Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Cancer Gene Therapy (2009) 16, 532–538 r 2009 Nature Publishing Group All rights reserved 0929-1903/09 $32.00 www.nature.com/cgt ORIGINAL ARTICLE Allele-specific cancer cell killing in vitro and in vivo targeting a single-nucleotide polymorphism in POLR2A ORF Mook, F Baas, MB de Wissel and K Fluiter Department of Neurogenetics, Academic Medical Center, Amsterdam, The Netherlands Cancer is one of the diseases for which RNA interference is a potential therapeutic approach. Genes involved in the promotion or maintenance of tumor growth are obvious targets for RNAi. RNAi is also considered an attractive additional approach to conventional chemotherapy for cancer treatment. Moreover, siRNAs have shown a high specificity for their molecular target mRNAs as they can selectively inhibit cancer-promoting genes that differ by a point mutation. Loss of heterozygosity (LOH) reduces genes to hemizygosity in cancer cells and presents an absolute difference between normal and cancer cells. The regions of LOH are usually much larger than the tumor suppressor gene, which is lost, and has been shown to contain genes that are essential for cell survival. Single-nucleotide polymorphisms (SNPs) are the most common type of genetic variation in man. SNPs in essential genes that are frequently affected by LOH can be used as a target for a therapy against cancer cells with LOH. We have designed siRNAs against the gene of the large subunit of RNA polymerase II (POLR2A), a gene located in close proximity to the tumor suppressor gene p53, which frequently shows LOH in cancer cells. It is shown in vitro that siRNA can selectively inhibit POLR2A expression dependent on its genotype. Furthermore, cancer cell proliferation and tumor growth inhibition in nude mice was genotype dependent. We conclude that siRNA can be used for genotype-specific inhibition of tumor growth targeting an SNP in POLR2A in vivo. Cancer Gene Therapy (2009) 16, 532–538; doi:10.1038/cgt.2008.104; published online 23 January 2009 Keywords: RNA interference; siRNA; cancer; single nucleotide polymorphism; POLR2A. Introduction RNA interference (RNAi)1–3 was first discovered in Caenorhabditis elegans as a potent gene-silencing mechanism.4 The inhibition of specific gene expression by means of RNAi has also been achieved in mammalian cells by directly introducing short dsRNA (siRNA) into cells, which avoids a response evoked by long dsRNA.5 Cancer is clearly an important potential target for RNAi-based therapies.6 Target molecules usually represent genes that have been shown previously to be relevant or rate limiting for tumor growth, including growth factors and receptors as well as antiapoptotic or downstream signal transduction proteins in cancer cells.7 Often these targeted molecules are not unique for cancer cells thereby limiting the therapeutic window or these molecules appear to be redundant leading to resistance. Therefore, targeting of molecules that are unique in cancer cells may lead to more specific therapeutic interventions. Correspondence: Dr ORF Mook, Department of Neurogenetics, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands. E-mail: [email protected] Received 22 August 2008; revised 13 October 2008; accepted 25 November 2008; published online 23 January 2009 One of the differences between cancer cells and normal cells is that cancer cells have lost large segments of DNA. The loss of large chromosomal regions, or even whole chromosomes, is an early event in the clonal evolution of cancers. Loss of heterozygosity (LOH) can involve 420% of the total genome in certain cancers.8 This irreversible difference between normal and tumor cells forms the basis of an approach for anticancer drug development, called allele-specific inhibition (ASI)9 (reviewed in10). We have previously identified a large number of single-nucleotide polymorphisms (SNPs) in genes that are essential for cell survival and are localized in chromosomal regions often involved in LOH in various cancer types.11 Inhibition of the hemizygous essential gene left in the cancer cells will lead to specific cancer cell death. The heterozygous normal cells will loose maximally half of the expression of the gene by this approach and cell survival is not affected. Classic antisense approaches have shown proof of principle of ASI in vitro9,11 and in vivo.12 However, antisense-based approaches have some specific limitations that can likely be overcome by the use of siRNA. Over the past few years, several studies have reported the selective silencing of mutant alleles resulting from single-nucleotide point mutations by RNAi.2,13–17 In line with that, systematic analysis of a single-nucleotidemismatched target showed that when the character and the position of the mismatch is carefully considered, a Allele-specific inhibition of tumor growth ORF Mook et al siRNA si10C si10T si7C si7T si10CM9 si10TM9 siGFP Antisense sequense 50 -30 GUCUUCGCCGUAGCGCAGCdTdG GUCUUCGCCAUAGCGCAGCdTdG UUCGCCGUAGCGCAGCUGCdAdC UUCGCCAUAGCGCAGCUGCdAdC GUCUUCGCAGUAGCGCAGCdTdG GUCUUCGCAAUAGCGCAGCdTdG GAUGAACUUCAGGGUCAGCdTdT single mismatch can render an siRNA ineffective.18,19 Therefore, the sequence specificity and the high success rate of finding a potent siRNA made it an attractive molecule for ASI. In our approach we used siRNA to target an essential gene (POLR2A) that is present in only one copy in the cancer cell but present as a heterozygous gene in normal cells. Heterozygosity due to a SNP in POLR2A protects normal cells whereas hemizygosity for POLR2A it is lethal for cancer cells. In this study we show in vitro that siRNAs can discriminate between two allelic forms of POLR2A that differ by a single nucleotide in the target sequence. Systemic delivery of unformulated siRNA resulted in genotype-specific growth inhibition in vivo. 533 POLR2A Table 1 Different siRNAs targeting POLR2A 3’ 5’ C G 3’ si10C 5’ Pos. 10 3’ 5’ U A 3’ si10T 3’ C G Pos. 7 si7C 3’ U A Pos. 7 si7T 3’ C G G A Pos. 10/9 siRNA synthesis Six siRNA duplexes (Proligo, Paris, France) targeted to the POLR2A C/T SNP at position 2673 relative to the start codon were designed (Table 1; Figure 1). A siRNA targeting eGFP (siGFP; Table 1) was purchased from MedProbe (Lund, Sweden) and was used as a control for in vivo experiments. In all siRNAs, a two DNA nucleotide overhang was added at the 30 end to stabilize the duplex. Sense and antisense strands were chemically synthesized and obtained annealed. si10CM9 3’ Cell lines The pancreatic cancer cell line MiaPaca II (T genotype) and the prostate cancer cell line 15PC3 (C genotype) were maintained at 37 1C and 5% CO2 by serial passage in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal calf serum, 2 mM L-glutamine, 100 U ml1 penicillin and 100 mg ml1 streptomycin. Transfections Transfections were performed in six-well culture plates with Lipofectamine 2000 as transfection agent. For transfections the siRNA concentrations ranged between 5 and 100 nM. Total RNA was isolated 24 h posttransfection. Protein samples were 72 h posttransfection. Northern blots Cells were harvested in Trizol (Gibco, Breda, The Netherlands) after removing the growth medium 24 h posttransfection. The RNA isolation was according to the manufacturer’s procedure. RNA was denatured using glyoxal and separated on 1% agarose gels, following U G A A 3’ Pos. 10/9 mRNA 5’ 5’ Materials and methods mRNA 5’ 5’ 3’ mRNA 5’ 5’ 3’ mRNA 5’ Pos. 10 5’ 3’ mRNA mRNA si10TM9 5’ Figure 1 Schematic representation of siRNAs and the relative position of the single-nucleotide polymorphism (SNP) in large subunit of RNA polymerase II (POLR2A) that were used in this study. standard protocols. RNA was subsequently transferred to Hybond-N þ membrane (Amersham, Roosendoal, The Netherlands) in 20 SSC. Following transfer the RNA was UV crosslinked and the membrane was baked for 4 h at 80 1C. To detect POLR2A and 28S mRNA, we used the probes previously described.20 Hybridizations and posthybridization washes were according to Church and Gilbert.21 POLR2A mRNA levels were quantified using Aida software version 3.44 (Raytest, Tilburg, The Netherlands) and normalized to the levels of 28S mRNA. Western blots Cell extracts were prepared in lysis buffer (PBS; 1% Triton X-100, 0.01% sodium azide), were subjected to SDS–polycrylamide gel (6%) electrophoresis (SDS–PAGE), and the resolved proteins were transferred electrophoretically to polyvinylidene fluoride (PVDF) membranes (Invitrogen, Breda, The Netherlands). POLR2A was detected with a mouse-anti-POLR2A monoclonal antibody (clone 8WG16; RDI). Elongation factor2a (rabbit-anti-eEF2a; Cell Signaling, Damvers, MA) was used as loading control. Chemiluminescent detection was performed on a LAS3000 (Fuji, Tokyo, Japan) in accordance with the manufacturer’s instructions. POLR2A protein levels were quantified using Aida software Cancer Gene Therapy Allele-specific inhibition of tumor growth ORF Mook et al 534 15PC3 1.50 1.50 0.50 0.75 0.50 si 10 C M oc k 0.00 si 7T 0.00 si 7C 0.25 si 10 T si 10 C M 9 si 10 TM 9 0.25 M oc k 0.75 1.00 si 7T 1.00 1.25 si 7C 1.25 si 10 T si 10 C M 9 si 10 TM 9 POLAR2A/28S 1.75 si 10 C POLAR2A/28S MiaPaca II 1.75 Figure 2 Quantification of densitometric analysis of northern blots of MiaPaca II cells (a) and 15PC3 cells (b) transfected with indicated siRNA targeting large subunit of RNA polymerase II (POLR2A) at concentrations of 25 nM. Bar graphs show the quantification of three independent experiments (mean±s.e.m.) of POLR2A over 28S ribosomal RNA. Ratios are normalized to mock transfection. version 3.44 (Raytest) and normalized to the levels of eEF2a. In vivo model Female NMRI nu/nu mice (8- to 10-weeks old; Charles River, Maastricht, The Netherlands) were injected subcutaneously with both 106 MiaPaca II (right flank) and 106 15PC3 cells (left flank) cells. The cells were injected within 1 h after harvesting by trypsin treatment. Before injection the cells were washed with phosphatebuffered saline (PBS), counted with a hemocytometer and subsequently mixed with Matrigel on ice (Collaborative Biomedical Products, Bedford, MA). At 1 week after tumor cell injection, unformulated si10C, si10T, si10CM9, si10TM9, siGFP (0.15 mg kg1) or saline was administered via low volume (0.2 ml) tail vein injections22,23 twice a week for 3 weeks. From the start of the treatment until killing, tumor growth was recorded three times a week. Tumor volumes were calculated according Meyer et al.24 Data points were fitted with GraphPad Prism using linear regression in order to calculate the tumor growth rates (GraphPad Prism Software Inc., San Diego, CA). Growth rates of tumors in siRNA-treated animals were normalized to the growth rates of tumors in saline treated animals. Aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT) levels in serum were determined using standard diagnostic procedures with the appropriate kits (Roche Diagnostics, Mannheim, Germany). For each treatment, five mice per group were used. All animal experiments were conducted under the institutional guidelines and according to the law; they were sanctioned by the animal ethics committee. Results In vitro genotype-specific inhibition of POLR2A mRNA and decreased POLR2A protein levels is dependent on the relative position of the SNP To test the sequence specificity of siRNA we used the human cancer lines MiaPaca II and 15PC3. These cell Cancer Gene Therapy lines differ in sequence of POLR2A at position c.2673. As POLR2A encodes an essential gene in mammalian cells inhibition of gene expression would result in diminished cell growth. POLR2A mRNA levels, protein levels and growth inhibition were used as readout for sequence specificity. In MiaPaca II cells (T genotype), POLR2A mRNA was downregulated by si10T. Its allelic variant si10C did not show downregulation of POLR2A mRNA. Introduction of one mismatch at ninth position of the siRNA targeting the T genotype (si10TM9) completely abrogated its ability to downregulate POLR2A. POLR2A expression in MiaPaca II cells was also not affected by a siRNA targeting the C genotype with an additional mismatch at ninth position (si10CM9), having two mismatches with this genotype (Figure 2a). We observed sequence-specific POLR2A mRNA knockdown with siRNA at concentration as high as 100 nM (data not shown). Shifting the position of the siRNA so that the SNP was opposite to the seventh position in the siRNA (si7T) resulted in loss of sequence specificity. Both si7T and si7C downregulated POLR2A independent of the c.2673 genotype (Figure 2a). This confirms previous findings that showed that ASI is dependent on the relative position of the mismatch. Transfection of the same set of siRNAs into 15PC3 cells (C genotype) showed similar results. si10C effectively downregulated POLR2A mRNA (Figure 2b). The siRNAs si10T, si10CM9 and si10TM9 were not effective, not even at a concentration of 100 nM, whereas si7C and si7T were both effective in POLR2A mRNA knockdown at all the concentrations used (Figure 2b). These results show that in in vitro experiments siRNA can discriminate between two genotypes that differ only in one nucleotide. Discrimination was observed over a range of siRNA concentrations up to 20 times the effective concentration of 5 nM. However, discrimination was dependent on the relative position of the SNP. POLR2A protein levels in MiaPaca II and 15PC3 cells (Figure 3) correlated with POLR2A mRNA levels. Allele-specific inhibition of tumor growth ORF Mook et al Effect of siRNAs targeting POLR2A on cell growth As POLR2A is an essential component of the transcriptional machinery, downregulation is cytotoxic. Therefore we can easily study whether allele-specific siRNA can mediate specific target knockdown while leaving its allelic variant that differs only for one nucleotide, unaffected. To test the siRNA specificity in a toxicity assay, MiaPaca II and 15PC3 cells were transfected with the same siRNAs against POLR2A and cell survival was observed for 4 days. At 1-day posttranfection there were no obvious differences between the transfections in either cell line, indicating similar seeding and recovery of the cells after plating them. At 5 days posttransfection, MiaPaca II cells (T-genotype) that were transfected with siRNAs that had no effect on POLR2A expression in this cell line (si10C, si10CM9, si10TM9) or mock transfection had grown to confluence (Figure 4). Transfection with siRNAs that knocked down POLR2A in this cell line (si10T, si7C and si7T) resulted in cell death and subsequent detachment from their surface (Figure 4). Likewise, 15PC3 cells (C genotype) transfected with siRNAs that had no effect on POLR2A expression in this cell line (si10T, si10CM9, si10TM9) grew at similar rates as mock transfected cells and all grew confluent in 5 days (Figure 4). siRNAs that MiaPaca II 15PC3 si7T Mock si7C si10TM9 si10T si10CM9 si10C si7T Mock si7C si10TM9 si10T si10CM9 si10C POLR2A eEF2α Figure 3 Representative western blot of three independent experiments of MiaPaca II and 15PC3 cells transfected with the indicated siRNA targeting the large subunit of RNA polymerase II (POLR2A) at a concentration of 25 nM. effectively lowered POLR2A mRNA and protein in 15PC3 cells (si10C, si7C and si7T) also resulted in cell death and detachment from the surface (Figure 4). This demonstrates that the fully matched siRNAs si10C and si10T targeting POLR2A were toxic but more importantly, for siRNAs carrying a single mismatch at position 9 or 10, no toxicity was observed in this time frame. Therefore, si10C and si10T are potential candidates for genotype-specific drugs. Genotype-specific tumor growth inhibition Since the final goal of our approach is the development of an allele-specific cancer drug we tested whether siRNA directed against POLR2A could inhibit growth of tumor xenografts dependent on their genotype. Growth of MiaPaca II xenografts was inhibited in mice treated with systemically administered si10C, si10T and si10TM9. In contrast to the in vitro experiments, a single mismatch of a siRNA with its target mRNA was toxic in vivo (Figure 5a). Animals treated with siRNA with 2 mismatches (si10CM9) and siRNA targeting eGFP showed similar growth rates of the MiaPaca II xenograft as in the saline treated animals (Figure 5a). Introduction of a second mismatch with the T-genotype was necessary to obtain genotype-specific growth inhibition. Likewise, in 15PC3 xenografts tumor growth was inhibited by si10C, si10T and si10CM9 (Figure 5b). No growth inhibition was observed with si10TM9 which has two mismatches with the C-genotype and siRNA targeting eGFP (Figure 5b). This demonstrates that also in 15PC3 xenografts two mismatches were needed to prevent tumor growth inhibition and again a single mismatch is not sufficient to render a siRNA ineffective in vivo. Therefore, si10CM9 and si10TM9 showed ASI of tumor growth (Figures 5a and b). Serum ASAT and ALAT levels, as determined after killing of the animals, did not differ significantly between the treatment groups indicating that Figure 4 Representative images of cultured MiaPaca II and 15PC3 cells tranfected for 5 days with the indicated siRNA targeting large subunit of RNA polymerase II (POLR2A) at a concentration of 25 nM. Cancer Gene Therapy 535 Allele-specific inhibition of tumor growth ORF Mook et al 536 ASI ASI 0.2 Relative growth rate 1.0 0.8 0.6 0.4 0.2 ) P1 0 10 + (p (p M 9 M M M 1 ) 9) (p at ch m G FP 2 M M 1 M M 2 si + 9 (p M 1 in ) P1 0 10 (p M M M 1 ) 9) (p at ch m G FP si e in Sa l e 0.0 0.0 Sa l Relative growth rate 0.4 si10TM9 0.6 1.2 si10T 0.8 si10C si10CM9 1.0 si10C si10TM9 si10T 1.2 si10CM9 15PC3 MiaPaca II Figure 5 In vivo growth inhibition due to siRNA administration (a, b). Selective tumor growth inhibition was observed in MiaPaca II xenografts (a) and 15PC3 xenografts (b). MM indicates the number of mismatches with the particular large subunit of RNA polymerase II (POLR2A) genotype of the tumor. Bar graphs show the mean±s.e.m. of the relative tumor growth rates (n ¼ 5). Allele-specific inhibition (ASI) indicates siRNAs that show genotype-specific tumor growth inhibition. there was no liver toxicity due to administration of the siRNAs (data not shown). Discussion Allele specific inhibition is a promising approach for the treatment of various diseases including cancer.10,13,14,25 Proof of principle for ASI has been shown previously using antisense oligonucleotides (ODNs)9,20 and siRNA16,17 in vitro. In vivo, tumor growth could be inhibited allele specifically using antisense oligonucleotides however only with a very small therapeutic window.12 ASI mediated by intravenously administered chemically synthesized siRNA in vivo has not been shown before. Here we show that ASI by siRNA can be applied both in vitro and in vivo. We show in two genotypically different cells lines that proper base pairing at the ninth and tenth position of the antisense strand of siRNA and its target RNA is important for its function because a mismatch at these positions rendered siRNA directed against POLR2A inactive. This is in line with the observation that RNAi is lost upon introduction of central mismatches (positions 9–11).13–18,25–27 The catalytic site is at that region and apparently loss of proper base pairing at those specific positions results in loss of RNAi. This was strengthened by the finding that the same mismatches (C:A and G:U) at the seventh position in the AS strand were tolerated. However, it has been shown recently that besides the position, the nature of the mismatch is also involved in target recognition.18,19 Unfortunately, these rules do not apply the same to different targets. For example, a C:A Cancer Gene Therapy mismatch at the tenth position did not affect gene silencing,26 resulted in target knockdown of only approximately 25%18 or as shown in our study was not tolerated at all. If this also applies for a G:U mismatch at the tenth position was not investigated in this particular studies. In our study, the introduction of a mismatch at the ninth position resulted in a G:A mismatch. In general, bulky purine–purine mismatches, A:A and G:A, are much less tolerated and therefore render an siRNA inactive.18 Specifically, mutated K-RASV12 was discriminated from its wild-type variant based on a A:G mismatach at position 9.2 This is in agreement with our study that showed loss of RNAi upon the introduction of a G:A mismatch at the ninth position. There are a limited number of exceptions to the central region; C:U mismatch at the sixteenth position,28 G:G mismatch at the seventeenth position for CEACAM6 siRNA22 and a C:T mismatch at the seventh position29 and studied systematically.18,19 We next tested if siRNA could inhibit growth of xenografted tumors in nude mice in a genotype (allele)specific manner. It has recently been shown by us23 and others that low dosages of unmodified siRNA can mediate RNAi in vivo.22,30,31 We show siRNA mediated genotype (allele)-specific inhibition of tumor growth. However, siRNAs carrying one mismatch, either at the ninth or tenth position, with its target retained activity in vivo. Combination of these two mismatches was necessary to render the siRNA ineffective and achieve ASI. It is unclear why in vitro selected genotype-specific siRNA lost its specificity in vivo but the observed effect in vivo must be sequence dependent as siRNA with two mismatches Allele-specific inhibition of tumor growth ORF Mook et al targeting POLR2A and siRNA targeting GFP did not inhibit tumor growth whereas fully matched or one mismatched siRNAs targeting POLR2A did inhibit tumor growth. Mouse and human POLR2A are highly homologous but differ at two bases from the human sequence in the targeted region of the mRNA. Thus, depending of the human genotype, two or three mismatches are present. These mismatches are sufficient prevent POLR2A knockdown in mouse tissues. Xenografts are composed of human cancer cells and mouse stromal cells. Stromal POLR2A is not inhibited and cannot be discriminated from human POLR2A on western or northern blots. It is therefore not possible to determine the human POLR2A levels in xenografts reliably. In conclusion, siRNA-mediated ASI is a potential therapeutic application to inhibit tumor growth in vivo. Abbreviations ASI, allele-specific inhibition; DMEM, Dulbecco’s modified Eagle’s medium; eEF2a, elongation factor-2a; LOH, loss of heterozygosity; ODNs, antisense oligonucleotides; POLR2A, large subunit of RNA polymerase II; PVDF, polyvinylidene fluoride; RNAi, RNA interference; RPA70, replication protein A, 70-kDa subunit; SNPs, single nucleotide polymorphisms; SDS–PAGE, SDS– polycrylamide gel electrophoresis. Acknowledgements This work was supported by the Dutch Cancer Society, project number 2003-2968 and the Stichting Kindergeneeskundig Kanker Onderzoek. References 1 Leung RK, Whittaker PA. RNA interference: from gene silencing to gene-specific therapeutics. Pharmacol Ther 2005; 107: 222–239. 2 Brummelkamp TR, Bernards R, Agami R. Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell 2002; 2: 243–247. 3 Zhang Z, Jiang G, Yang F, Wang J. Knockdown of mutant K-ras expression by adenovirus-mediated siRNA inhibits the in vitro and in vivo growth of lung cancer cells. Cancer Biol Ther 2006; 5: 1481–1486. 4 Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by doublestranded RNA in Caenorhabditis elegans. Nature 1998; 391: 806–811. 5 Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001; 411: 494–498. 6 Kim DH, Rossi JJ. Strategies for silencing human disease using RNA interference. Nat Rev Genet 2007; 8: 173–184. 7 Aigner A. Applications of RNA interference: current state and prospects for siRNA-based strategies in vivo. Appl Microbiol Biotechnol 2007; 76: 9–21. 8 Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature 1998; 396: 643–649. 9 Basilion JP, Schievella AR, Burns E, Rioux P, Olson JC, Monia BP et al. Selective killing of cancer cells based on loss of heterozygosity and normal variation in the human genome: a new paradigm for anticancer drug therapy. Mol Pharmacol 1999; 56: 359–369. 10 Fluiter K, Housman D, Ten Asbroek AL, Baas F. Killing cancer by targeting genes that cancer cells have lost: allelespecific inhibition, a novel approach to the treatment of genetic disorders. Cell Mol Life Sci 2003; 60: 834–843. 11 Ten Asbroek AL, Olsen J, Housman D, Baas F, Stanton Jr V. Genetic variation in mRNA coding sequences of highly conserved genes. Physiol Genomics 2001; 5: 113–118. 12 Fluiter K, ten Asbroek AL, van Groenigen M, Nooij M, Aalders MC, Baas F. Tumor genotype-specific growth inhibition in vivo by antisense oligonucleotides against a polymorphic site of the large subunit of human RNA polymerase II. Cancer Res 2002; 62: 2024–2028. 13 Abdelgany A, Wood M, Beeson D. Allele-specific silencing of a pathogenic mutant acetylcholine receptor subunit by RNA interference. Hum Mol Genet 2003; 12: 2637–2644. 14 Ding H, Schwarz DS, Keene A, Affar el B, Fenton L, Xia X et al. Selective silencing by RNAi of a dominant allele that causes amyotrophic lateral sclerosis. Aging Cell 2003; 2: 209–217. 15 Martinez LA, Naguibneva I, Lehrmann H, Vervisch A, Tchenio T, Lozano G et al. Synthetic small inhibiting RNAs: efficient tools to inactivate oncogenic mutations and restore p53 pathways. Proc Natl Acad Sci USA 2002; 99: 14849–14854. 16 Miller VM, Gouvion CM, Davidson BL, Paulson HL. Targeting Alzheimer’s disease genes with RNA interference: an efficient strategy for silencing mutant alleles. Nucleic Acids Res 2004; 32: 661–668. 17 Miller VM, Xia H, Marrs GL, Gouvion CM, Lee G, Davidson BL et al. Allele-specific silencing of dominant disease genes. Proc Natl Acad Sci USA 2003; 100: 7195–7200. 18 Du Q, Thonberg H, Wang J, Wahlestedt C, Liang Z. A systematic analysis of the silencing effects of an active siRNA at all single-nucleotide mismatched target sites. Nucleic Acids Res 2005; 33: 1671–1677. 19 Schwarz DS, Ding H, Kennington L, Moore JT, Schelter J, Burchard J et al. Designing siRNA that distinguish between genes that differ by a single nucleotide. PLoS Genet 2006; 2: 1307–1318. 20 ten Asbroek AL, Fluiter K, van Groenigen M, Nooij M, Baas F. Polymorphisms in the large subunit of human RNA polymerase II as target for allele-specific inhibition. Nucleic Acids Res 2000; 28: 1133–1138. 21 Church GM, Gilbert W. Genomic sequencing. Proc Natl Acad Sci USA 1984; 81: 1991–1995. 22 Duxbury MS, Matros E, Ito H, Zinner MJ, Ashley SW, Whang EE. Systemic siRNA-mediated gene silencing: a new approach to targeted therapy of cancer. Ann Surg 2004; 240: 667–674; discussion 675–676. 23 Mook OR, Baas F, de Wissel MB, Fluiter K. Evaluation of locked nucleic acid-modified small interfering RNA in vitro and in vivo. Mol Cancer Ther 2007; 6: 833–843. 24 Meyer T, Regenass U, Fabbro D, Alteri E, Rosel J, Muller M et al. A derivative of staurosporine (CGP 41 251) shows selectivity for protein kinase C inhibition and in vitro antiproliferative as well as in vivo anti-tumor activity. Int J Cancer 1989; 43: 851–856. Cancer Gene Therapy 537 Allele-specific inhibition of tumor growth ORF Mook et al 538 25 Kurosawa T, Igarashi S, Nishizawa M, Onodera O. Selective silencing of a mutant transthyretin allele by small interfering RNAs. Biochem Biophys Res Commun 2005; 337: 1012–1018. 26 Dykxhoorn DM, Schlehuber LD, London IM, Lieberman J. Determinants of specific RNA interference-mediated silencing of human beta-globin alleles differing by a single nucleotide polymorphism. Proc Natl Acad Sci USA 2006; 103: 5953–5958. 27 Li Y, Yokota T, Matsumura R, Taira K, Mizusawa H. Sequence-dependent and independent inhibition specific for mutant ataxin-3 by small interfering RNA. Ann Neurol 2004; 56: 124–129. 28 Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE. EphA2: a determinant of malignant cellular behavior and a Cancer Gene Therapy potential therapeutic target in pancreatic adenocarcinoma. Oncogene 2004; 23: 1448–1456. 29 Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE. RNA interference targeting the M2 subunit of ribonucleotide reductase enhances pancreatic adenocarcinoma chemosensitivity to gemcitabine. Oncogene 2004; 23: 1539–1548. 30 Liang Z, Yoon Y, Votaw J, Goodman MM, Williams L, Shim H. Silencing of CXCR4 blocks breast cancer metastasis. Cancer Res 2005; 65: 967–971. 31 Ocker M, Neureiter D, Lueders M, Zopf S, Ganslmayer M, Hahn EG et al. Variants of bcl-2 specific siRNA for silencing antiapoptotic bcl-2 in pancreatic cancer. Gut 2005; 54: 1298–1308.