CTNNA2
NM_004389
CCUGAUGAAUGCUGUUGUCTT
164





CTNNA2
NM_004389
GCACAAUACGGUGACCAAUTT
165





CTNNA2
NM_004389
UCACAUCUUGGAGGAUGUGTT
166





AXIN1
AF009674
GAAAGUGAGCGACGAGUUUTT
167





AXIN1
AF009674
GUGCCUUCAACACAGCUUGTT
168





AXIN1
AF009674
UGAAUAUCCAAGAGCAGGGTT
169





EPHB3
NM_004443
GAAGAUCCUGAGCAGUAUCTT
170





EPHB3
NM_004443
GCUGCAGCAGUACAUUGCUTT
171





EPHB3
NM_004443
UACCCUGGACAAGCUCAUCTT
172





DDR1
NM_013994
AACAAGAGGACACAAUGGCTT
173





DDR1
NM_013994
AGAGGUGAAGAUCAUGUCGTT
174





DDRI
NM_013994
UCGCAGACUUUGGCAUGAGTT
175





CLK2
NM_003993
AUCGUUAGCACCUUAGGAGTT
176





CLK2
NM_003993
CCCCUGCCUUGUACAUAAUTT
177





CLK2
NM_003993
GUACAAGGAAGCAGCUCGATT
178





C20orf97
NM_021158
AGUCCCAGGUGGGACUCUUTT
179





C20orf97
NM_021158
CUGGCAUCCUUGAGCUGACTT
180





C20orf97
NM_021158
GACUGUUCUGGAAUGAGGGTT
181






X95425
ACUGCCAGGAGUAAGAACUTT
182






X95425
CUAUUACUGCAGAGGGCUUTT
183






X95425
UGCAUCCUGCAGAGUAUCUTT
184





RPS6KA6
NM_014496
CCUCCUUUCAAACCUGCUUTT
185





RPS6KA6
NM_014496
GAGGUUCUGUUUACAGAGGTT
186





RPS6KA6
NM_014496
UCAGCCAGUGCAGAUUCAATT
187






AB002301
AGACAAAGAGGGGACCUUCTT
188






AB002301
GAAAGUCUAUCCGAAGGCUTT
189






AB002301
UGCCUCCCUGAAACUUCGATr
190





GPRK6
NM_002082
AAGCAAGAAAUGGCGGCAGTT
191





GPRK6
NM_002082
GAGCUGAAUGUCUUUGGGCTT
192





GPRK6
NM_002082
UGUAUAUAGCGACCAGAGCTT
193





GSK3A
NM_019884
CUUCAGUGCUGGUGAACUCTT
194





GSK3A
NM_019884
GCUGGACCACUGCAAUAUUTT
195





GSK3A
NM_019884
GUGGCUUACACGGACAUCATT
196





RAD51L1
NM_133510
AACAGGACCGUACUGCUUGTT
197





RAD51L1
NM_133510
GAAGCCUUUGUUCAGGUCUTT
198





RAD51L1
NM_133510
GAGAGGCAUCCUCCUUGAATT
199





NOTCH4
NM_004557
CCAGCACUGACUACUGUGUTT
200





NOTCH4
NM_004557
GGAACUCGAUGCUUGUCAGTT
201





NOTCH4
NM_004557
UGCGAGGAAGAUACGGAGUTT
202





MCM3
NM_002388
GCAGAUGAGCAAGGAUGCUTT
203





MCM3
NM_002388
GUACAUCCAUGUGGCCAAATT
204





MCM3
NM_002388
UGGGUCAUGAAAGCUGCCATT
205





FZD4
NM_012193
AGAACCUCGGCUACAACGUTT
206





FZD4
NM_012193
UCCGCAUCUCCAUGUGCCATT
207





FZD4
NM_012193
UCGGCUACAACGUGACCAATT
208

















TABLE IIIB










siRNA sequences used in screens of DNA damaging



agents: doxorubicin screen


















GENE_


SEQ ID


SYMBOL
SEQUENCE_ID
SENSE_SEQ
NO














AATK
AB014541
CGCAAGAAGAAGGCCGUGUTT
209






AATK
AB014541
CGCUGGUGCAAUGUUUUCUTT
210





AATK
AB014541
GAAUCCCUACCGAGACUCUTT
211





ABL1
NM_007313
AAACCUCUACACGUUCUGCTT
212





ABL1
NM_007313
CUAAAGGUGAAAAGCUCCGTT
213





ABL1
NM_007313
UCCUGGCAAGAAAGCUUGATT
214





ACVR2
NM_001616
AAGAUGGCCACAAACCUGCTT
215





ACVR2
NM_001616
AGAUAAACGGCGGCAUUGUTT
216





ACVR2
NM_001616
GACAUGCAGGAAGUUGUUGTT
217





ACVR2B
NM_001106
CGGGAGAUCUUCAGCACACTT
218





ACVR2B
NM_001106
GAGAUUGGCCAGCACCCUUTT
219





ACVR2B
NM_001106
GCCCAGGACAUGAGUGUCUTT
220





ADRBK2
NM_005160
CGAGGAUGAGGCAUCUGAUTT
221





ADRBK2
NM_005160
CUGAAGUCCCUUUUGGAGGTT
222





ADRBK2
NM_005160
GAACUUCCCUUUGGUCAUCTT
223





AKT1
NM_005163
GCUGGAGAACCUCAUGCUGTT
224





AKT1
NM_005163
AGACGUUUUUGUGCUGUGGTT
225





AKT1
NM_005163
CGCACCUUCCAUGUGGAGATT
226





AKT2
NM_001626
AGAUGGCCACAUCAAGAUCTT
227





AKT2
NM_001626
GUCAUCAUUGCCAAGGAUGTT
228





AKT2
NM_001626
UGCCAGCUGAUGAAGACCGTT
229





ALK
NM_004304
ACCAGAGACCAAAUGUCACTT
230





ALK
NM_004304
AUAAGCCCACCAGCUUGUGTT
231





ALK
NM_004304
UCAACACCGCUUUGCCGAUTT
232





ALS2CR7
NM_139158
CUGGCUGAUUUUGGUCUUGTT
233





ALS2CR7
NM_139158
GCCUUCAUGUUGUCUGGAATT
234





ALS2CR7
NM_139158
UCCACACCAAAGAGACACUTT
235





AXIN1
AF009674
GAAAGUGAGCGACGAGUUUTT
236





AXIN1
AF009674
GUGCCUUCAACACAGCUUGTT
237





AXIN1
AF009674
UGAAUAUCCAAGAGCAGGGTT
238





BLK
NM_001715
AGUCACGAGCGUUCGAAAATT
239





BLK
NM_001715
CAACAUGAAGGUGGCCAUUTT
240





BLK
NM_001715
GCACUAUAAGAUCCGCUGCTT
241





BMPR2
NM_001204
CAAAUCUGUGAGCCCAACATT
242





BMPR2
NM_001204
CAAGAUGUUCUUGCACAGGTT
243





BMPR2
NM_001204
GAACGGCUAUGUGCGUUUATT
244





BRAF
NM_004333
ACACUUGGUAGACGGGACUTT
245





BRAF
NM_004333
GUCAAUCAUCCACAGAGACTT
246





BRAF
NM_004333
UUGCAUGUGGAAGUGUUGGTT
247





C20orf97
NM_021158
AGUCCCAGGUGGGACUCUUTT
248





C20orf97
NM_021158
CUGGCAUCCUUGAGCUGACTT
249





C20orf97
NM_021158
GACUGUUCUGGAAUGAGGGTT
250





CAMK2G
BC021269
GACAUUGUGGCCAGAGAGUTT
251





CAMK2G
BC021269
GAUGAGGACCUCAAAGUGCTT
252





CAMK2G
BC021269
GGCUGGAGCCUAUGAUUUCTT
253





CCND3
NM_001760
AAAGCAUGCCCAGACCUUUTT
254





CCND3
NM_001760
AAGGAUCUUUGUGGCCAAGTT
255





CCND3
NM_001760
CUACCUGGAUCGCUACCUGTT
256





CCNE2
NM_057749
CCACAGAUGAGGUCCAUACTT
257





CCNE2
NM_057749
CUGGGGCUUUCUUGACAUGTT
258





CCNE2
NM_057749
GUGGUUAAGAAAGCCUCAGTT
259





CCNG1
NM_004060
AUGGAUUGUUUCUGGGCGUTT
260





CCNG1
NM_004060
CUAUCAGUCUUCCCACAGCTT
261





CCNG1
NM_004060
CUUGCCACUUGAAAGGAGATT
262





CCNG2
NM_004354
CCUCGAGAAAAAGGGCUGATT
263





CCNG2
NM_004354
GCUCAGCUGAAAGCUUGCATT
264





CCNG2
NM_004354
UGCCUAGCCGAGUAUUCUUTT
265





CCNH
NM_001239
GACCCGCUAUCCCAUAUUGTT
266





CCNH
NM_001239
GCCAGCAAUGCCAAGAUCUTT
267





CCNH
NM_001239
UUGCCCUGACUGCCAUUUUTT
268





CCNT2
NM_058241
AGCGCCAGUAAAGAAGAACTT
269





CCNT2
NM_058241
AGGGCAGCCAGUUGUCAUUTT
270





CCNT2
NM_058241
CCACCACUCCAAAAUGAGCTT
271





CDC25B
NM_021874
AGGAUGAUGAUGCAGUUCCTT
272





CDC25B
NM_021874
GACAAGGAGAAUGUGCGCUTT
273





CDC25B
NM_021874
GAGCCCAGUCUGUUGAGUUTT
274





CDC42
NM_044472
ACCUUAUGGAAAAGGGGUGTT
275





CDC42
NM_044472
CCAUCCUGUUUGAAAGCCUTT
276





CDC42
NM_044472
CCCAAAAGGAAGUGCUGUATT
277





CDK3
NM_001258
CGAGAGGAAGCUCUAUCUGTT
278





CDK3
NM_001258
GAGAGGAUGCAUCUGGGGATT
279





CDK3
NM_001258
GAUCAGACUGGAUUUGGAGTT
280





CDK4
NM_000075
CAGUCAAGCUGGCUGACUUTT
281





CDK4
NM_000075
GCGAAUCUCUGCCUUUCGATT
282





CDK4
NM_000075
GGAUCUGAUGCGCCAGUUUTT
283





CDK4
NM_000075
CCCUGGUGUUUGAGCAUGUTT
284





CDK4
NM_000075
CUGACCGGGAGAUCAAGGUTT
285





CDK4
NM_000075
GAGUGUGAGAGUCCCCAAUTT
286





CDKN1A
NM_078467
AACUAGGCGGUUGAAUGAGTT
287





CDKN1A
NM_078467
CAUACUGGCCUGGACUGUUTT
288





CDKN1A
NM_078467
GAUGGUGGCAGUAGAGGCUTT
289





CDKN1C
NM_000076
AAAAACCGGGAUUCCGGCCTT
290





CDKN1C
NM_000076
GCGCAAGAGAUCAGCGCCUTT
291





CDKN1C
NM_000076
GUGGACAGCGACUCGGUGCTT
292





CDKN2B
NM_004936
ACACAGAGAAGCGGAUUUCTT
293





CDKN2B
NM_004936
CUCCAAGAGGUGGGUAAUUTT
294





CDKN2B
NM_004936
UGUCUGCUGAGGAGUUAUGTT
295





CDKN3
NM_005192
CCUGCCUUAAAAAUUACCGTT
296





CDKN3
NM_005192
GAACUAAAGAGCUGUGGUATT
297





CDKN3
NM_005192
GAGGAUCCGGGGCAAUACATT
298





CENPE
NM_001813
GAAAAUGAAGCUUUGCGGGTT
299





CENPE
NM_001813
GAAGAGAUCCCAGUGCUUCTT
300





CENPE
NM_001813
UCUGAAAGUGACCAGCUCATT
301





CHEK1
NM_001274
CCAGUUGAUGUUUGGUCCUTT
302





CHEK1
NM_001274
UCUCAGACUUUGGCUUGGCTT
303





CHEK1
NM_001274
UUCUAUGGUCACAGGAGAGTT
304





CHUK
NM_001278
AAAGGCUGCUCACAAGUUCTT
305





CHUK
NM_001278
AGCUGCUCAACAAACCAGATT
306





CHUK
NM_001278
AUGAGGAACAGGGCAAUAGTT
307





CREBBP
NM_004380
GACAUCCCGAGUCUAUAAGTT
308





CREBBP
NM_004380
GCACAAGGAGGUCUUCUUCTT
309





CREBBP
NM_004380
UGGAGGAGAAUUAGGCCUUTT
310





CTNNA1
NM_001903
CGUUCCGAUCCUCUAUACUTT
311





CTNNA1
NM_001903
UGACAUCAUUGUGCUGGCCTT
312





CTNNA1
NM_001903
UGACCAAAGAUGACCUGUGTT
313





CTNNA2
NM_004389
CCUGAUGAAUGCUGUUGUCTT
314





CTNNA2
NM_004389
GCACAAUACGGUGACCAAUTT
315





CTNNA2
NM_004389
UCACAUCUUGGAGGAUGUGTT
316





CTNNAL1
NM_003798
AAGUGUUGUUGCUGGCAGATT
317





CTNNAL1
NM_003798
ACUUGAGAAGCUUUUGGGGTT
318





CTNNAL1
NM_003798
CUAGAGGUUUUUGCUGCAGTT
319





CUL5
NM_003478
AAGAGUGAGCUGGUCAAUGTT
320





CUL5
NM_003478
AUUUUGGAGUGCUUGGGCATT
321





CUL5
NM_003478
UGGGUAAACAGGGCAGCAATT
322





DAPK2
NM_014326
GAAUAUUUUUGGGACGCCGTT
323





DAPK2
NM_014326
UCCAAGAGGCUCUCAGACATT
324





DAPK2
NM_014326
UCUCAGAAGGUCCUCCUGATT
325





DVL1
NM_004421
GGAGGAGAUCUUUGAUGACTT
326





DVL1
NM_004421
GUACGCCAGCAGCUUGCUGTT
327





DVL1
NM_004421
UCGGAUCACACGGCACCGATT
328





DVL3
NM_004423
ACCCCAGUGAGUUCUUUGUTT
329





DVL3
NM_004423
CCUGGACAAUGACACAGAGTT
330





DVL3
NM_004423
GUUCAUUUAAGCCUCAGGGTT
331





DYRK3
NM_003582
CCAUGUUUGCAUGGCCUUUTT
332





DYRK3
NM_003582
CUUCUGGAGCAAUCCAAACTT
333





DYRK3
NM_003582
UCUUUGGAUGCCCUCCACATT
334





ECT2
NM_018098
ACUGGCUAAAGAUGCUGUGTT
335





ECT2
NM_018098
GACCAUGGGAAAAUUGUGGTT
336





ECT2
NM_018098
GCUUAGUACAGCGGGUUGATT
337





EIF4EBP1
NM_004095
CCACCCCUUCCUUAGGUUGTT
338





EIF4EBP1
NM_004095
CUCACCUGUGACCAAAACATT
339





EIF4EBP1
NM_004095
UAGCCCAGAAGAUAAGCGGTT
340





ELK1
NM_005229
GCCAUUCCUUUGUCUGCCATT
341





ELK1
NM_005229
GUGAAAGUAGAAGGGCCCATT
342





ELK1
NM_005229
UUCAAGCUGGUGGAUGCAGTT
343





EPHB3
NM_004443
GAAGAUCCUGAGCAGUAUCTT
344





EPHB3
NM_004443
GCUGCAGCAGUACAUUGCUTT
345





EPHB3
NM_004443
UACCCUGGACAAGCUCAUCTT
346





ERBB3
NM_001982
CUUUCUGAAUGGGGAGCCUTT
347





ERBB3
NM_001982
UACACACACCAGAGUGAUGTT
348





ERBB3
NM_001982
UGACAGUGGAGCCUGUGUATT
349





ERBB4
NM_005235
GAGUACUCUAUAGUGGCCUTT
350





ERBB4
NM_005235
GCUUCCCAGUCCAAAUGACTT
351





ERBB4
NM_005235
UGACAGUGGAGCAUGUGUUTT
352





ERN1
NM_001433
AAGCCUUACGGUCAUGAUGTT
353





ERN1
NM_001433
GAAUAAUGAAGGCCUGACGTT
354





ERN1
NM 001433
GAUGAUUGCGAUGGAUCCUTT
355





FGFR3
NM_000142
AACAUCAUCAACCUGCUGGTT
356





FGFR3
NM_000142
CACUUCCAGCAUUUAGCUGTT
357





FGFR3
NM_000142
CACUUCUUACGCAAUGCUUTT
358





FOXO1A
NM_002015
CUAUGCGUACUGCAUAGCATT
359





FOXO1A
NM_002015
GACAACGACACAUAGCUGGTT
360





FOXO1A
NM_002015
UACAAGGAACCUCAGAGCCTT
361





FZD4
NM_012193
CCAUCUGCUUGAGCUACUUTT
362





FZD4
NM_012193
GUUGACUUACCUGACGGACTT
363





FZD4
NM_012193
UUGGCAAAGGCUCCUUGUATT
364





FZD4
NM_012193
AGAACCUCGGCUACAACGUTT
365





FZD4
NM_012193
UCCGCAUCUCCAUGUGCCATT
366





FZD4
NM_012193
UCGGCUACAACGUGACCAATT
367





FZD9
NM_003508
GACUUUCCAGACCUGGCAGTT
368





FZD9
NM_003508
GAUCGGGGUCUUCUCCAUCTT
369





FZD9
NM_003508
GGACUUCGCGCUGGUCUGGTT
370





GRB2
NM_002086
AUACGUCCAGGCCCUCUUUTT
371





GRB2
NM_002086
CGGGCAGACCGGCAUGUUUTT
372





GRB2
NM_002086
UGCAGCACUUCAAGGUGCUTT
373





GUCY2D
NM_000180
GAAAUUCCCAGGGGAUCAGTT
374





GUCY2D
NM_000180
GACAGACCGGCUGCUUACATT
375





GUCY2D
NM_000180
GUCACGGAACUGCAUAGUGTT
376





GUK1
NM_000858
CGGCAAAGAUUACUACUUUTT
377





GUK1
NM_000858
GGAGCCCGGCCUGUUUGAUTT
378





GUK1
NM_000858
UCAAGAAAGCUCAAAGGACTT
379





HDAC3
NM_003883
CCCAGAGAUUUUUGAGGGATT
380





HDAC3
NM_003883
UGCCUUCAACGUAGGCGAUTT
381





HDAC3
NM_003883
UGGUACCUAUUAGGGAUGGTT
382





HDAC4
NM_006037
AGAGGACGUUUUCUACGGCTT
383





HDAC4
NM_006037
AUCUGUUUGCAAGGGGAAGTT
384





HDAC4
NM_006037
CAAGAUCAUCCCCAAGCCATT
385





HSPCA
NM_005348
ACACCUGGAGAUAAACCCUTT
386





HSPCA
NM_005348
CCUAUGGGUCGUGGAACAATT
387





HSPCA
NM_005348
UAACCUUGGUACUAUCGCCTT
388





ICAM1
NM_000201
CAGCUAAAACCUUCCUCACTT
389





ICAM1
NM_000201
AACACAAAGGCCCACACUUTT
390





ICAM1
NM_000201
CAGAGUGGAAGACAUAUGCTT
391





IMPDH2
NM_000884
AGAGGGAAGACUUGGUGGUTT
392





IMPDH2
NM_000884
CACUCAUGCCAGGACAUUGTT
393





IMPDH2
NM_000884
GAAGAAUCGGGACUACCCATT
394





INPP5D
NM_005541
AGCAUUAAGAAGCCCAGUGTT
395





INPP5D
NM_005541
GAACAAGCACUCAGAGCAGTT
396





INPP5D
NM_005541
UCCCAUCAACAUGGUGUCCTT
397





IRS2
NM_003749
CACAGCCGUUCGAUGUCCATT
398





IRS2
NM_003749
GUACAUCGCCAUCGACGUGTT
399





IRS2
NM_003749
GUACCUGAUCGCCCUCUACTT
400





KIF26A
XM_050278
AUGCGGAAUUUGCCGUGGGTT
401





KIF26A
XM_050278
GCACAAGCACCUGUGUGAGTT
402





KIF26A
XM_050278
GUCGUACACCAUGAUCGGGTT
403





KIF4B
AF241316
CCUGCAGCAACUGAUUACCTT
404





KIF4B
AF241316
GAACUUGAGAAGAUGCGAGTT
405





KIF4B
AF241316
GAAGAGGCCCACUGAAGUUTT
406





KIF5B
NM_004521
AAUGCAUCUCGUGAUCGCATT
407





KIF5B
NM_004521
AGACAGUUGGAGGAAUCUGTT
408





KIF5B
NM_004521
AUCGGCAACUUUAGCGAGUTT
409





KRAS2
NM_033360
GAAAAGACUCCUGGCUGUGTT
410





KRAS2
NM_033360
GGACUCUGAAGAUGUACCUTT
411





KRAS2
NM_033360
GGCAUACUAGUACAAGUGGTT
412





MAP2K2
NM_030662
ACCACACCUUCAUCAAGCGTT
413





MAP2K2
NM_030662
AGUCAGCAUCGCGGUUCUCTT
414





MAP2K2
NM_030662
GAAGGAGAGCCUCACAGCATT
415





MAP3K1
AF042838
UCACUUAGCAGCUGAGUCUTT
416





MAP3K1
AF042838
UUGACAGCACUGGUCAGAGTT
417





MAP3K1
AF042838
UUGGCAAGAACUUCUUGGCTT
418





MAP3K4
NM_005922
AGAACGAUCGUCCAGUGGATT
419





MAP3K4
NM_005922
GGUACCUCGAUGCCAUAGUTT
420





MAP3K4
NM_005922
UUUUGGACUAGUGCGGAUGTT
421





MAP3K5
NM_005923
AGAAUUGGCAGCUGAGUUGTT
422





MAP3K5
NM_005923
UGCAGCAGCUAUUGCACUUTT
423





MAP3K5
NM_005923
UGUACAGCUUGGAAGGAUGTT
424





MAP4K2
NM_004579
GAAUCCUAAGAAGAGGCCGTT
425





MAP4K2
NM_004579
GAGGAGGUCUUUCAUUGGGTT
7426





MAP4K2
NM_004579
GAUAGUCAAGCUAGACCCATT
427





MAP4K3
NM_003618
AAUGGGAUGCUGGCAAUGATT
428





MAP4K3
NM_003618
AUCCUUACACGGGCCAUAATT
429





MAP4K3
NM_003618
CUGGACCUCUGUCAGAACUTT
430





MAPK8
NM_139049
CACCCGUACAUCAAUGUCUTT
431





MAPK8
NM_139049
GGAAUAGUAUGCGCAGCUUTT
432





MAPK8
NM_139049
GUGAUUCAGAUGGAGCUAGTT
433





MAPRE1
NM_012325
GAGUAUUAACAGCCUGGACTT
434





MAPRE1
NM_012325
GCUAAGCUAGAACACGAGUTT
435





MAPRE1
NM_012325
UAGAGGAUGUGUUUCAGCCTT
436





MARK1
NM_018650
ACAACAGCACUCUUCAGUCTT
437





MARK1
NM_018650
CUGCGAGAGCGAGUUUUACTT
438





MARK1
NM_018650
UGUGUAUUCUGGAGGUAGCTT
439





MATK
NM_002378
AGCGGAAACACGGGACCAATT
440





MATK
NM_002378
GUGUGAUGUGACAGCCCAGTT
441





MATK
NM_002378
UGUCACUGAAAGAGGUGUCTT
442





MCC
NM_002387
AGUUGAGGAGGUUUCUGCATT
443





MCC
NM_002387
GACUUAGAGCUGGGAAUCUTT
444





MCC
NM_002387
GGAUUAUAUCCAGCAGCUCTT
445





MCM3
NM_002388
GCAGAUGAGCAAGGAUGCUTT
446





MCM3
NM_002388
GUACAUCCAUGUGGCCAAATT
447





MCM3
NM_002388
UGGGUCAUGAAAGCUGCCATT
448





MET
NM_000245
AUGCCUCUGGAGUGUAUUCTT
449





MET
NM_000245
AUGCGCCCAUCCUUUUCUGTT
450





MET
NM_000245
GAUCUGGGCAGUGAAUUAGTT
451





MPHOSPH1
NM_016195
AGAGGAACUCUCUGCAAGCTT
452





MPHOSPH1
NM_016195
CUGAAGAAGCUACUGCUUGTT
453





MPHOSPH1
NM_016195
GACAUGCGAAUGACACUAGTT
454





MPHOSPH1
NM_016195
AAGUUUGUGUCCCAGACACTT
455





MPHOSPH1
NM_016195
AAUGGCAGUGAAACACCCUTT
456





MPHOSPH1
NM_016195
AUGAAGGAGAGUGAUCACCTT
457





MYO3A
NM_017433
AAAGCUACCGAUGUCAGGGTT
458





MYO3A
NM_017433
AAAUCCCGAGUUAUCCACCTT
459





MYO3A
NM_017433
GGCUAAUGAAAGGUGCUGGTT
460





NEK1
AB067488
AAGUGACAUUUGGGCUCUGTT
461





NEK1
AB067488
AUGCACGUGCUGCUGUACUTT
462





NEK1
AB067488
GAAGGACCUUCUGAUUCUGTT
463





NFKB2
NM_002502
AGGAUUCUCAUGGGAAGGGTT
464





NFKB2
NM_002502
GAAGAACAUGAUGGGGACUTT
465





NFKB2
NM_002502
GAUUGAGCGGCCUGUAACATT
466





NOTCH1
AF308602
AGGCAAGCCCUGCAAGAAUTT
467





NOTCH1
AF308602
AUAUCGACGAUUGUCCAGGTT
468





NOTCH1
AF308602
CACUUACACCUGUGUGUGCTT
469





NOTCH3
NM_000435
AAUGGCUUCCGCUGCCUCUTT
470





NOTCH3
NM_000435
GAACAUGGCCAAGGGUGAGTT
471





NOTCH3
NM_000435
GAGUCUGGGACCUCCUUCUTT
472





NOTCH4
NM_004557
CCAGCACUGACUACUGUGUTT
473





NOTCH4
NM_004557
GGAACUCGAUGCUUGUCAGTT
474





NOTCH4
NM_004557
UGCGAGGAAGAUACGGAGUTT
475





NR4A2
NM_006186
AAGGCCGGAGAGGUCGUUUTT
476





NR4A2
NM_006186
CAUCGACAUUUCUGCCUUCTT
477





NR4A2
NM_006186
GUCACAUGGGCAGAGAUAGTT
478





NRAS
NM_002524
AAGAGCCACUUUCAAGCUGTT
479





NRAS
NM_002524
AGUAGCAACUGCUGGUGAUTT
480





NRAS
NM_002524
CCUCUACAGGGAGCAGAUUTT
481





PAK1
NM_002576
CCGCUGUCUCGAUAUGGAUTT
482





PAK1
NM_002576
GGACCGAUUUUACCGAUCCTT
483





PAK1
NM_002576
UGGAUGGCUCUGUCAAGCUTT
484





PDGFRB
NM_002609
AAAGAAGUACCAGCAGGUGTT
485





PDGFRB
NM_002609
UCCAUCCACCAGAGUCUAGTT
486





PDGFRB
NM_002609
UUUGCUGAGCUCCAUCGGATT
487





PDZGEF2
NM_016340
AACCCUCAUCCACAGGUGATT
488





PDZGEF2
NM_016340
CCGACUGAGUACAUCGAUGTT
489





PDZGEF2
NM_016340
GCCAGAUUCGACUGAUUGUTT
490





PIK3C2A
NM_002645
AACGAGGAAUCCGACAUUCTT
491





PIK3C2A
NM_002645
UGAUGAGCCCAUCCUUUCATT
492





PIK3C2A
NM_002645
UGCUUCAACGGAUGUAGCATT
493





PIK3CA
NM_006218
AGGUGCACUGCAGUUCAACTT
494





PIK3CA
NM_006218
UGGCUUUGAAUCUUUGGCCTT
495





PIK3CA
NM_006218
UUCAGCUAGUACAGGUCCUTT
496





PIK3CB
NM_006219
AAGUUCAUGUCAGGGCUGGTT
497





PIK3CB
NM_006219
AAUGCGCAAAUUCAGCGAGTT
498





PIK3CB
NM_006219
CAAAGAUGCCCUUCUGAACTT
499





PKD2
NM_000297
CGGCUAGUACGUGAAGAGUTT
500





PKD2
NM_000297
CUUAUAGUGGAGCUGGCUATT
501





PKD2
NM_000297
GAUAGCGGACAUAGCUCCATT
502





PLCG1
NM_002660
ACUACGUGGAAGAGAUGGUTT
503





PLCG1
NM_002660
AGGCAAGAAGUUCCUUCAGTT
504





PLCG1
NM_002660
GGAGAAUGGUGACCUCAGUTT
505





POLS
NM_006999
ACAGAGACGCCGAAAGUACTT
506





POLS
NM_006999
CUAGCGACAUAGACCUGGUTT
507





POLS
NM_006999
GCAAAUGAAUUGGCCUGGCTT
508





PRKACB
NM_002731
AAACCCUUGGAACAGGUUCTT
509





PRKACB
NM_002731
CAAGAUGACAUCUGAGCUCTT
510





PRKACB
NM_002731
UGUCUGAUCGAUCAUGCAGTT
511





PRKCL1
NM_002741
CACAAGAUCGUCUACAGGGTT
512





PRKCL1
NM_002741
CACCAGUGAAGUCAGCACUTT
513





PRKCL1
NM_002741
GAUUUCAAGUUCCUGGCGGTT
514





PRKCL2
NM_006256
AAGUGGCUCCUCAUUGUACTT
515





PRKCL2
NM_006256
AUCAUUCUGGCACCUUCAGTT
516





PRKCL2
NM_006256
GUAAAAGCGGAAGUAGUCGTT
517





PRKCQ
NM_006257
AAAUGAAGCAAGGCCGCCATT
518





PRKCQ
NM_006257
ACGGAGUACAGACGUCUCATT
519





PRKCQ
NM_006257
GGAAGCAAAGGACCUUCUGTT
520





PRKG2
NM_006259
AAGACUGGAUCCUCAGCAGTT
521





PRKG2
NM_006259
CAAGUGCAUCCAGCUGAACTT
522





PRKG2
NM_006259
GAAAUUCCUGCACAAUGGGTT
523





PRKWNK3
AJ409088
ACCAAGCAGCCAGCUAUACTT
524





PRKWNK3
AJ409088
CUAAUGACAUCUGGGACCUTT
525





PRKWNK3
AJ409088
CUACGAAGGAAAACGUCAGTT
526





PRKY
NM_002760
AGACAGUGAAGCUGGUUGUTT
527





PRKY
NM_002760
GAAUUUCUGAGGACGAGCUTT
528





PRKY
NM_002760
UCAGAUUUGGGCCAGAGUUTT
529





PTEN
NM_000314
UGGAGGGGAAUGCUCAGAATT
530





PTEN
NM_000314
AAGGCAGCUAAAGGAAGUGTT
531





PTEN
NM_000314
UAAAGAUGGCACUUUCCCGTT
532





PTK2
NM_005607
CUUGGACGAUGUAUUGGAGTT
533





PTK2
NM_005607
GACUGAAAAUGCUUGGGCATT
534





PTK2
NM_005607
UCCCACACAUCUUGCUGACTT
535





PTK6
NM_005975
AACACCCUCUGCAAAGUUGTT
536





PTK6
NM_005975
CCGUGGUUCUUUGGCUGCATT
537





PTK6
NM_005975
UCAGGCUUAUCCGAUGUGCTT
538





RAB2
NM_002865
AAUUGGCCCUCAGCAUGCUTT
539





RAB2
NM_002865
GAAGGUGAAGCUUUUGCACTT
540





RAB2
NM_002865
GAGGUUUCAGCCAGUGCAUTT
541





RAD51L1
NM_133510
AACAGGACCGUACUGCUUGTT
542





RAD51L1
NM_133510
GAAGCCUUUGUUCAGGUCUTT
543





RAD51L1
NM_133510
GAGAGGCAUCCUCCUUGAATT
544





RAF1
NM_002880
CACUCUCUACCGAAGAUCATT
545





RAF1
NM_002880
GAUCCUAAAGGUUGUCGACTT
546





RAF1
NM_002880
GGAAGCCAUUUGCAGUGCUTT
547





RASAL2
NM_004841
AGUACCAGGAUUCUUCAGCTT
548





RASAL2
NM_004841
CUUAGUUCUGGGCCAUGUATT
549





RASAL2
NM_004841
GACCCCACUGACAGUGAUUTT
550





RASD1
NM_016084
CAAAACCAAGGAGAACGUGTT
551





RASD1
NM_016084
CCUAAGGAGGACCUUUUUGTT
552





RASD1
NM_016084
GAAACCGUCAUGCCCGCUUTT
553





RHOK
NM_002929
AGUACACAGCAGGUUCAUCTT
554





RHOK
NM_002929
CGUGAAUGAGGAGAACCCUTT
555





RHOK
NM_002929
GUUUAAGGAGGGGCCUGUGTT
556





SOS1
NM_005633
AUUGACCACCAGGUUUCUGTT
557





SOS1
NM_005633
CUUACAAAAGGGAGCACACTT
558





SOS1
NM_005633
UAUCAGACCGGACCUCUAUTT
559





SRC
NM_005417
CAAUUCGUCGGAGGCAUCATT
560





SRC
NM_005417
GCAGUGCCUGCCUAUGAAATT
561





SRC
NM_005417
GGGGAGUUUGCUGGACUUUTT
562





SRC
NM_005417
GAACCGGAUGCAGUUGAGCTT
563





SRC
NM_005417
GCCGGAAUACAAGAACGGGTT
564





SRC
NM_005417
GUGGCUCUUAUCCGCAUGATT
565





SRPK1
NM_003137
CGCUUAUGGAACGUGAUACTT
566





SRPK1
NM_003137
GCAACAGAAUGGCAGCGAUTT
567





SRPK1
NM_003137
GUUCUAAUCGGAUCUGGCUTT
568





STAT2
NM_005419
AAAGCCUGCAUCAGAGCUCTT
569





STAT2
NM_005419
AGUUAAUCUCCAGGAACGGTT
570





STAT2
NM_005419
UUUGCUCAGCCCAAACCUUTT
571





STAT4
NM_003151
ACACAGAUCUGCCUCUAUGTT
572





STAT4
NM_003151
CCCUACAAUAAAGGCCGGUTT
573





STAT4
NM_003151
UUAGGAAGGUCCUUCAGGGTT
574





STK10
NM_005990
AGACCAGUACUUCCUCCAGTT
575





STK10
NM_005990
CCAUACUCAGAACUCCUCUTT
576





STK10
NM_005990
GAAAAAGCAUCAGGGGGAATT
577





STK38L
BC028603
AGACACCUUGACAGAAGAGTT
578





STK38L
BC028603
CUCUGGGGAUUUCUCAAUGTT
579





STK38L
BC028603
GGAAGUAAAUGCAGGCCAGTT
580





STK6
NM_003600
ACAGUCUUAGGAAUCGUGCTT
581





STK6
NM_003600
GCACAAAAGCUUGUCUCCATT
582





STK6
NM_003600
UUGCAGAUUUUGGGUGGUCTT
583





STK6
NM_003600
CACCCAAAAGAGCAAGCAGTT
584





STK6
NM_003600
CCUCCCUAUUCAGAAAGCUTT
585





STK6
NM_003600
GACUUUGAAAUUGGUCGCCTT
586





STMN1
NM_005563
AACUGCACAGUGCUGUUGGTT
587





STMN1
NM_005563
UACCCAACGCACAAAUGACTT
588





STMN1
NM_005563
UGGCUAGUACUGUAUUGGCTT
589





SYK
NM_003177
AGAACUGGGCUCUGGUAAUTT
590





SYK
NM_003177
AGAAGUUCGACACGCUCUGTT
591





SYK
NM_003177
GGAAAACCUCAUCAGGGAATT
592





TCF1
NM_000545
AGCCGUGGUGGAGACCCUUTT
593





TCF1
NM_000545
AGUCAAGGAGAAAUGCGGUTT
594





TCF1
NM_000545
CCUCGUCACGGAGGUGCGUTT
595





TGFBR1
NM_004612
GACAUGAUUCAGCCACAGATT
596





TGFBR1
NM_004612
UUCCUCGAGAUAGGCCGUUTT
597





TGFBR1
NM_004612
UUUGGGAGGUCAGUUGUUCTT
598





TGFBR2
NM_003242
CCAGAAAUCCUGCAUGAGCTT
599





TGFBR2
NM_003242
GCAGAACACUUCAGAGCAGTT
600





TIE
NM_005424
AAAAAGGGAUCUGGGGAUGTT
601





TIE
NM_005424
CGUGACGUUAAUGAACCUGTT
602





TIE
NM_005424
GAGCAACGGAUCCUACUUCTT
603





TK1
NM_003258
AAGCACAGAGUUGAUGAGATT
604





TK1
NM_003258
CCUUGCUGGGACUUGGAUCTT
605





TK1
NM_003258
CGCCGGGAAGACCGUAAUUTT
606





TLE1
NM_005077
AGAUGACAAGAAGCACCACTT
607





TLE1
NM_005077
AGGAAGGUGGAUGAUAAGGTT
608





TLE1
NM_005077
CACCUGUUUCCAAACCUUGTT
609





TLK2
NM_006852
AGAGCUGGAUCAUCCCAGATT
610





TLK2
NM_006852
AGGCGUUUAUUCGACGAUGTT
611





ThK2
NM_006852
CACUUGACGGUUGUCCCUUTT
612





TOP3A
NM_004618
GAUCCUCCCUGUCUAUGAGTT
613





TOP3A
NM_004618
GCAAAGAAAUUGGACGAGGTT
614





TOP3A
NM_004618
GGCGAAAACAUCGGGUUUGTT
615





TYRO3
NM_006293
GACUAACAAAGGCAGCUGUTT
616





TYRO3
NM_006293
GCAGCUUGCAUGAAGGAGUTT
617





TYRO3
NM_006293
UGCCCCUUUCCAACUGUCUTT
618





VHL
NM_000551
AGGAAAUAGGCAGGGUGUGTT
619





VHL
NM_000551
CAGAACCCAAAAGGGUAAGTT
620





VHL
NM_000551
GAUCUGGAAGACCACCCAATT
621





WASL
NM_003941
AAACAGGAGGUGUUGAAGCTT
622





WASL
NM_003941
AAGUGGAGCAGAACAGUCGTT
623





WASL
NM_003941
GGACAAUCCACAGAGAUCUTT
624





WEE1
NM_003390
AUCGGCUCUGGAGAAUUUGTT
625





WEE1
NM_003390
CAAGGAUCUCCAGUCCACATT
626





WEE1
NM_003390
UGUACCUGUGUGUCCAUCUTT
627





WNT1
NM_005430
ACGGCGUUUAUCUUCGCUATT
628





WNT1
NM_005430
CCCUCUUGCCAUCCUGAUGTT
629





WNT1
NM_005430
CUAUUUAUUGUGCUGGGUCTT
630





WNT2
NM_003391
AUUUGCCCGCGCAUUUGUGTT
631





WNT2
NM_003391
AACGGGCGAUUAUCUCUGGTT
632





WNT2
NM_003391
AGAAGAUGAAUGGUCUGGCTT
633





ZW10
NM_004724
ACAGUUGCAGGAGUUUUCCTT
634





ZW10
NM_004724
CAAACUGUCAGGCAGCAUUTT
635





ZW10
NM_004724
GCCAGCUUGCAAGAAAUUGTT
636






XM_170783
ACCGACACUUUGGCUUCCATT
637






XM_170783
GAUGAGCGCGGGAAUGUUGTT
638






XM_170783
UGGCCGAGGCCUUCAAGCUTT
639






XM_064050
CAUCAAUCACUCUCUGCUGTT
640






XM_064050
CUAACCCAGGAUGUUCAGGTT
641






XM_064050
GACACUCACCAUGCUGAAATT
642






XM_066649
AAGGGUGACUUUGUGUCCUTT
643






XM_066649
ACCAGGAACAAACCUGUUGTT
644






XM_066649
UUUGAAGGUGGCCCUCCUATT
645






NM_005200
AUGAAGCCUCACCAGGACUTT
646






NM_005200
CACUUUUCCCUCAACGAGGTT
647






NM_005200
UAGUAGCAAAGCAGGAAGGTT
648






NM_139286
GUUGUAGGAGGCAGUGAUGTT
649






NM_139286
UCUCAAUUUGGAAGUCUUGTT
650






NM_139286
UGAUCAAUGAUCGGAUUGGTT
651






NM_013366
CGAUCUGCAGGCCAACAUCTT
652






NM_013366
GAAGUAUGAGCAGCUCAAGTT
653






NM_013366
GGACCUCUUCAUCAAUGAGTT
654






NM_014885
ACCAGGAUUUGGAGUGGAUTT
655






NM_014885
CAAGGCAUCCGUUAUAUCUTT
656






NM_014885
GUGGCUGGAUUCAUGUUCCTT
657






NM_016263
CCAGAUCCUUGUCUGGAAGTT
658






NM_016263
CGACAACAAGCUGCUGGUCTT
659






NM_016263
GAAGCUGUCCAUGUUGGAGTT
660






NM_013367
AGCCAGCAGAUGUAAUUGGTT
661






NM_013367
CAUUUCAAUGAGGCUCCAGTT
662






NM_013367
GUCAUUUACAGAGUGGCUCTT
663






NM_018492
AGGACACUUUGGGUACCAGTT
664






NM_018492
GACCCUAAAGAUCGUCCUUTT
665






NM_018492
GCUGAGGAGAAUAUGCCUCTT
666






NM_006087
CGUGUACUACAACGAGGCCTT
667






NM_006087
UCCCCUCUGACUCCAACUUTT
668






NM_006087
CGAGGCACUCUACGACAUCTT
669






NM_016231
GCAAUGAGGACAGCUUGUGTT
670






NM_016231
UCUCCUUGUGAACAGCAACTT
671






NM_016231
UGUAGCUUUCCACUGGAGUTT
672






XM_095827
AAGGUCUUUACGCCAGUACTT
673






XM_095827
GGAAUGUAUCCGAGCACUGTT
674






XM_095827
UAAGCCUGGUGGUGAUCUUTT
675






NM_145754
CUCAAGGGAAAUAUCCGUGTT
676






NM_145754
GUGUGUUGUGCCUGCUGAATT
677






NM_145754
UCAGGCAUGGCAUUAAAACTT
678






XM_168069
CAAAGUUAUUAGCCCCAAGTT
679






XM_168069
CAGAGGCCAAGUAUAUCAATT
680






XM_168069
CCUGCAGAUUUGCACAGCGTT
681






NM_021170
AUCCUGGAGAUGACCGUGATT
682






NM_021170
GCCGGUCAUGGAGAAGCGGTT
683






NM_021170
UGGCCCUGAGACUGCAUCGTT
684






NM_019089
CCCCUCCAUGCUCAGAACUTT
685






NM_019089
CCUAUCUGGGAAGCCUGUGTT
686






NM_019089
UGCCCCAGUGACAAUAACATT
687






NM_016653
ACCAGGGCCAAAAUUAUGGTT
688






NM_016653
AUAGUGAACCUGGAACUGGTT
689






NM_016653
GCAGUUGCCCCAGAAGUUUTT
690






NM_016281
AGAACACACUGCUUGGUUGTT
691






NM_016281
GACAGUGAACAUGGAACCATT
692






NM_016281
GAGAACUUGCAGCACACACTT
693






NM_012119
ACCUGCCAACCUGCUCAUCTT
694






NM_012119
GAUCUCCUUUAAGGAGCAGTT
695






NM_012119
GCAGCUGUGUAUUUAAGGATT
696






AB002301
AGACAAAGAGGGGACCUUCTT
697






AB002301
GAAAGUCUAUCCGAAGGCUTT
698






AB002301
UGCCUCCCUGAAACUUCGATT
699






NM_018401
AGGUAUGCAUCGUGCAGAATT
700






NM_018401
GCAAUCAAACCGUCAUGACTT
701






NM_018401
UAUCCUGCUGGAUGAACACTT
702






NM_006622
GCAAGGUAUACAAUGCCGUTT
703






NM_006622
UAACUCAGCAACCCAGCAATT
704






NM_006622
UGCCUUGAAGACAGUACCATT
705






A1278633
CCUCAGCCGUAUAAUACGUTT
706






A1278633
CUGCUCUGUUCAAUCCCAGTT
707






A1278633
CUGGGAUUGGCCACCUCUUTT
708






NM_152524
AGAAGGAGAGUGUCAGGUUTT
709






NM_152524
UAUGUACCCCGUUCAGCAATT
710






NM_152524
UUUUGCCUUGGAGUGCUCCTT
711






NM_019013
AAAACCCCCGGGAGUCGUCTT
712






NM_019013
AGUGGCACCAAGUGGCUGGTT
713






NM_019013
GAAACCUGCUUUGUCAUUUTT
714






A1338451
CUGAUGCACUUUGCUGCAGTT
715






A1338451
CUGCAGGUUCAAAUCCCAGTT
716






A1338451
GGGGAAAAAGCUUUGCGUUTT
717






NM_018410
AAAGACCCAGGCUAUCAGATT
718






NM_018410
CAGACCCCAAAUCCAUAAGTT
719






NM_018410
GUCAGUGUCACCCAGCAAATT
720






NM_018123
UAUCGAGCCACCAUUUGUGTT
721






NM_018123
UGAUGCAUAUAGCCGCAACTT
722






NM_018123
UGCACAGGGCCAAAGUUGATT
723

















TABLE IIIC










siRNA sequences used in screens of DNA damaging



agents: camptothecin screen











GENE


SEQ ID



SYMBOL
SEQUENCE_ID
SENSE_SEQ
NO














AATK
AB014541
CGCAAGAAGAAGGCCGUGUTT
724






AATK
AB014541
CGCUGGUGCAAUGUUUUCUTT
725





AATK
AB014541
GAAUCCCUACCGAGACUCUTT
726





ABL1
NM_007313
AAACCUCUACACGUUCUGCTT
727





ABL1
NM_007313
CUAAAGGUCAAAAGCUCCGTT
728





ABL1
NM_007313
UCCUGGCAAGAAAGCUUGATT
729





ABL2
NM_007314
AUCAGUGAUGUGGUGCAGATT
730





ABL2
NM_007314
GACUCGGACACUGAAGAAATT
731





ABL2
NM_007314
UGGCACAGCAGGUACUAAATT
732





ACVR2
NM_001616
AAGAUGGCCACAAACCUGCTT
733





ACVR2
NM_001616
AGAUAAACGGCGGCAUUGUTT
734





ACVR2
NM_001616
GACAUGCAGGAAGUUGUUGTT
735





ACVR2B
NM_001106
CGGGAGAUCUUCAGCACACTT
736





ACVR2B
NM_001106
GAGAUUGGCCAGCACCCUUTT
737





ACVR2B
NM_001106
GCCCAGGACAUGAGUGUCUTT
738





AKT2
NM_001626
AGAUGGCCACAUCAAGAUCTT
739





AKT2
NM_001626
GUCAUCAUUGCCAAGGAUGTT
740





AKT2
NM_001626
UGCCAGCUGAUGAAGACCGTT
741





ANAPC5
NM_016237
ACAGUGCUGAACUUGGCUUTT
742





ANAPC5
NM_016237
CCAAAUGUCAGAGGCACAUTT
743





ANAPC5
NM_016237
UCAAACUGAUGGCUGAAGGTT
744





AXIN1
AF009674
GAAAGUGAGCGACGAGUUUTT
745





AXIN1
AF009674
GUGCCUUCAACACAGCUUGTT
746





AXIN1
AF009674
UGAAUAUCCAAGAGCAGGGTT
747





BCL2
NM_000633
AGGACAUUUGUUGGAGGGGTT
748





BCL2
NM_000633
UCUACCAAUUGUGCCGAGATT
749





BCL2
NM_000633
UGAAGAACGUGGACGCUUUTT
750





BLK
NM_001715
AGUCACGAGCGUUCGAAAATT
751





BLK
NM_001715
CAACAUGAAGGUGGCCAUUTT
752





BLK
NM_001715
GCACUAUAAGAUCCGCUGCTT
753





BMPR1B
NM_001203
ACAGAUUGGAAAAGGUCGCTT
754





BMPR1B
NM_001203
GAAGUUACGCCCCUCAUUCTT
755





BMPR1B
NM_001203
UAUUUGCAGCACAGACGGATT
756





BMPR2
NM_001204
CAAAUCUGUGAGCCCAACATT
757





BMPR2
NM_001204
CAAGAUGUUCUUGCACAGGTT
758





BMPR2
NM_001204
GAACGGCUAUGUGCGUUUATT
759





BRCA1
NM_007296
ACUUAGGUGAAGCAGCAUCTT
760





BRCA1
NM_007296
GGGCAGUGAAGACUUGAUUTT
761





BRCA1
NM_007296
UGAAGUGGGCUCCAGUAUUTT
762





BRCA2
NM_000059
CAAAUGGGCAGGACUCUUATT
763





BRCA2
NM_000059
CUGUUCAGCCCAGUUUGAATT
764





BRCA2
NM_000059
UCUCCAAGGAAGUUGUACCTT
765





C20orf97
NM_021158
AGUCCCAGGUGGGACUCUUTT
766





C20orf97
NM_021158
CUGGCAUCCUUGAGCUGACTT
767





C20orf97
NM_021158
GACUGUUCUGGAAUGAGGGTT
768





CAMK2D
NM_001221
ACCAGAUGGAGUAAAGGAGTT
769





CAMK2D
NM_001221
GCACCCUAAUAUUGUGCGATT
770





CAMK2D
NM_001221
UUGGCAGACUUUGGCUUAGTT
771





CCND1
NM_053056
CAUGUAACCGGCAUGUUUCTT
772





CCND1
NM_053056
CCCACAGCUACUUGGUUUGTT
773





CCND1
NM_053056
UGACCCCGCACGAUUUCAUTT
774





CCNE2
NM_057749
CCACAGAUGAGGUCCAUACTT
775





CCNE2
NM_057749
CUGGGGCUUUCUUGACAUGTT
776





CCNE2
NM_057749
GUGGUUAAGAAAGCCUCAGTT
777





CCNT2
NM_058241
AGCGCCAGUAAAGAAGAACTT
778





CCNT2
NM_058241
AGGGCAGCCAGUUGUCAUUTT
779





CCNT2
NM_058241
CCACCACUCCAAAAUGAGCTT
780





CDC14B
NM_033331
GGCCAUCCCCUCCAUUAAUTT
781





CDC14B
NM_033331
GUAAUUGAAAGGCAGUGCCTT
782





CDC14B
NM_033331
UUGCUAUCACUGUGGCUCUTT
783





CDC16
NM_003903
AGUGGCUUCAAAGAUCCCUTT
784





CDC16
NM_003903
GCAUGUCGUUACCUUGCAGTT
785





CDC16
NM_003903
UAAGCCUAGUGAAACGGUCTT
786





CDC23
NM_004661
AGCAACUGCUGCUUAUUGCTT
787





CDC23
NM_004661
AGCAAGCAAGGAGAUAGGATT
788





CDC23
NM_004661
CCUUCUUUAUGUCAGGAGCTT
789





CDC25B
NM_021874
AGGAUGAUGAUGCAGUUCCTT
790





CDC25B
NM_021874
GACAAGGAGAAUGUGCGCUTT
791





CDC25B
NM_021874
GAGCCCAGUCUGUUGAGUUTT
792





CDC34
NM_004359
ACGUGGACGCCUCCGUGAUTT
793





CDC34
NM_004359
CACCUACUACGAGGGCGGCTT
794





CDC34
NM_004359
CAUCUACGAGACGGGGGACTT
795





CDC37
NM_007065
CGCAUGGAGCAGUUCCAGATT
796





CDC37
NM_007065
GACGGCUUCAGCAAGAGCATT
797





CDC37
NM_007065
GAUUAAGACAGCCGAUCGCTT
798





CDC42
NM_044472
ACCUUAUGGAAAAGGGGUGTT
799





CDC42
NM_044472
CCAUCCUGUUUGAAAGCCUTT
800





CDC42
NM_044472
CCCAAAAGGAAGUGCUGUATT
801





CDC45L
NM_003504
CACCUGCUCAAGUCCUUUGTT
802





CDC45L
NM_003504
GAUCCUUCAGGCCUUGUUCTT
803





CDC45L
NM_003504
UGACAGUGAUGGGUCAGAGTT
804





CDK2
NM_001798
AUGAUAGCGGGGGCUAAGUTT
805





CDK2
NM_001798
GAGCUAUCUGUUCCAGCUGTT
806





CDK2
NM_001798
UCUAUUGCUUCACCAUGGCTT
807





CDK2AP1
NM_004642
AGCAAAUACGCGGAGCUGCTT
808





CDK2AP1
NM_004642
CUGCCCAGGUUUUUUUUGUTT
809





CDK2AP1
NM_004642
GUUACAGUUCAUCUCCCCUTT
810





CDK4
NM_000075
CCCUGGUGUUUGAGCAUGUTT
811





CDK4
NM_000075
CUGACCGGGAGAUCAAGGUTT
812





CDK4
NM_000075
GAGUGUGAGAGUCCCCAAUTT
813





CDK5R2
NM_003936
AGGCGAGAGCCGACUCAAGTT
814





CDK5R2
NM_003936
CCUGGACCGCUAGGGAUACTT
815





CDK5R2
NM_003936
CGCAACCGCGAGAACCUUCTT
816





CDK7
NM_001799
AACUGGCAGAUUUUGGCCUTT
817





CDK7
NM_001799
CUGUCCAGUGGAAACCUUATT
818





CDK7
NM_001799
UAGAACCGCCUUAAGAGAGTT
819





CDKL5
NM_003159
ACAGUACCCAAUUCCGACATT
820





CDKL5
NM_003159
GGAGAAUACUUCUGCUGUGTT
821





CDKL5
NM_003159
UCAGCCACAAUGAUGUCCUTT
822





CDKN1A
NM_078467
AACUAGGCGGUUGAAUGAGTT
823





CDKN1A
NM_078467
CAUACUGGCCUGGACUGUUTT
824





CDKN1A
NM_078467
GAUGGUGGCAGUAGAGGCUTT
825





CHEK1
NM_001274
AUCGAUUCUGCUCCUCUAGTT
826





CHEK1
NM_001274
CUGAAGAAGCAGUCGCAGUTT
827





CHEK1
NM_001274
UGCCUGAAAGAGACUUGUGTT
828





CHEK1
NM_001274
CCAGUUGAUGUUUGGUCCUTT
829





CHEK1
NM_001274
UCUCAGACUUUGGCUUGGCTT
830





CHEK1
NM_001274
UUCUAUGGUCACAGGAGAGTT
831





CHFR
NM_018223
AGACUGCGUCCUUUUCGUCTT
832





CHFR
NM_018223
GAUACCAGCACCAGUGGAATT
833





CHFR
NM_018223
GCAUACCUCAUCCAGCAUCTT
834





CKAP2
NM_018204
CCAAUCACAAGUCCuAUUGTT
835





CKAP2
NM_018204
CUUGUGCGACCUCCUAUUATT
836





CKAP2
NM_018204
GAGAGAAAAGCUCGUCUGATT
837





CREBBP
NM_004380
AUUUUUGCGGCGCCAGAAUTT
838





CREBBP
NM_004380
GAAAAACGGAGGUCGCGUUTT
839





CREBBP
NM_004380
GAAAACAAAUGCCCCGUGCTT
840





CSF1R
NM_005211
AGUGCAGAAAGUCAUCCCATT
841





CSF1R
NM_005211
CAACCUGCAGUUUGGUAAGTT
842





CSF1R
NM_005211
UGAGCCAAGUGGCAGCUAATT
843





CTNNA1
NM_001903
CGUUCCGAUCCUCUAUACUTT
844





CTNNA1
NM_001903
UGACAUCAUUGUGCUGGCCTT
845





CTNNA1
NM_001903
UGACCAAAGAUGACCUGUGTT
846





CTNNAL1
NM_003798
AAGUGUUGUUGCUGGCAGATT
847





CTNNAL1
NM_003798
AACUGAGAAGCUUUUGGGGTT
848





CTNNAL1
NM_003798
CUAGAGGUUUUUGCUGCAGTT
849





CTNNBIP1
NM_020248
AAAUUUGCGCCUCGGUAUCTT
850





CTNNBIP1
NM_020248
ACCUAAGUCCUUCCACCUGTT
851





CTNNB1P1
NM_020248
CACCCUGGAUGCUGUUGAATT
852





CUL1
NM_003592
GACCGCAAACUACUGAUUCTT
853





CUL1
NM_003592
GCCAGCAUGAUCUCCAAGUTT
854





CUL1
NM_003592
UAGACAUUGGGUUCGCCGUTT
855





DAPK2
NM_014326
GAAUAUUUUUGGGACGCCGTT
856





DAPK2
NM_014326
UCCAAGAGGCUCUCAGACATT
857





DAPK2
NM_014326
UCUCAGAAGGUCCUCCUGATT
858





DCC
NM_005215
ACAUCGUGGUGCGAGGUUATT
859





DCC
NM_005215
AUGAGCCGCCAAUUGGACATT
860





DCC
NM_005215
AUGGCAAGUUUGGAAGGACTT
861





DDR1
NM_013994
AACAAGAGGACACAAUGGCTT
862





DDR1
NM_013994
AGAGGUGAAGAUCAUGUCGTT
863





DDR1
NM_013994
UCGCAGACUUUGGCAUGAGTT
864





DMPK
NM_004409
CAAGUGGGACAUGCUGAAGTT
865





DMPK
NM_004409
UAAAAGGCCCUCCAUCUGCTT
866





DMPK
NM_004409
UUGGCCCUGUUCAGCAAUGTT
867





DTX1
NM_004416
AACCCACCUGAUGAGGACUTT
868





DTX1
NM_004416
GACCGAGUUUGGAUCCAACTT
869





DTX1
NM_004416
GAUGGAGUUCCACCUCAUCTT
870





DYRK3
NM_003582
CCAUGUUUGCAUGGCCUUUTT
871





DYRK3
NM_003582
CUUCUGGAGCAAUCCAAACTT
872





DYRK3
NM_003582
UCUUUGGAUGCCCUCCACATT
873





ECU
NM_018098
ACUGGCUAAAGAUGCUGUGTT
874





ECU
NM_018098
GACCAUGGGAAAAUUGUGGTT
875





ECU
NM_018098
GCUUAGUACAGCGGGUUGATT
876





EGR2
NM_000399
CACUACCACCCUUUCCUGUTT
877





EGR2
NM_000399
GUGCAAUGUGAUGGGAGGATT
878





EGR2
NM_000399
UGUUACCGGAGCUGAUUUGTT
879





ELK1
NM_005229
GCCAUUCCUUUGUCUGCCATT
880





ELK1
NM_005229
GUGAAAGUAGAAGGGCCCATT
881





ELK1
NM_005229
UUCAAGCUGGUGGAUGCAGTT
882





ELK1
NM_005229
AGGACCCUUUCAAUGUCCCTT
883





ELK1
NM_005229
CUCUCAUUAUCUCCUCCACTT
884





ELK1
NM_005229
GCUCUCCUUCCAGUUUCCATT
885





EPHA4
NM_004438
CUGGCUACGAACUGAUUGGTT
886





EPHA4
NM_004438
GAUUCCUAUCCGGUGGACUTT
887





EPHA4
NM_004438
GCUAUCGUAUAGUUCGGACTT
888





EPHB3
NM_004443
GAAGAUCCUGAGCAGUAUCTT
889





EPHB3
NM_004443
GCUGCAGCAGUACAUUGCUTT
890





EPHB3
NM_004443
UACCCUGGACAAGCUCAUCTT
891





ETS1
NM_005238
UUCAGCCUGAAAGGUGUAGTT
892





ETS1
NM_005238
ACGCUACGUGUACCGCUUUTT
893





ETS1
NM_005238
UGACUACCCCUCGGUCAUUTT
894





FLT1
NM_002019
ACAUCGAAAACAGCAGGUGTT
895





FLT1
NM_002019
AGGAGGAGUGCAUCUUUGGTT
896





FLT1
NM_002019
UGGAUGAGGACUUUUGCAGTT
897





FOXO1A
NM_002015
CUAUGCGUACUGCAUAGCATT
898





FOXO1A
NM_002015
GACAACGACACAUAGCUGGTT
899





FOXO1A
NM_002015
UACAAGGAACCUCAGAGCCTT
900





FRAT1
NM_005479
AAGCUAAUGACGAGGAACCTT
901





FRAT1
NM_005479
CCAUGGUGAAGUGCUUGGATT
902





FRAT1
NM_005479
UAACAGCUGCAAUUCCCUGTT
903





FRK
NM_002031
ACUAUAGACUUCCGCAACCTT
904





FRK
NM_002031
CAGUAGAUUGCUGUGGCCUTT
905





FRK
NM_002031
CUCCAUACAGCUUCUGAAGTT
906





FZD9
NM_003508
GACUUUCCAGACCUGGCAGTT
907





FZD9
NM_003508
GAUCGGGGUCUUCUCCAUCTT
908





FZD9
NM_003508
GGACUUCGCGCUGGUCUGGTT
909





GPRK6
NM_002082
AAGCAAGAAAUGGCGGCAGTT
910





GPRK6
NM_002082
GAGCUGAAUGUCUUUGGGCTT
911





GPRK6
NM_002082
UGUAUAUAGCGACCAGAGCTT
912





GUK1
NM_000858
CGGCAAAGAUUACUACUUUTT
913





GUK1
NM_000858
GGAGCCCGGCCUGUUUGAUTT
914





GUK1
NM_000858
UCAAGAAAGCUCAAAGGACTT
915





HDAC3
NM_003883
CCCAGAGAUUUUUGAGGGATT
916





HDAC3
NM_003883
UGCCUUCAACGUAGGCGAUTT
917





HDAC3
NM_003883
UGGUACCUAUUAGGGAUGGTT
918





HDAC4
NM_006037
AGAGGACGUUUUCUACGGCTT
919





HDAC4
NM_006037
AUCUGUUUGCAAGGGGAAGTT
920





HDAC4
NM_006037
CAAGAUCAUCCCCAAGCCATT
921





HDAC5
NM_005474
AAACUGUUCUCAGAUGCCCTT
922





HDAC5
NM_005474
CCCAACUUGAAAGUGCGUUTT
923





HDAC5
NM_005474
UGAGAUGCACUCCUCCAGUTT
924





HDAC9
NM_058176
AAGCUUCUUGUAGCUGGUGTT
925





HDAC9
NM_058176
AUAUUGCCUGGACAGGUGGTT
926





HDAC9
NM_058176
CAGCAACAAGAACUCCUAGTT
927





HSPCB
NM_007355
AGCAUUCAUGGAGGCUCUUTT
928





HSPCB
NM_007355
AUUGACAUCAUCCCCAACCTT
929





HSPCB
NM_007355
CUCAGCUUUUGUGGAGCGATT
930





IRS1
NM_005544
AGGGCAGUGGAGACUAUAUTT
931





IRS1
NM_005544
CCAGAGUGCCAAAGUGAUCTT
932





IRS1
NM_005544
GGAUAUAUUUGGCUGGGUGTT
933





KIF17
XM_027915
GAUAACGGCUUCUGGAAGATT
934





KIF17
XM_027915
GCAAAAGCAACUUUGGCAGTT
935





KIF17
XM_027915
GCUCAAUAUCAGCUGGGAATT
936





KIF25
NM_005355
GAGCUAUACCAUGCUGGGATT
937





KIF25
NM_005355
GGAUGGACGGACAGAGGUUTT
938





KIF25
NM_005355
GUUACUGGUGAUUCUCUGCTT
939





KIF26A
XM_050278
AUGCGGAAUUUGCCGUGGGTT
940





KIF26A
XM_050278
GCACAAGCACCUGUGUGAGTT
941





KIF26A
XM_050278
GUCGUACACCAUGAUCGGGTT
942





KIF2C
NM_006845
ACAAAAACGGAGAUCCGUCTT
943





KIF2C
NM_006845
AUAAGCAGCAAGAAACGGCTT
944





KIF2C
NM_006845
GAAUUUCGGGCUACUUUGGTT
945





KIF3B
NM_004798
AAACGGUCCAUUGGUAGGATT
946





KIF3B
NM_004798
AAGUGGAAGGAAGUCGGGATT
947





KIF3B
NM_004798
UGCCAAGCAGUUUGAACUGTT
948





KIF4B
AF241316
CCUGCAGCAACUGAUUACCTT
949





KIF4B
AF241316
GAACUUGAGAAGAUGCGAGTT
950





KIF4B
AF241316
GAAGAGGCCCACUGAAGUUTT
951





KRAS2
NM_033360
GAAAAGACUCCUGGCUGUGTT
952





KRAS2
NM_033360
GGACUCUGAAGAUGUACCUTT
953





KRAS2
NM_033360
GGCAUACUAGUACAAGUGGTT
954





LATS2
NM_014572
AACAGCCAUCCAAGUCUUCTT
955





LATS2
NM_014572
AACCUACCAGCAGAAGGUUTT
956





LATS2
NM_014572
UAGGCUUUUCAGGACCUUCTT
957





MAP2K7
NM_005043
AGUCCUACAGGAAGAGCCCTT
958





MAP2K7
NM_005043
GCUACUUGAACACAGCUUCTT
959





MAP2K7
NM_005043
UCAACGACCUGGAGAACUUTT
960





MAP3K1
AF042838
UCACUUAGCAGCUGAGUCUTT
961





MAP3K1
AF042838
UUGACAGCACUGGUCAGAGTT
962





MAP3K1
AF042838
UUGGCAAGAACUUCUUGGCTT
963





MAP3K4
NM_005922
AGAACGAUCGUCCAGUGGATT
964





MAP3K4
NM_005922
GGUACCUCGAUGCCAUAGUTT
965





MAP3K4
NM_005922
UUUUGGACUAGUGCGGAUGTT
966





MAP4K5
NM_006575
AAGGCUGCCACAAAUGUUGTT
967





MAP4K5
NM_006575
GAAACAGAAGCACGAGAUGTT
968





MAP4K5
NM_006575
UCUCUACAUCUUGGCUGGATT
969





MAPK13
NM_002754
CUCACAGUGGAUGAAUGGATT
970





MAPK13
NM_002754
GAUCAUGGGGAUGGAGUUCTT
971





MAPK13
NM_002754
UACAGCCUUUCAAGCAGAGTT
972





MAPK8
NM_139049
CACCCGUACAUCAAUGUCUTT
973





MAPK8
NM_139049
GGAAUAGUAUGCGCAGCUUTT
974





MAPK8
NM_139049
GUGAUUCAGAUGGAGCUAGTT
975





MAPRE1
NM_012325
GAGUAUUAACAGCCUGGACTT
976





MAPRE1
NM_012325
GCUAAGCUAGAACACGAGUTT
977





MAPRE1
NM_012325
UAGAGGAUGUGUUUCAGCCTT
978





MAPRE3
NM_012326
CAGCUUUGUUCAGGGGCAGTT
979





MAPRE3
NM_012326
CUUCGUGACAUCGAGCUCATT
980





MAPRE3
NM_012326
GGAUUACAACCCUCUGCUGTT
981





MARK1
NM_018650
ACAACAGCACUCUUCAGUCTT
982





MARK1
NM_018650
CUGCGAGAGCGAGUUUUACTT
983





MARK1
NM_018650
UGUGUAUUCUGGAGGUAGCTT
984





MCC
NM_002387
AGUUGAGGAGGUUUCUGCATT
985





MCC
NM_002387
GACUUAGAGCUGGGAAUCUTT
986





MCC
NM_002387
GGAUUAUAUCCAGCAGCUCTT
987





MCM3
NM_002388
AGGAUUUUGUGGCCUCCAUTT
988





MCM3
NM_002388
GUCUCAGCUUCUGCGGUAUTT
989





MCM3
NM_002388
UCCAGGUUGAAGGCAUUCATT
990





MCM3
NM_002388
GCAGAUGAGCAAGGAUGCUTT
991





MCM3
NM_002388
GUACAUCCAUGUGGCCAAATT
992





MCM3
NM_002388
UGGGUCAUGAAAGCUGCCATT
993





MLH1
NM_000249
AACUGAAAGCCCCUCCUAATT
994





MLH1
NM_000249
GAUGGAAAUAUCCUGCAGCTT
995





MLH1
NM_000249
UGCUGUUAGUCGAGAACUGTT
996





MYB
NM_005375
ACAAGAGGUGGAAUCUCCATT
997





MYB
NM_005375
GGUUAUCUGCAGGAGUCUUTT
998





MYB
NM_005375
UCGAACAGAUGUGCAGUGCTT
999





MYO3A
NM_017433
AAAGCUACCGAUGUCAGGGTT
1000





MYO3A
NM_017433
AAAUCCCGAGUUAUCCACCTT
1001





MYO3A
NM_017433
GGCUAAUGAAAGGUGCUGGTT
1002





NEK1
AB067488
AAGUGACAUUUGGGCUCUGTT
1003





NEK1
AB067488
AUGCACGUGCUGCUGUACUTT
1004





NEK1
AB067488
GAAGGACCUUCUGAUUCUGTT
1005





NF1
NM_000267
AUCCUUCAACAAGGCACAGTT
1006





NF1
NM_000267
GUAACUUCAGCAGAGCGAATT
1007





NF1
NM_000267
UACAUGACUCCAUGGCUGUTT
1008





NFKB2
NM_002502
AGGAUUCUCAUGGGAAGGGTT
1009





NFKB2
NM_002502
GAAGAACAUGAUGGGGACUTT
1010





NFKB2
NM_002502
GAUUGAGCGGCCUGUAACATT
1011





NTRK1
NM_002529
CAACGGCAACUACACGCUGTT
1012





NTRK1
NM_002529
CGCCACAGCAUCAAGGAUGTT
1013





NTRK1
NM_002529
GAGUGGUCUCCGUUUCGUGTT
1014





OSR1
NM_005109
GAUACACAAAGAUGGGCUGTT
1015





OSR1
NM_005109
AAACAGCUCAGGCUUUGUCTT
1016





OSR1
NM_005109
GAAUAGUGGCUUACCGCUUTT
1017





PAK1
NM_002576
CCGCUGUCUCGAUAUGGAUTT
1018





PAK1
NM_002576
GGACCGAUUUUACCGAUCCTT
1019





PAK1
NM_002576
UGGAUGGCUCUGUCAAGCUTT
1020





PCNA
NM_002592
AAUUGCGGAUAUGGGACACTT
1021





PCNA
NM_002592
AGUCCAAAGUCUGAUCUGGTT
1022





PCNA
NM_002592
UUUCCUGUGCAAAAGACGGTT
1023





PDGFRB
NM_002609
AAAGAAGUACCAGCAGGUGTT
1024





PDGFRB
NM_002609
UCCAUCCACCAGAGUCUAGTT
1025





PDGFRB
NM_002609
UUUGCUGAGCUGCAUCGGATT
1026





PDZGEF2
NM_016340
AACCCUCAUCCACAGGUGATT
1027





PDZGEF2
NM_016340
CCGACUGAGUACAUCGAUGTT
1028





PDZGEF2
NM_016340
GCCAGAUUCGACUGAUUGUTT
1029





PIK3C2A
NM_002645
AACGAGGAAUCCGACAUUCTT
1030





PIK3C2A
NM_002645
UGAUGAGCCCAUCCUUUCATT
1031





PIK3C2A
NM_002645
UGCUUCAACGGAUGUAGCATT
1032





POLS
NM_006999
ACAGAGACGCCGAAAGUACTT
1033





POLS
NM_006999
CUAGCGACAUAGACCUGGUTT
1034





POLS
NM_006999
GCAAAUGAAUUGGCCUGGCTT
1035





PPARG
NM_015869
AAUGACAGACCUCAGACAGTT
1036





PPARG
NM_015869
UAAGCCUCAUGAAGAGCCUTT
1037





PPARG
NM_015869
UGUCAGUACUGUCGGUUUCTT
1038





PRC1
NM_003981
AAGCAUAUCCGUCUGUCAGTT
1039





PRC1
NM_003981
AGGCUUCCAAAUCUGAUGCTT
1040





PRC1
NM_003981
GGAACUCUUUGAAGGUGUCTT
1041





PRKACA
NM_002730
GAAUGGGGUCAACGAUAUCTT
1042





PRKACA
NM_002730
GGACGAGACUUCCUCUUGATT
1043





PRKACA
NM_002730
GUGUGGCAAGGAGUUUUCUTT
1044





PRKCB1
NM_002738
AGAGCAUGCAUUUUUCCGGTT
1045





PRKCB1
NM_002738
GGAGCCCCAUGCUGUAUUUTT
1046





PRKCB1
NM_002738
UUGGAUGUUAGCGGUACUCTT
1047





PRKCL1
NM_002741
CACAAGAUCGUCUACAGGGTT
1048





PRKCL1
NM_002741
CACCAGUGAAGUCAGCACUTT
1049





PRKCL1
NM_002741
GAUUUCAAGUUCCUGGCGGTT
1050





PRKCM
NM_002742
AAUGAAUGAGGAGGGUAGGTT
1051





PRKCM
NM_002742
CCUUCAUCACCCUGGUGUUTT
1052





PRKCM
NM_002742
GUUCCCUGAAUGUGGUUUCTT
1053





PRKWNK3
AJ409088
ACCAAGCAGCCAGCUAUACTT
1054





PRKWNK3
AJ409088
CUAAUGACAUCUGGGACCUTT
1055





PRKWNK3
AJ409088
CUACGAAGGAAAACGUCAGTT
1056





PRKY
NM_002760
AGACAGUGAAGCUGGUUGUTT
1057





PRKY
NM_002760
GAAUUUCUGAGGACGAGCUTT
1058





PRKY
NM_002760
UCAGAUUUGGGCCAGAGUUTT
1059





PTEN
NM_000314
UGGAGGGGAAUGCUCAGAATT
1060





PTEN
NM_000314
AAGGCAGCUAAAGGAAGUGTT
1061





PTEN
NM_000314
UAAAGAUGGCACUUUCCCGTT
1062





PTK6
NM_005975
AACACCCUCUGCAAAGUUGTT
1063





PTK6
NM_005975
CCGUGGUUCUUUGGCUGCATT
1064





PTK6
NM_005975
UCAGGCUUAUCCGAUGUGCTT
1065





PTTG1
NM_004219
AACAGCCAAGCUUUUCUGCTT
1066





PTTG1
NM_004219
GGCUUUGGGAACUGUCAACTT
1067





PTTG1
NM_004219
UCUGUUGCAGUCUCCUUCATT
1068





RALA
NM_005402
AGACAGGUUUCUGUAGAAGTT
1069





RALA
NM_005402
GUCCAGAUCGAUAUCUUAGTT
1070





RALA
NM_005402
GUUUAGCCAAGAGAAUCAGTT
1071





RALBP1
NM_006788
AAUGAAGAGGUCCAAGGGATT
1072





RALBP1
NM_006788
AGGACCCGUGCAUCUUACUTT
1073





RALBP1
NM_006788
GcUAAAAGAcAGGAGUGUGTT
1074





RAP1A
NM_002884
CAGUGUAUGCUCGAAAUCCTT
1075





RAP1A
NM_002884
GAUGAGCGAGUAGUUGGCATT
1076





RAP1A
NM_002884
UUGGAAAGUGCCAGCAUUCTT
1077





RASA2
NM_006506
AACUGAUGACCUGGGGUCUTT
1078





RASA2
NM_006506
CAAGCAGAGAGCUCACCUATT
1079





RASA2
NM_006506
GAAAACAAGCAAUCCGCAGTT
1080





RET
NM_000323
CUUCGCAGAAAAGAGUCGGTT
1081





RET
NM_000323
GACAUCCAGGAUCCACUGUTT
1082





RET
NM_000323
GUGUGCCGAACUUCACUACTT
1083





RHOK
NM_002929
AGUACACAGCAGGUUCAUCTT
1084





RHOK
NM_002929
CGUGAAUGAGGAGAACCCUTT
1085





RHOK
NM_002929
GUUUAAGGAGGGGCCUGUGTT
1086





RPS6KA6
NM_014496
CCUCCUUUCAAACCUGCUUTT
1087





RPS6KA6
NM_014496
GAGGUUCUGUUUACAGAGGTT
1088





RPS6KA6
NM_014496
UCAGCCAGUGCAGAUUCAATT
1089





SGK2
NM_016276
AGAGCCUUAUGAUCGAGCATT
1090





SGK2
NM_016276
CUCUAUCAUGCCUGCUCCUTT
1091





SGK2
NM_016276
GAGAAGGACCUGUGAAACUTT
1092





SKP2
NM_005983
AAGAACCAGGAGAUAUGGGTT
1093





SKP2
NM_005983
GGUCUCUGGUGUUUGUAAGTT
1094





SKP2
NM_005983
UUUGCCCUGCAGACUUUGCTT
1095





SRC
NM_005417
GAACCGGAUGCAGUUGAGCTT
1096





SRC
NM_005417
GCCGGAAUACAAGAACGGGTT
1097





SRC
NM_005417
GUGGCUCUUAUCCGCAUGATT
1098





SRPK1
NM_003137
CGCUUAUGGAACGUGAUACTT
1099





SRPK1
NM_003137
GCAACAGAAUGGCAGCGAUTT
1100





SRPK1
NM_003137
GUUCUAAUCGGAUCUGGCUTT
1101





STAT3
NM_139276
AUGCCACAGGCCACCUAUATT
1102





STAT3
NM_139276
CGACCUGCAGCAAUACCAUTT
1103





STAT3
NM_139276
GAAUCACAUGCCACUUUGGTT
1104





STAT4
NM_003151
ACACAGAUCUGCCUCUAUGTT
1105





STAT4
NM_003151
CCCUACAAUAAAGGCCGGUTT
1106





STAT4
NM_003151
UUAGGAAGGUCCUUCAGGGTT
1107





STAT5A
NM_003152
CCUGUGGAACCUGAAACCATT
1108





STAT5A
NM_003152
GUCUAUGAUGCUGUUGCCCTT
1109





STAT5A
NM_003152
UGAGAUGAUUCAGAAGGGGTT
1110





STK4
NM_006282
CACCAUUUUGGAUGGCUCCTT
1111





STK4
NM_006282
GGAAAACCAGAUCAACAGCTT
1112





STK4
NM_006282
UUCUGGAUGGCUUGCCUCATT
1113





STK6
NM_003600
ACAGUCUUAGGAAUCGUGCTT
1114





STK6
NM_003600
GCACAAAAGCUUGUCUCCATT
1115





STK6
NM_003600
UUGCAGAUUUUGGGUGGUCTT
1116





TCF3
M31523
AAAGACCCCGUGUAAACCUTT
1117





TCF3
M31523
ACCUCAAGGCCAGCUCAAUTT
1118





TCF3
M31523
AUGGGGCAUUUUGUUGGGATT
1119





TERT
NM_003219
CACCAAGAAGUUCAUCUCCTT
1120





TERT
NM_003219
GAGUGUCUGGAGCAAGUUGTT
1121





TERT
NM_003219
GUUUGGAAGAACCCCACAUTT
1122





TGFBR1
NM_004612
GACAUGAUUCAGCCACAGATT
1123





TGFBR1
NM_004612
UUCCUCGAGAUAGGCCGUUTT
1124





TGFBR1
NM_004612
UUUGGGAGGUCAGUUGUUCTT
1125





TK2
NM_004614
GAUGCCAGAAGUGGACUAUTT
1126





TK2
NM_004614
UACCUGGAAGCAAUUCACCTT
1127





TK2
NM_004614
UUAUGCUGCAUUUGGCUGGTT
1128





TOP2B
NM_001068
ACAUUCCCUGGAGUGUACATT
1129





TOP2B
NM_001068
GAGGAUUUAGCGGCAUUUGTT
1130





TOP2B
NM_001068
GCUGCUGGACUGCAUAAAGTT
1131





TOP3A
NM_004618
GAUCCUCCCUGUCUAUGAGTT
1132





TOP3A
NM_004618
GCAAAGAAAUUGGACGAGGTT
1133





TOP3A
NM_004618
GGCGAAAACAUCGGGUUUGTT
1134





TOP3B
NM_003935
CAAAUGGGACAAAGUGGACTT
1135





TOP3B
NM_003935
CUUUGACCUGAAGGGCUCUTT
1136





TOP3B
NM_003935
UCCAGUCCUUCAAACCAGATT
1137





WASL
NM_003941
AAACAGGAGGUGUUGAAGCTT
1138





WASL
NM_003941
AAGUGGAGCAGAACAGUCGTT
1139





WASL
NM_003941
GGACAAUCCACAGAGAUCUTT
1140





WEE1
NM_003390
AUCGGCUCUGGAGAAUUUGTT
1141





WEE1
NM_003390
CAAGGAUCUCCAGUCCACATT
1142





WEE1
NM_003390
UGUACCUGUGUGUCCAUCUTT
1143





WISP1
NM_003882
AAAUGCCUGUCUCUAGCUGTT
1144





WISP1
NM_003882
AUGGCCAGUUUUCUGGUAGTT
1145





WISP1
NM_003882
CCUGGGCAUUGUUGAGGUUTT
1146





WISP3
NM_003880
ACAGUUUUGUCACUGGCCCTT
1147





WISP3
NM_003880
CAAAAUGGACUCCCUGCUCTT
1148





WISP3
NM_003880
CCAGGGGAAAUCUGCAAUGTT
1149





WNT1
NM_005430
ACGGCGUUUAUCUUCGCUATT
1150





WNT1
NM_005430
CCCUCUUGCCAUCCUGAUGTT
1151





WNT1
NM_005430
CUAUUUAUUGUGCUGGGUCTT
1152





WNT2
NM_003391
AUUUGCCCGCGCAUUUGUGTT
1153





WNT2
NM_003391
AACGGGCGAUUAUCUCUGGTT
1154





WNT2
NM_003391
AGAAGAUGAAUGGUCUGGCTT
1155





WT1
NM_024426
CACUGGCACACUGCUCUUATT
1156





WT1
NM_024426
GACAAGAUACCGGUGCUUCTT
1157





WT1
NM_024426
GACACCAAAGGAGACAUACTT
1158






NM_017719
AGACCUAAUCACACGGAUGTT
1159






NM_017719
AGAUAGCGGGUUCACCUACTT
1160






NM_017719
GUUGACAGACUUUGGGUUCTT
1161






XM_168069
ACUCCAUCUGGUUGACCUGTT
1162






XM_168069
GAUUCAGGUGGAACUGAACTT
1163






XM_168069
GCACCAAGCUCCUCUGAUGTT
1164






XM_170783
ACCGACACUUUGGCUUCCATT
1165






XM_170783
GAUGAGCGCGGGAAUGUUGTT
1166






XM_170783
UGGCCGAGGCCUUCAAGCUTT
1167






XM_064050
CAUCAAUCACUCUCUGCUGTT
1168






XM_064050
CUAACCCAGGAUGUUCAGGTT
1169






XM_064050
GACACUCACCAUGCUGAAATT
1170






XM_066649
AAGGGUGACUUUGUGUCCUTT
1171






XM_066649
ACCAGGAACAAACCUGUUGTT
1172






XM_066649
UUUGAAGGUGGCCCUCCUATT
1173






XM_089006
AAAUCGAGAAGGAGGCUCATT
1174






XM_089006
AUAGUGACCGUCCCUUUGATT
1175






XM_089006
CCAGGUUCCUCCAAAGAUGTT
1176






NM_145754
AAGGGUUCAGCAUCUGACUTT
1177






NM_145754
CCUGGAGACAUUGCACCAGTT
1178






NM_145754
GGUGCUACCUCCUUUCCAGTT
1179






NM_017596
AGUUGCCCACCCUGUUUUUTT
1180






NM_017596
GAAAGAAUCCGUCCGCAUGTT
1181






NM_017596
GCAGCCAGAACUCUCAAAGTT
1182






NM_139286
GUUGUAGGAGGCAGUGAUGTT
1183






NM_139286
UCUCAAUUUGGAAGUCUUGTT
1184






NM_139286
UGAUCAAUGAUCGGAUUGGTT
1185






NM_014885
ACCAGGAUUUGGAGUGGAUTT
1186






NM_014885
CAAGGCAUCCGUUAUAUCUTT
1187






NM_014885
GUGGCUGGAUUCAUGUUCCTT
1188






NM_016263
CCAGAUCCUUGUCUGGAAGTT
1189






NM_016263
CGACAACAAGCUGCUGGUCTT
1190






NM_016263
GAAGCUGUCCAUGUUGGAGTT
1191






NM_013367
AGCCAGCAGAUGUAAUUGGTT
1192






NM_013367
CAUUUCAAUGAGGCUCCAGTT
1193






NM_013367
GUCAUUUACAGAGUGGCUCTT
1194






NM_018492
AGGACACUUUGGGUACCAGTT
1195






NM_018492
GACCCUAAAGAUCGUCCUUTT
1196






NM_018492
GCUGAGGAGAAUAUGCCUCTT
1197






XM_168069
CAAAGUUAUUAGCCCCAAGTT
1198






XM_168069
CAGAGGCCAAGUAUAUCAATT
1199






XM_168069
CCUGCAGAUUUGCACAGCGTT
1200






NM_021170
AUCCUGGAGAUGACCGUGATT
1201






NM_021170
GCCGGUCAUGGAGAAGCGGTT
1202






NM_021170
UGGCCCUGAGACUGCAUCGTT
1203






NM_019089
CCCCUCCAUGCUCAGAACUTT
1204






NM_019089
CCUAUCUGGGAAGCCUGUGTT
1205






NM_019089
UGCCCCAGUGACAAUAACATT
1206






AK024504
AGAGAGCUGGACCAUUCAUTT
1207






AK024504
AUGAGCAAUGCGGAUAGCUTT
1208






AK024504
GCCAUGUGUCUGAUGACAUTT
1209






AB002301
AGACAAAGAGGGGACCUUCTT
1210






AB002301
GAAAGUCUAUCCGAAGGCUTT
1211






AB002301
UGCCUCCCUGAAACUUCGATT
1212






NM_018401
AGGUAUGCAUCGUGCAGAATT
1213






NM_018401
GCAAUCAAACCGUCAUGACTT
1214






NM_018401
UAUCCUGCUGGAUGAACACTT
1215






NM_016457
CAUUGUCCACUGUGACUUGTT
1216






NM_016457
UGAAGUGGCCAUUCUGCAGTT
1217






NM_016457
UGUGGACAUUGCCACUGUCTT
1218






NM_005200
AUGAUCGCACCGCAGAGGUTT
1219






NM_005200
UACAUGACGUACUUGAGUGTT
1220






NM_005200
UGCUAAGGGGAUCGGACAUTT
1221






NM_024322
ACCACUCCGGAUACAUCACTT
1222






NM_024322
ACUAAGGCGUCUGCGAGAUTT
1223






NM_024322
GGACCUCACAGCAACUCUUTT
1224






NM_017769
CUGGUUGCAGUUCCAUUCCTT
1225






NM_017769
GUGAGCAUCCUGGAUCAAATT
1226






NM_017769
UUCAGAGAGUCCACACACCTT
1227






NM_019013
AAAACCCCCGGGAGUCGUCTT
1228






NM_019013
AGUGGCACCAAGUGGCUGGTT
1229






NM_019013
GAAACCUGCUUUGUCAUUUTT
1230






AI338451
CUGAUGCACUUUGCUGCAGTT
1231






AI338451
CUGCAGGUUCAAAUCCCAGTT
1232






AI338451
GGGGAAAAAGCUUUGCGUUTT
1233






NM_018123
UAUCGAGCCACCAUUUGUGTT
1234






NM_018123
UGAUGCAUAUAGCCGCAACTT
1235






NM_018123
UGCACAGGGCCAAAGUUGATT
1236









6.4. Example 4
BRCA1/BARD1 E3 Ubiquitin Ligase as an Anti-Cancer Drug Target

Examples 2 and 3 describe siRNA screens to identify genes that enhance cell killing by DNA damaging agents. In this example, HeLa cells were treated with or without cisplatin, and sensitization by a member of the BRCC complex were investigated (FIG. 19). Prominent amongst the genes whose disruption sensitized cells to DNA damage were BRCA1, BRCA2, BARD1 and RAD51, all members of the BRCC complex that enhances cellular survival following DNA damage (Dong et al., Mol Cell. 2003 November; 12 (5):1087-99). Sensitization by BRCA1, BRCA2 and BARD1 was dose dependent with respect to cisplatin concentration, but sensitization by RAD51 was only seen at low cisplatin concentration (FIG. 1). In other experiments, it was found that disruption of BRCA1 and BRCA2 decreased the IC50 concentrations for cisplatin inhibition of HeLa cell growth >˜4-fold (data not shown). Silencing by BRCA1, BRCA2 and BARD1 siRNA pools ranged from ˜85%-98% (data not shown). Table IV lists siRNA sequences of BARD1 and RAD51 used in this example.


These findings were remarkable in that products of the BRCA1, BRCA2, BARD21 and RAD51 genes are associated with a holoenzyme complex (BRCC) that enhances cellular survival following DNA damage (Dong et al., Mol Cell. 2003 November; 12 (5):1087-99). This complex has E3 Ub ligase activity, most of which can be recovered as a BRCA1/BARD1 heterodimer (Dong et al., Mol Cell. 2003 November; 12 (5):1087-99; Brzovic et al., Nat Struct Biol. 2001 October; 8 (10):833-7). These findings strongly implicate BRCC in mediating sensitivity to cisplatin in our siRNA screens. Surprisingly, siRNA pools to members of the FANC complex (FANCA, FANCC, FANCE and FANCF), another multisubunit complex implicated in determining resistance to cisplatin (Taniguchi et al., Nat Med. 2003 May; 9 (5):568-74), did not increase sensitivity in our assays (data not shown).


To determine if the sensitization to cisplatin by BRCA1 or BRCA2 disruption was affected by the presence or absence of TP53 expression in the target cells, matched pairs of TP53 positive and negative cells generated by stable expression of short hairpin RNAs (shRNAs) targeting TP53 (see, Example 2) were used. TP53-positive or negative cells were supertransfected with siRNA pools to BRCA1 or BRCA2, treated with cisplatin and analyzed for cell growth using Alamar Blue (FIG. 20). TP53-negative cells were ˜10-fold more sensitive to cisplatin when transfected with BRCA1 or BRCA2 siRNAs (IC50˜0.1 nM) than with control siRNA (luciferase, IC50-˜1 nM). The sensitization to cisplatin following BRCA1 or BRCA2 disruption was even more pronounced at lower cisplatin concentrations. TP53-positive cells were less sensitized to cisplatin following BRCA1 or BRCA2 disruption (IC50 ˜0.4 nM). Sensitization to cisplatin following BRCA1 or BRCA2 disruption was similar in magnitude in this assay to the sensitization seen following disruption of CHEK1 (data not shown). Sensitization to DNA damaging agents following BRCA1 and BRCA2 disruption was also investigated using cell cycle analysis. TP53-positive and negative cells were supertransfected with siRNA pools to BRCA1 or BRCA2, treated with one of several DNA damaging agents (cisplatin, camptothecin, doxorubicin and bleomycin) and analyzed for cell cycle distribution by flow cytometry. In all cases, TP53-negative cells were more sensitive to DNA damage following BRCA1 or BRCA2 disruption than in luciferase-transfected cells (data not shown). The response of these cells to bleomycin following BRCA1 disruption is shown in FIG. 21. BRCA1 disruption resulted in more sub-G1 cells (dead cells) following bleomycin treatment of TP53-negative than TP53-positive cells. We conclude that cells lacking TP53 are more sensitive to DNA damage following BRCA1 disruption. FIG. 22 shows results that demonstrate that RAD51/Doxorubicin synergy is greater in TP53− cells.


The cell lines used in this example were HeLa cells, TP53-positive A549 cells and TP53-negative A549 cells. The matched pair of TP53 positive and negative cells were generated by stable transfection of short hairpin RNAs (shRNAs) targeting TP53 (monthly highlt highlight, November 2003). The cells were transfected using pools of siRNAs (pool of 3 siRNA per gene) at 100 nM (each siRNA at 33 nM), or with single siRNA at 100 nM. The following siRNAs were used in our study: Luc control, BRCA1, BRCA2 and BARD1 pool. These transfected cells were then treated with varying concentrations of DNA damaging agents. The concentration for each agent used in the cell cycle analysis is as follows: for HeLa cells, Doxorubicin (10 nM), Camptothecin (6 nM), Cisplatin (400 ng/ml), Mitomycin C (40 nM), Bleomycin (100 ng/ml); for the other cell lines, Doxorubicin (200 nM), Camptothecin (200 nM), Cisplatin (2 ug/ml), Mitomycin C (400 nM), Bleomycin (5 ug/ml).


siRNA transfection was carried out as follows: one day prior to transfection, 2000 (or 100) microliters of a chosen cell line, e.g., cervical cancer HeLa cells (ATCC, Cat. No. CCL-2), grown in DMEM/10% fetal bovine serum (Invitrogen, Carlsbad, Calif.) to approximately 90% confluency were seeded in a 6-well (or 96-well) tissue culture plate at 45,000 (or 2000) cells/well. For each transfection 70 microliters of OptiMEM (Invitrogen) was mixed with 5 microliter of siRNA (Dharmacon, Lafayette, Colo.) from a 20 micromolar stock. For each transfection, a ratio of 20 microliter of OptiMEM was mixed with 5 microliter of Oligofectamine reagent (Invitrogen) and incubated 5 minutes at room temperature. Then 25-microliter OptiMEM/Oligofectamine mixture was mixed with the 75-microliter of OptiMEM/siRNA mixture, and incubated 15-20 minutes at room temperature. 100 (or 10) microliter of the transfection mixture was aliquoted into each well of the 6-well (or 96-well) plate and incubated for 4 hours at 37° C. and 5% CO2.


After 4 hours, 100 microliter/well of DMEM/10% fetal bovine serum with or without DNA damage agents was added to each well to reach the final concentration of each agents as described above. The plates were incubated at 37° C. and 5% CO2 for another 68 hours. Samples from the 6-well plates were analyzed for cell cycle profiles and samples from 96-well plates were analyzed for cell growth with Alamar Blue assay.


For cell cycle analysis, the supernatant from each well was combined with the cells that were harvested by trypsinization. The mixture was then centrifuged at 1200 rpm for 5 minutes. The cells were then fixed with ice cold 70% ethanol for ˜30 minutes. Fixed cells were washed once with PBS and resuspended in 0.5 ml of PBS containing Propidium Iodide (10 microgram/ml) and RNase A(1 mg/ml), and incubated at 37° C. for 30 min. Flow cytometric analysis was carried out using a FACSCalibur flow cytometer (Becton Dickinson) and the data was analyzed using FlowJo software (Tree Star, Inc). The Sub-G1 cell population was used to measure cell death. If the summation of the Sub-G1 population from the (siRNA+DMSO) sample and (Luc+drug) sample is larger than the Sub-G1 population of (siRNA+drug) sample, we define that as sensitization of siRNA silencing to DNA damage.


For Alamar Blue assay, the media from the 96-well plates was removed, and 100 uL/well complete media containing 10% (vol/vol) alamarBlue reagent (BioSource International, Inc) and 1/100th volume 1M Hepes buffer tissue culture reagent was added. The plates were then incubated 1-4 hours at 37° C. and fluorescence was measured by exciting at 544 nm and detecting emission at 590 nm with SPECTRAMax Gemini-Xs Spectrofluorometer (Molceular Devices). The fluorescence signal was corrected for background (no cells). Cell response (survival) in the presence of DNA damaging agents was measured as a percentage of control cell growth in the absence of DNA damaging agents.


Many functions have been ascribed to BRCA1, but the only know enzymatic function is E3 Ub ligase activity. This activity is enhanced by association of BARD1 with BRCA1 and results in autoubiquitylation of the BRCA1/BARD1 complex via an unconventional K6 linkage of ubiquitin (Wu-Baer et al., J Biol. Chem. 2003 Sep. 12; 278 (37):34743-6; Chen et al., J Biol. Chem. 2002 Jun. 14; 277 (24):22085-92), Available evidence suggests that the BRCA1 E3 Ub ligase activity is required for its DNA repair function. Cancer-predisposing mutations within the BRCA1 RING domain abolish its Ub ligase activity and these mutants are unable to reverse gamma-radiation hypersensitivity of BRCA1-null human breast cancer cells (Ruffner et al., Proc Natl Acad Sci USA. 2001 Apr. 24; 98 (9):5134-9). In addition, siRNA-mediated disruption of BRCA1 blocks deposition of polyubiquitin structures in nuclear foci that are sites of DNA repair and checkpoint activation in gamma-irradiated cells (Morris et al., Hum Mol Genet. 2004 Apr. 15; 13 (8):807-17). It is important to note that the ubiquitin linkage (K6) mediated by BRCA1 is distinct from the ubiquitin linkage (K48) that marks proteins for degradation by the proteasome (Wu-Baer et al., J Biol. Chem. 2003 Sep. 12; 278 (37):34743-6; Morris et al., Hum Mol Genet. 2004 Apr. 15; 13 (8):807-17). The function of the K6 linkage is currently unknown, but may serve a signaling function.


Taken together, these findings and those in the literature suggest that an inhibitor of BRCA1 E3 Ub ligase activity might be an effective anti-cancer agent because it would enhance the therapeutic window for DNA damaging agents towards tumor cells (most of which are TP53-negative) relative to normal cells (TP53-positive). Dose-dependence of BRCA1 levels on enhanced sensitivity to cisplatin versus deposition of polyubiquitin in nuclear foci is carried out to gain insight into whether these events are causally linked. Chemical inhibitors of BRCA1 E3 Ub ligase activity are also investigated to establish the role of ubiquitylation in repair of DNA damage.


Evidence suggesting the existence of other E3 Ub ligases with roles in DNA damage repair comes from studies in yeast (Spence et al., Mol Cell Biol. 1995 March; 15 (3):1265-73) showing that DNA damage repair requires Ub ligases with non-proteolytic specificity (K63 linkage). To expedite the identification of those involved in DNA damage repair, we are adding siRNAs for multiple E3 ligases with similar domain structures to BRCA1 (RING finger domain ligases) to our siRNA library with the expectation that those that sensitize cells to DNA damage will be revealed by our library screens.


Table IV siRNA sequences of BARD1 and RAD51

SEQCONTENTSSEQUENCEGENEIDIDSENSE SEQIDNAMENO5093CAGUAAUUCUUAAGGCUAATTNM_000465BARD112375094CUCCUGAGAAGGUCUGCAATTNM_000465BARD112385095CGCAGAAGCAGGCUCAACATTNM_000465BARD112396920GUUAGAGCAGUGUGGCAUATTNM_002875RAD5112406921GGUAUGCACUGCUUAUUGUTTNM_002875RAD5112416922CAGAUUGUAUCUGAGGAAATTNM_002875RAD511242


7. REFERENCES CITED

All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.


Many modifications and variations of the present invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only, and the invention is to be limited only by the terms of the appended claims along with the full scope of equivalents to which such claims are entitled.

Claims
  • 1. A method for identifying a gene whose product modulates the effect of an agent on a cell of a cell type, said method comprising (a) contacting a plurality of groups of one or more cells of said cell type with said agent, wherein each said group of one or more cells comprises one or more different small interfering RNAs (siRNAs) from among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different genes in cells of said cell type; (b) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes; and (c) identifying a gene as said gene whose product modulates the effect of said agent on a cell of said cell type if the effect of said agent on said group of one or more cells comprising said one or more different siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes.
  • 2. The method of claim 1, wherein each said group of cells comprising one or more of said plurality of siRNAs is obtained by transfection with said one or more siRNAs prior to said step of contacting.
  • 3. A method for identifying a gene whose product modulates the effect of an agent on a cell of a cell type, said method comprising (a) transfacting each of a plurality of groups of one or more cells of said cell type with a composition comprising one or more different small interfering RNAs (siRNAs) from among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different genes in cells of said cell type; (b) contacting each of said plurality of groups of one or more cells with said agent; (c) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which is not transfected with an siRNA targeting any one of said different genes; and (d) identifying a gene as said gene whose product modulates the effect of said agent on a cell of said cell type if the effect of said agent on said group of one or more cells comprising said one or more different siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes.
  • 4. The method of any one of claims 1-3, wherein the effect of said agent on said group of one or more cells comprising said siRNA is enhanced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes.
  • 5. The method of any one of claims 1-3, wherein the effect of said agent on said group of one or more cells comprising said siRNA is reduced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different genes.
  • 6. The method of any one of claims 1-3, wherein said agent acts on a gene other than any one of said different genes targeted by said plurality of siRNAs, or a protein encoded thereof.
  • 7. The method of claim 3, wherein said plurality of siRNAs comprises at least k different siRNAs targeting at least one gene of said different genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 8. The method of claim 7, wherein said one or more different siRNAs targeting said at least one gene comprise 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 9. The method of claim 7, wherein said plurality of siRNAs comprises at least k different siRNAs targeting each of at least 2 different genes of said different genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 10. The method of claim 9, wherein said one or more different siRNAs targeting each said at least 2 different genes comprise 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 11. The method of claim 9, wherein said plurality of siRNAs comprises at least k different siRNAs targeting each of said different genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 12. The method of claim 11, wherein said one or more different siRNAs targeting each of said different genes comprise 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 13. The method of claim 5, wherein said cell type is a cancer cell type.
  • 14. The method of claim 13, wherein said cell type is a cancer cell type, and wherein said effect is growth inhibitory effect.
  • 15. The method of claim 12, wherein said agent is a KSP inhibitor.
  • 16. The method of any one of claims 7-15, wherein said plurality of different genes comprises at least N different genes, wherein N is selected from the group consisting of 5, 10, 100, 1,000, and 5,000 different genes.
  • 17. The method of any one of claims 1-3, wherein said different genes are different endogenous genes.
  • 18. A method for identifying a gene which interacts with a primary target gene in a cell of a cell type, said method comprising (a) contacting a plurality of groups of one or more cells of said cell type with an agent, wherein said agent modulates the expression of said primary target gene and/or the activity of a protein encoded by said primary target gene, and wherein each said group of cells comprises one or more different siRNAs among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different secondary genes in said cell; (b) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes; and (c) identifying a gene as said gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said group of one or more cells comprising one or more siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes.
  • 19. The method of claim 18, wherein each said group of cells comprising one or more of said plurality of siRNAs is obtained by transfection with said one or more siRNA prior to said step of contacting.
  • 20. A method for identifying a gene which interacts with a primary target gene in a cell of a cell type, said method comprising (a) transfacting each of a plurality of groups of one or more cells of said cell type with a composition comprising one or more different small interfering RNAs (siRNAs) from among a plurality of different siRNAs, said one or more different siRNAs targeting a same gene, and said plurality of different siRNAs comprising siRNAs targeting respectively different genes in cells of said cell type; (b) contacting said plurality of groups of one or more cells of said cell type with an agent, wherein said agent modulates the expression of said primary target gene and/or the activity of a protein encoded by said primary target gene; (c) comparing the effect of said agent on each said group of one or more cells to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes; and (d) identifying a gene as said gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said group of one or more cells comprising one or more siRNAs targeting said gene is different as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes.
  • 21. The method of any one of claims 18-20, wherein said agent is an siRNA targeting and silencing said primary target gene.
  • 22. The method of any one of claims 18-20, wherein said agent is an inhibitor of said primary target gene.
  • 23. The method of any one of claims 18-20, wherein the effect of said agent on said group of one or more cells comprising said one or more siRNAs is enhanced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes.
  • 24. The method of any one of claims 18-20, wherein the effect of said agent on said group of one or more cell comprising said one or more siRNAs is reduced as compared to the effect of said agent on a cell of said cell type which does not comprise an siRNA targeting any one of said different secondary genes.
  • 25. The method of claim 20, wherein said plurality of siRNAs comprises at least k different siRNAs targeting at least one of said different secondary genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 26. The method of claim 25, wherein said one or more different siRNAs targeting said at least one gene comprise 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 27. The method of claim 18, wherein said plurality of siRNAs comprises at least k different siRNAs targeting each of at least 2 different genes of said different secondary genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 28. The method of claim 27, wherein said one or more different siRNAs targeting each said at least 2 different genes comprise 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 29. The method of claim 27, wherein said plurality of siRNAs comprises at least k different siRNAs targeting each of said different secondary genes, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 30. The method of claim 29, wherein said one or more different siRNAs targeting each of said different genes comprise 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 31. The method of claim 22, wherein said primary target gene is KSP.
  • 32. The method of claim 18, wherein said plurality of different genes comprises at least N different genes, wherein N is selected from the group consisting of 5, 10, 100, 1,000, and 5,000 different genes.
  • 33. The method of any one of claims 18-20, wherein said different secondary genes are different endogenous genes.
  • 34. The method of any one of claims 18-20, wherein said cell type is a cancer cell type.
  • 35. The method of claim 8 or 26, wherein the total siRNA concentration of said one or more siRNAs in said composition is an optimal concentration for silencing said target gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 36. The method of claim 35, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 37. The method of claim 35, wherein the concentration of each said one or more siRNA is about the same.
  • 38. The method of claim 35, wherein the respective concentrations of said one or more siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 39. The method of claim 35, wherein none of the siRNAs in said composition has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said one or more siRNAs.
  • 40. The method of claim 35, wherein at least one siRNA in said composition has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said one or more siRNAs.
  • 41. The method of claim 8 or 26, wherein the number of different siRNAs and the concentration of each siRNA in said composition is chosen such that said composition causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 42. A method for treating a mammal having a cancer, comprising administering to said mammal a therapeutically sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, wherein said mammal is subject to a therapy comprising administering to said mammal a therapeutically sufficient amount of a KSP inhibitor.
  • 43. A method for treating a mammal having a cancer, comprising administering to said mammal i) a therapeutically sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, and ii) a therapeutically sufficient amount of a KSP inhibitor.
  • 44. The method of claim 42 or 43, wherein said agent reduces the expression of said STK6 or TPX2 gene in cells of said cancer.
  • 45. The method of claim 42 or 43, wherein said agent comprises an siRNA targeting said STK6 or TPX2 gene.
  • 46. The method of claim 45, wherein said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said STK6 or TPX2 gene.
  • 47. The method of claim 46, wherein the total siRNA concentration of said different siRNAs in said agent is an optimal concentration for silencing said STK6 or TPX2 gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 48. The method of claim 47, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 49. The method of claim 47, wherein the concentration of each said different siRNA is about the same.
  • 50. The method of claim 47, wherein the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 51. The method of claim 47, wherein none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 52. The method of claim 47, wherein at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs.
  • 53. The method of claim 47, wherein the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 54. The method of claim 45, wherein said mammal is a human, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • 55. A method for treating a mammal having a cancer, comprising administering to said mammal i) a therapeutically sufficient amount of a first agent, said first agent regulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, and ii) a therapeutically sufficient amount of a second agent, said second agent regulating the expression of a KSP gene and/or activity of a protein encoded by said KSP gene.
  • 56. The method of claim 55, wherein said first agent comprises an siRNA targeting said STK6 or TPX2 gene, and said second agent comprises an siRNA targeting said KSP gene.
  • 57. The method of claim 56, wherein said first agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said STK6 or TPX2 gene.
  • 58. The method of claim 57, wherein the total siRNA concentration of said different siRNAs in said first agent is an optimal concentration for silencing said STK6 or TPX2 gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 59. The method of claim 58, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 60. The method of claim 58, wherein the concentration of each said different siRNA is about the same.
  • 61. The method of claim 58, wherein the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 62. The method of claim 58, wherein none of the siRNAs in said first agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 63. The method of claim 58, wherein at least one siRNA in said first agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs.
  • 64. The method of claim 58, wherein the number of different siRNAs and the concentration of each siRNA in said first agent is chosen such that said first agent causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 65. The method of claim 56, wherein said mammal is a human, and wherein said siRNA targeting said STK6 gene is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • 66. The method of claim 45 or 56, wherein said mammal is a human, and wherein said siRNA targeting said TPX2 gene is selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.
  • 67. A method for evaluating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said method comprising determining an expression level of a STK6 or TPX2 gene in said cell, wherein said expression level above a predetermined threshold level indicates that said cell is resistant to the growth inhibitory effect of said KSP inhibitor.
  • 68. The method of claim 67, wherein said expression level of said STK6 or TPX2 gene is determined by a method comprising measuring the expression level of said STK6 or TPX2 gene using one or more polynucleotide probes, each of said one or more polynucleotide probes comprising a nucleotide sequence in said STK6 or TPX2 gene.
  • 69. The method of claim 67 or 68, wherein said one or more polynucleotide probes are polynucleotide probes on a microarray.
  • 70. A method for evaluating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said method comprising determining a level of abundance of a protein encoded by a STK6 or TPX2 gene in said cell, wherein said level of abundance of said protein above a predetermined threshold level indicates that said cell is resistant to the growth inhibitory effect of said KSP inhibitor.
  • 71. A method for evaluating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said method comprising determining a level of activity of a protein encoded by a STK6 or TPX2 gene in said cell, wherein said activity level above a predetermined threshold level indicates that said cell is resistant to the growth inhibitory effect of said KSP inhibitor.
  • 72. The method of claim 70 or 71, wherein said cell is a human cell.
  • 73. A method for regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, comprising contacting said cell with a sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene.
  • 74. A method for regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor in a mammal, comprising administering to said mammal a therapeutically sufficient amount of an agent, said agent regulating the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene.
  • 75. A method for regulating growth of a cell, comprising contacting said cell with i) a sufficient amount of an agent that regulates the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene; and ii) a sufficient amount of a KSP inhibitor.
  • 76. The method of claim 73, 74, or 75, wherein said agent reduces the expression of said STK6 or TPX2 gene in said cell.
  • 77. The method of claim 73, 74, or 75, wherein said agent comprises an siRNA targeting said STK 6 gene.
  • 78. The method of claim 77, wherein said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said STK6 or TPX2 gene.
  • 79. The method of claim 78, wherein the total siRNA concentration of said different siRNAs in said agent is an optimal concentration for silencing said STK6 or TPX2 gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 80. The method of claim 79, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 81. The method of claim 79, wherein the concentration of each said different siRNA is about the same.
  • 82. The method of claim 79, wherein the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 83. The method of claim 79, wherein none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 84. The method of claim 79, wherein at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs.
  • 85. The method of claim 79, wherein the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, less than 0.01% of silencing of any off-target genes.
  • 86. The method of claim 77, wherein said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • 87. The method of claim 77, wherein said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.
  • 88. A method of identifying an agent that is capable of regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, wherein said agent is capable of modulating the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene, said method comprising comparing inhibitory effect of said KSP inhibitor on cells expressing said STK6 or TPX2 gene in the presence of said agent with inhibitory effect of said KSP inhibitor on cells expressing said STK6 or TPX2 gene in the absence of said agent, wherein a difference in said inhibitory effect of said KSP inhibitor identifies said agent as capable of regulating resistance of said cell to the growth inhibitory effect of said KSP inhibitor.
  • 89. A method of identifying an agent that is capable of regulating resistance of a cell to the growth inhibitory effect of a KSP inhibitor, wherein said agent is capable of modulating the expression of a STK6 or TPX2 gene and/or activity of a protein encoded by said STK6 or TPX2 gene, said method comprising: (a) contacting a first cell expressing said STK6 or TPX2 gene with said KSP inhibitor in the presence of said agent and measuring a first growth inhibitory effect; (b) contacting a second cell expressing said STK6 or TPX2 gene with said KSP inhibitor in the absence of said agent and measuring a second growth inhibitory effect; and (c) comparing said first and second inhibitory effects measured in said step (a) and (b), wherein a difference between said first and second inhibitory effects identifies said agent as capable of regulating resistance of a cell to the growth inhibitory effect of said KSP inhibitor.
  • 90. The method of claim 88 or 89, wherein said agent comprises a molecule which reduces expression of said STK6 or TPX2 gene.
  • 91. The method of claim 88 or 89, wherein said agent is an siRNA targeting said STK6 or TPX2 gene.
  • 92. The method of claim 91, wherein said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • 93. The method of claim 91, wherein said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.
  • 94. A cell comprising one or more different small interfering RNAs (siRNAs) targeting a STK6 or TPX2 gene in said cell.
  • 95. The cell of claim 94, wherein said one or more different siRNAs comprises 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 96. The cell of claim 95, wherein said cell is produced by transfection using a composition of said one or more different siRNAs, wherein the total siRNA concentration of said composition is an optimal concentration for silencing said STK6 or TPX2 gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 97. The cell of claim 96, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 98. The cell of claim 96, wherein the concentration of each said different siRNA is about the same.
  • 99. The cell of claim 96, wherein the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 100. The cell of claim 96, wherein none of the siRNAs in said composition has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 101. The cell of claim 96, wherein at least one siRNA in said composition has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs.
  • 102. The cell of claim 96, wherein the number of different siRNAs and the concentration of each siRNA in said composition is chosen such that said composition causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 103. The cell of claim 94, wherein said cell is a human cell.
  • 104. The cell of claim 103, wherein said cell is a human cell, and wherein each of said one or more different siRNAs is selected from the group consisting of siRNAs described by SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • 105. The cell of claim 103, wherein said cell is a human cell, and wherein said siRNA is selected from the group consisting of siRNAs described by SEQ ID NO:1237, SEQ ID NO:1238, and SEQ ID NO:1239.
  • 106. The cell of claim 94, wherein said cell is a murine cell.
  • 107. A microarray for diagnosing resistance of a cell to the growth inhibitory effect of a KSP inhibitor, said microarray comprising one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in a STK6 or TPX2 gene.
  • 108. A kit for diagnosis of resistance of a cell to the growth inhibitory effect of a KSP inhibitor, comprising in one or more containers one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in a STK6 or TPX2 gene.
  • 109. A kit for screening for agents which regulate resistance of a cell to the growth inhibitory effect of a KSP inhibitor, comprising in one or more containers (i) the cell of claim 94; and (ii) a KSP inhibitor.
  • 110. A kit for treating a mammal having a cancer, comprising in one or more containers (i) a sufficient amount of an agent that regulates the expression of a STK6 or TPX2 gene and/or the activity of a protein encoded by said STK6 or TPX2 gene; and (ii) a KSP inhibitor.
  • 111. The method of any one of claims 42-43, 67, 70-71, 74-75 and 88-89, wherein said KSP inhibitor is (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine.
  • 112. The method of claim 1, 2, or 3, wherein said contacting step (a) is carried out separately for each said group of one or more cells.
  • 113. The method of claim 18, 19, or 20, wherein said contacting step (a) is carried out separately for each said group of one or more cells.
  • 114. The kit of claim 109 or 110, wherein said KSP inhibitor is (1S)-1-{[(2S)-4-(2,5-difluorophenyl)-2-phenyl-2,5-dihydro-1H-pyrrol-1-yl]carbonyl}-2-methylpropylamine.
  • 115. A method for identifying a gene that interacts with a primary target gene in a cell of a cell type, said method comprising (a) contacting one or more cells of said cell type with an agent, wherein said agent modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene, and wherein said one or more cells express a first small interference RNA (siRNA) targeting said primary target gene; (b) comparing the effect of said agent on said one or more cells of said clone to the effect of said agent on a cell of said cell type not expressing said first siRNA; and (c) identifying said secondary target gene as a gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said one or more cells expressing said first siRNA is different as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA.
  • 116. A method for identifying a gene which interacts with a primary target gene in a cell of a cell type, said method comprising (a) generating a clone of cells of said cell type which express a first small interference RNA (siRNA) targeting said primary target gene; (b) contacting one or more cells of said clone with an agent, wherein said agent modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; (c) comparing the effect of said agent on said one or more cells of said clone to the effect of said agent on a cell of said cell type not expressing said first siRNA; and (d) identifying said secondary target gene as a gene that interacts with said primary target gene in a cell of said cell type if the effect of said agent on said one or more cells expressing said first siRNA is different as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA.
  • 117. The method of claim 116, wherein said first siRNA is expressed by a nucleotide sequence integrated in the genome of said cells.
  • 118. The method of claim 116, wherein said agent comprises one or more second siRNAs targeting and silencing said secondary target gene.
  • 119. The method of claim 116, wherein said agent is an inhibitor of said secondary target gene.
  • 120. The method of claim 118, wherein the effect of said agent on said one or more cells expressing said first siRNA is enhanced as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA.
  • 121. The method of claim 118, wherein the effect of said agent on said one or more cells expressing said first siRNA is reduced as compared to the effect of said agent on a cell of said cell type not expressing said first siRNA.
  • 122. The method of claim 120, wherein said one or more second siRNAs comprises at least k different siRNAs, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 123. The method of claim 122, wherein the total siRNA concentration of said at least k different siRNAs in said agent is an optimal concentration for silencing said secondary target gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 124. The method of claim 123, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 125. The method of claim 123, wherein the concentration of each said at least k different siRNA is about the same.
  • 126. The method of claim 123, wherein the respective concentrations of said at least k different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 127. The method of claim 123, wherein none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 128. The method of claim 123, wherein at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said at least k different siRNAs.
  • 129. The method of claim 123, wherein the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 130. The method of claim 122, wherein said cell type is a cancer cell type, and wherein said primary target gene is p53.
  • 131. The method of claim 130, further comprising a step (e) repeating steps (b)-(d) for each of a plurality of different secondary target genes.
  • 132. The method of claim 131, wherein said plurality of secondary target genes comprises at least the number of different genes selected from the group consisting of 5, 10, 100, 1,000, and 5,000 different genes.
  • 133. The method of claim 132, wherein said effect is a change in the sensitivity of cells of said cell type to a drug.
  • 134. The method of claim 133, wherein said drug is a DNA damaging agent.
  • 135. The method of claim 134, wherein said DNA damaging agent is selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation.
  • 136. The method of claim 135, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin.
  • 137. A method of evaluating the responsiveness of cells of a cell type to treatment of a drug, comprising (a) contacting one or more cells of said cell type with said drug, wherein said one or more cells express a first small interference RNA (siRNA) targeting a primary target gene, and wherein said one or more cells are subject to treatment of a composition that modulates the expression of one or more secondary target genes and/or the activity of one or more proteins encoded respectively by said one or more secondary target genes; (b) contacting one or more cells of said cell type with said drug, wherein said one or more cells do not express a small interference RNA (siRNA) targeting said primary target gene, and wherein said one or more cells are subject to treatment of said agent that modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; and (c) comparing the effect of said drug on said one or more cells measured in step (a) to the effect of said drug on said one or more cells measured in step (b), thereby evaluating the responsiveness of said cells to treatment of said drug.
  • 138. A method for evaluating the responsiveness of cells of a cell type to treatment of a drug, said method comprising (a) generating a clone of cells of said cell type which express a first small interference RNA (siRNA) targeting a primary target gene; (b) contacting one or more cells of said clone which express said first siRNA with said drug, wherein said one or more cells are subject to treatment of an agent that modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; (c) contacting one or more cells of said cell type which do not express a small interference RNA (siRNA) targeting said primary target gene with said drug, wherein said one or more cells are subject to treatment of said agent that modulates the expression of a secondary target gene and/or the activity of a protein encoded by said secondary target gene; and (d) comparing the effect of said drug on said one or more cells measured in step (b) to the effect of said drug on said one or more cells measured in step (c), thereby evaluating the responsiveness of said cells to treatment of said drug.
  • 139. The method of claim 137 or 138, wherein the effect of said drug on said one or more cells expressing said first siRNA is enhanced as compared to the effect of said drug on a cell of said cell type not expressing said first siRNA.
  • 140. The method of claim 137 or 138, wherein the effect of said drug on said one or more cells expressing said first siRNA is reduced as compared to the effect of said drug on a cell of said cell type not expressing said first siRNA.
  • 141. The method of claim 137 or 138, wherein said composition comprises one or more inhibitors of said one or more secondary target gene.
  • 142. The method of claim 137 or 138, wherein said composition comprises one or more second siRNAs targeting and silencing said one or more secondary target gene.
  • 143. The method of claim 142, wherein said one or more second siRNAs comprises at least k different siRNAs, wherein said k is selected from the group consisting of 2, 3, 4, 5, 6 and 10.
  • 144. The method of claim 143, wherein the total siRNA concentration of said at least k different siRNAs in said agent is an optimal concentration for silencing said secondary target gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 145. The method of claim 144, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 146. The method of claim 144, wherein the concentration of each said at least k different siRNA is about the same.
  • 147. The method of claim 144, wherein the respective concentrations of said at least k different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 148. The method of claim 144, wherein none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 149. The method of claim 144, wherein at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said at least k different siRNAs.
  • 150. The method of claim 144, wherein the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 151. The method of claim 137 or 138, wherein said cell type is a cancer cell type, and wherein said primary target gene is p53.
  • 152. The method of claim 138, further comprising a step (e) repeating steps (b)-(d) for each of a plurality of different secondary target genes.
  • 153. The method of claim 137, further comprising a step (d) repeating steps (a)-(b) for each of a plurality of different secondary target genes.
  • 154. The method of claim 152 or 153, wherein said plurality of secondary target genes comprises at least the number of different genes selected from the group consisting of 5, 10, 100, 1,000, and 5,000 different genes.
  • 155. The method of claim 154, wherein said drug is a DNA damaging agent.
  • 156. The method of claim 155, wherein said DNA damaging agent is selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation.
  • 157. The method of claim 156, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin.
  • 158. A method for treating a mammal having a cancer, comprising administering to said mammal a therapeutically sufficient amount of an agent, said agent regulating the expression of a gene and/or activity of a protein encoded by said gene, wherein said mammal is subject to a therapy comprising administering to said mammal a therapeutically sufficient amount of a composition comprising one or more DNA damaging agents.
  • 159. A method for treating a mammal having a cancer, comprising administering to said mammal i) a therapeutically sufficient amount of an agent, said agent regulating the expression of a gene and/or activity of a protein encoded by said gene, and ii) a therapeutically sufficient amount of a composition comprising one or more DNA damaging agents.
  • 160. The method of claim 158 or 159, wherein said agent reduces the expression of said gene in cells of said cancer.
  • 161. The method of claim 158 or 159, wherein said agent enhances the expression of said gene in cells of said cancer.
  • 162. The method of claim 161, wherein said one or more DNA damaging agents are selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation, and wherein said gene is selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 163. The method of claim 161, wherein said one or more DNA damaging agents are selected from the group consisting of doxorubicin, camptothecin, and cisplatin, and wherein said gene is selected from the group consisting of EPHB3, Wee1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 164. The method of claim 163, wherein said agent comprises an siRNA targeting said gene.
  • 165. A method for evaluating sensitivity of a cell to the growth inhibitory effect of an agent, said method comprising determining a transcript level of each of one or more genes in said cell, wherein each said transcript level below a predetermined threshold level for a respective gene indicates that said cell is sensitive to the growth inhibitory effect of said DNA damaging agent.
  • 166. The method of claim 165, wherein said agent is a DNA damaging agent selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation, and wherein said gene is selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 167. The method of claim 165, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin, and wherein said gene is selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 168. The method of any one of claims 166-167, wherein said one or more genes comprises at least about 5 to about 50 different genes.
  • 169. The method of claim 168, wherein each said transcript level is a 1.5-fold, 2-fold or 3-fold reduction from said threshold level.
  • 170. The method of any one of claims 166-167, wherein said transcript level of said gene is determined by a method comprising measuring the transcript level of said gene using one or more polynucleotide probes, each of said one or more polynucleotide probes comprising a nucleotide sequence in said gene.
  • 171. The method of claim 170, wherein said one or more polynucleotide probes are polynucleotide probes on a microarray.
  • 172. A method for evaluating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, said method comprising determining a level of abundance of a protein encoded by a gene in said cell, wherein said level of abundance of said protein below a predetermined threshold level indicates that said cell is sensitive to the growth inhibitory effect of said DNA damaging agent.
  • 173. A method for evaluating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, said method comprising determining a level of activity of a protein encoded by a gene in said cell, wherein said activity level above a predetermined threshold level indicates that said cell is sensitive to the growth inhibitory effect of said DNA damaging agent.
  • 174. The method of claim 172 or 173, wherein said DNA damaging agent is selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation, and wherein said gene is selected from the group consisting of EPHB3, Wee1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 175. The method of claim 174, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin, and wherein said gene is selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 176. The method of claim 172 or 173, wherein said cell is a human cell.
  • 177. A method for regulating sensitivity of a cell to DNA damage, comprising contacting said cell with a sufficient amount of an agent, said agent regulating the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene.
  • 178. The method of claim 177, wherein said DNA damage is caused by a DNA damaging agent.
  • 179. The method of claim 178, wherein said DNA damaging agent is selected from the group consisting of topoisomerase I inhibitor, topoisomerase II inhibitor, DNA binding agent, and ionizing radiation.
  • 180. The method of claim 179, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin.
  • 181. A method for regulating growth of a cell, comprising contacting said cell with i) a sufficient amount of an agent that regulates the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene; and ii) a sufficient amount of a DNA damaging agent.
  • 182. The method of claim 177 or 181, wherein said agent reduces the expression of said gene in said cell.
  • 183. The method of claim 177 or 181, wherein said agent comprises an siRNA targeting said gene.
  • 184. The method of claim 183, wherein said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said gene.
  • 185. The method of claim 184, wherein the total siRNA concentration of said different siRNAs in said agent is an optimal concentration for silencing said gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 186. The method of claim 185, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 187. The method of claim 185, wherein the concentration of each said different siRNA is about the same.
  • 188. The method of claim 185, wherein the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 189. The method of claim 185, wherein none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 190. The method of claim 185, wherein at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs.
  • 191. The method of claim 185, wherein the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, or less than 0.01% of silencing of any off-target genes.
  • 192. A method of identifying an agent that is capable of regulating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, wherein said agent is capable of modulating the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene, said method comprising comparing inhibitory effect of said DNA damaging agent on cells expressing said gene in the presence of said agent with inhibitory effect of said DNA damaging agent on cells expressing said gene in the absence of said agent, wherein a difference in said inhibitory effect of said DNA damaging agent identifies said agent as capable of regulating sensitivity of said cell to the growth inhibitory effect of said DNA damaging agent.
  • 193. A method of identifying an agent that is capable of regulating sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, wherein said agent is capable of modulating the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or activity of a protein encoded by said gene, said method comprising: (a) contacting a first cell expressing said gene with said DNA damaging agent in the presence of said agent and measuring a first growth inhibitory effect; (b) contacting a second cell expressing said gene with said DNA damaging agent in the absence of said agent and measuring a second growth inhibitory effect; and (c) comparing said first and second inhibitory effects measured in said step (a) and (b), wherein a difference between said first and second inhibitory effects identifies said agent as capable of regulating sensitivity of a cell to the growth inhibitory effect of said DNA damaging agent.
  • 194. The method of claim 192 or 193, wherein said cell expresses an siRNA targeting a primary target gene.
  • 195. The method of claim 194, wherein said primary target gene is p53.
  • 196. The method of claim 192 or 193, wherein said agent comprises a molecule that reduces expression of said gene.
  • 197. The method of claim 196, wherein said agent comprises an siRNA targeting said gene.
  • 198. The method of claim 197, wherein said agent comprises 2, 3, 4, 5, 6, or 10 different siRNAs targeting said gene.
  • 199. The method of claim 198, wherein the total siRNA concentration of said different siRNAs in said agent is an optimal concentration for silencing said gene, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing substantially.
  • 200. The method of claim 199, wherein said optimal concentration is a concentration further increase of which does not increase the level of silencing by more than 20%, more than 10%, or more than 5%.
  • 201. The method of claim 199, wherein the concentration of each said different siRNA is about the same.
  • 202. The method of claim 199, wherein the respective concentrations of said different siRNAs are different from each other by less than 50%, less than 20%, or less than 10%.
  • 203. The method of claim 199, wherein none of the siRNAs in said agent has a concentration that is more than 80%, more than 50%, or more than 20% of said total siRNA concentration of said different siRNAs.
  • 204. The method of claim 199, wherein at least one siRNA in said agent has a concentration that is more than 20% or more than 50% of said total siRNA concentration of said different siRNAs.
  • 205. The method of claim 199, wherein the number of different siRNAs and the concentration of each siRNA in said agent is chosen such that said agent causes less than 10%, less than 1%, less than 0.1%, less than 0.01% of silencing of any off-target genes.
  • 206. A cell comprising one or more different small interfering RNAs (siRNAs) targeting a gene selected from the group consisting of EPHB3, Wee1, ELK1, BRCA1, BRCA2, BARD1, and RAD51 in said cell.
  • 207. The cell of claim 206, wherein said one or more different siRNAs comprises 2, 3, 4, 5, 6, or 10 different siRNAs.
  • 208. The cell of claim 206, wherein said cell is a human cell.
  • 209. The cell of claim 208, wherein said cell is a murine cell.
  • 210. A microarray for diagnosing sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, said microarray comprising one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in one or more genes selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 211. A kit for diagnosis of sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, comprising in one or more containers one or more polynucleotide probes, wherein each said polynucleotide probe comprises a nucleotide sequence in a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51.
  • 212. A kit for screening for agents which regulate sensitivity of a cell to the growth inhibitory effect of a DNA damaging agent, comprising in one or more containers (i) the cell of any one of claims 206-211; and (ii) said DNA damaging agent.
  • 213. A kit for treating a mammal having a cancer, comprising in one or more containers (i) a sufficient amount of an agent that regulates the expression of a gene selected from the group consisting of EPHB3, WEE1, ELK1, STK6, BRCA1, BRCA2, BARD1, and RAD51 and/or the activity of a protein encoded by said gene; and (ii) a DNA damaging agent.
  • 214. The method of any one of claims 192-193, wherein said DNA damaging agent is selected from the group consisting of a topoisomerase I inhibitor, a topoisomerase II inhibitor, a DNA binding agent, and ionizing radiation.
  • 215. The method of claim 214, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin.
  • 216. The kit of claim 212, wherein said DNA damaging agent is selected from the group consisting of a topoisomerase I inhibitor, a topoisomerase II inhibitor, a DNA binding agent, and ionizing radiation.
  • 217. The method of claim 216, wherein said DNA damaging agent is selected from the group consisting of doxorubicin, camptothecin, and cisplatin.
  • 218. The method of claim 21, 117, 137 or 138, wherein level of silencing of said primary target gene is controlled.
Parent Case Info

This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Patent Application No. 60/554,284, filed on Mar. 17, 2004, U.S. Provisional Patent Application No. 60/548,568, filed on Feb. 27, 2004, and U.S. Provisional Patent Application No. 60/505,229, filed on Sep. 22, 2003, each of which is incorporated by reference herein in its entirety.

Provisional Applications (3)
Number Date Country
60554284 Mar 2004 US
60548568 Feb 2004 US
60505229 Sep 2003 US