力达霉素对卵巢癌抑制作用及联合Hsp90抑制剂格尔德霉素及其衍生物的协同作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
细胞中受到外界DNA损伤物质的侵害后会激活自身修复DNA的途径来进行DNA的损伤后修复,这是许多诱导DNA损伤药物发生耐药或剂量升高的重要原因之一。通过降低细胞内部的DNA损伤后修复水平来增敏DNA损伤药物是寻找放疗、化疗增敏剂可行的方法。细胞在接受DNA损伤因素的作用后是否能够存活主要取决于DNA双链断裂程度。ATM/ATR等PIKKs成员在应答DNA损伤、维持基因组稳定性过程中扮演重要的角色,它们传递基因组损伤的信号、阻滞细胞周期进程并且促进DNA修复。ATM主要对DNA双链断裂进行应答,下游最重要的底物是Chk2;ATR对DNA单链断裂、交联或者复制叉进行应答,下游的重要底物有Chk1和BRCA1等。Chk1和Chk2都将细胞阻滞在G2/M期,抑制DNA复制,促进受损的DNA进行修复。
     力达霉素(lidamycin, LDM;又名C-1027)是烯二炔类(enediyne)抗肿瘤抗生素家族的成员之一,通过诱导DNA断裂可以达到强烈的肿瘤细胞杀伤作用,在中国正在进行Ⅱ期临床试验。作为一种DNA损伤药物,力达霉素同样能够激活细胞内的DNA损伤修复通路,这可能是影响其疗效的主要原因之一。Hsp90是人们非常关注的治疗肿瘤的一个靶点,目前有多种Hsp90抑制剂正在进行临床试验,单独的Hsp90抑制剂并不能发挥很好的效果,但研究中发现Hsp90抑制剂与很多种化疗药物联合应用效果良好,能够起到增敏的作用。关于Hsp90抑制剂增敏化疗药物的机制众说纷纭,可能根据化疗药物的不同,Hsp90所抑制的通路所起的作用也不尽相同。其中许多研究证实Hsp90抑制剂可以降低基因组的稳定性。本研究主要研究了Hsp90抑制剂格尔德霉素及其衍生物CNDG与力达霉素联合给药后对力达霉素在人卵巢癌SKOV-3细胞中的增敏作用,观察联合给药后细胞的生长、增殖、DNA损伤水平,探讨Hsp90抑制剂对力达霉素增敏的作用机制。
     Hsp90抑制剂我们选择了典型的格尔德霉素(Geldanamycin, GDM)和由本实验室合成的格尔德霉素衍生物CNDG。
     一、力达霉素对卵巢癌细胞的影响
     MTT检测表明,LDM (0.01nM、0.05nM、0.1nM、0.5nM和0.1nM)作用于SKOV-3细胞24小时后,可以呈剂量依赖性的抑制细胞增殖,细胞凋亡比率随浓度增加而升高。Western blot检测显示0.01nM的力达霉素可以增强HER2的磷酸化水平,而0.1和1 nM的力达霉素降低HER2的磷酸化。在体外模拟缺氧条件用CoC12处理细胞体外模拟肿瘤的缺氧状态,结果显示力达霉素可以明显的降低缺氧诱导因子HIF-1α的水平。
     二、Hsp90抑制剂可以增强力达霉素对卵巢癌SKOV-3、乳腺癌MCF-7、肺癌A-549、肝癌Bel-7402细胞的杀伤作用
     用MTT法检测力达霉素与格尔德霉素(GDM)联合应用对卵巢癌SKOV-3细胞增殖的影响,计算力达霉素(0.01nM、0.05nM)与格尔德霉素(10nM、50nM,100nM、500nM, 100nM)单药及联合后对卵巢癌SKOV-3细胞增殖的抑制作用,并通过公式计算其联合指数,力达霉素与格尔德霉素100nM、500nM联合作用有明显的联合抑制细胞增殖的作用。CNDG与力达霉素联合可显示与格尔德霉素相似的作用。实验表明力达霉素与Hsp90抑制剂联合应用可以明显增强抑制细胞增殖的水平。
     同样的实验条件,我们选择了三种其他种类的肿瘤细胞进行相应的实验,Hsp90抑制剂同样能够增强力达霉素对细胞的杀伤能力,伴随着诱导凋亡的增加、DNA损伤后修复的减少,G2/M期阻滞降低等现象,说明Hsp90抑制剂对力达霉素的增效作用是可以广泛适用于多种肿瘤细胞。
     三、Hsp90抑制剂增强力达霉素对SKOV-3细胞诱导凋亡的作用
     以不同浓度格尔德霉素预处理SKOV-3细胞16小时,力达霉素(0.01nM, 0.05 nM)作用于24 h后,PARP的切割水平在联合应用时大幅度升高;用Annexin V-PI双染检测凋亡细胞,其中0.05 nmol/L LDM作用于SKOV-3细胞凋亡率为14%,200nM格尔德霉素作用于SKOV-3细胞凋亡率为16.1%,两者联合后细胞的凋亡比例为32.2%,单独用药与联合用药相比P<0.01。联合指数0.89。
     四、Hsp90抑制剂增强力达霉素对卵巢癌SKOV-3细胞的DNA双链断裂作用
     力达霉素杀伤肿瘤最主要是通过切割DNA,导致DNA双链断裂。我们借助γH2AX水平来了解细胞内DNA双链断裂的程度,通过Western blot和免疫荧光实验可以看到,γH2AX在力达霉素和格尔德霉素联合应用的情况下细胞内的γH2AX明显增加,尤其是细胞核内的γH2AX水平,说明格尔德霉素的联合应用能够增强力达霉素的DNA断裂能力。
     五、Hsp90抑制剂增强力达霉素对卵巢癌SKOV-3细胞的杀伤作用是通过抑制力达霉素诱导的DNA损伤后修复实现的
     在用0.05 nM力达霉素处理细胞1小时后,细胞内的γH2AX大量增加,说明在此期间细胞内产生大量的DNA双链断裂,但6小时后yH2AX阳性细胞减少或细胞内γH2AX染色程度减弱,细胞内本来存在的双链断裂减少了;而用格尔德霉素预处理的细胞内的γH2AX没有发生大的变化,所以格尔德霉素的预处理可以抑制由力达霉素诱导的DNA损伤后修复。
     六、Hsp90抑制剂可以抑制力达霉素诱导的G2/M期阻滞
     不同剂量力达霉素处理SKOV-3细胞24 h,流式细胞仪检测细胞周期分布变化。结果显示,力达霉素诱导SKOV-3细胞发生G2/M期阻滞,该作用呈剂量相关。随着力达霉素浓度增加,G2/M期细胞比例逐渐增加。
     用不同剂量的格尔德霉素预处理细胞16小时,再用不同剂量的力达霉素处理细胞24小时,用流式细胞仪检测细胞周期分布。在SKOV-3细胞中,0.05 nM力达霉素处理细胞可以使G2/M期细胞从15.28%增加到76.79%,而经过200nM格尔德霉素预处理细胞G2/M期百分比大大下降,从76.79%下降到29.33%。说明格尔德霉素可以抑制力达霉素在SKOV-3细胞中诱导的G2/M期细胞周期阻滞。
     七、Hsp90抑制剂可以降低某些与基因组稳定性相关的蛋白或抑制其活性
     Western blot分析Hsp90抑制剂可以抑制多种与基因组稳定性相关的蛋白水平或抑制其活性,其中包括ATM、ATRIP、BRCA1、RPA70。其中,ATRIP的总蛋白水平的下降可以被蛋白酶体抑制剂MG132的作用所阻断。
     结论:
     本研究首次研究了Hsp90抑制剂对烯二炔类抗生素力达霉素的增敏效果。在卵巢癌SKOV-3细胞中Hsp90抑制剂能够通过抑制DNA损伤后修复增加力达霉素对DNA的断裂作用,从而增强力达霉素的细胞毒作用。同时在其他种类的细胞:人乳腺癌MCF-7、人肺癌A549和人肝癌Bel-7402中也可以看到相类似的作用。总之,Hsp90抑制剂可以降低细胞基因组稳定性,抑制力达霉素所诱导的DNA损伤后修复作用,增加力达霉素对细胞造成的DNA损伤,从而增强其抗肿瘤活性。
Potency of Lidamycin on ovaraian carcinoma suppression and Hsp90 inhibitors Geldanamycin and its analogue CNDG enhance the cytotoxicity of lidamycin
     Tumor resistance to DNA damaging agents is often associated with enhanced DNA repair activity. Reducing DNA damage repair is proved to be an effective way to sensitize cancer cells to DNA damaging agents. Enediyne lidamycin (LDM, also called C-1027) showed extremely potent cytotoxicity by inducing DNA damage and Hsp90 is an interesting target for anticancer treatment recently. In this research we investigated whether Hsp90 inhibitors, geldanamycin (GDM) and its analogue CNDG, could further enhance the potent cytotoxicity of LDM. The survival of cells after DNA double-strand breaks (DSBs) is heavily dependent on the DNA damage repair. Phosphatidylinositol-3 kinase-related kinases (PIKKs) members such as ATM and ATR play pivotal roles in response to DNA damage and maintaining genome integrity. They transduce genomic stress signals to halt cell cycle progression and promote DNA repair. After DNA damaging agent treatment PIKKs is activated to attempt the cell cycle arrest and DNA damage repair. ATM responds mainly to DSBs, whereas ATR is activated by single stranded DNA and stalled DNA replication forks. Further, the activation of ATM and ATR triggers G2/M phase arrest and some DNA repair enzymes. Chk2 can be directly activated by ATM and Chkl by ATR. Both Chkl and Chk2 can arrest cell cycle at G2/M phase, retarding DNA replication and stimulating the repair of damaged DNA.
     Lidamycin (LDM) is an enediyne antitumor antibiotic produced by Streptomyces globisporus C-1027. It is currently being evaluated in phaseⅡclinical trials as a potential chemotherapeutic agent in China. LDM induces DSBs leading potent cytotoxicity and marked inhibition of tumor growth in vivo. As a DNA damaging agent LDM induced DNA damage repair and this might decrease the effect of LDM.
     Hsp90 is important for stabilization and trafficking of tyrosine and serine/threonine kinases that are activated in response to genotoxic stress, including those that are essential for survival of cancer cells. GDM is an ansamycin antibiotic that inhibits HSP90 by binding to the NH2-terminal ATP binding domain, leading to degradation of HSP90 clients. Some Hsp90 inhibitors are in phaseⅠ/Ⅱclinical trials in combination with radiation or other chemotherapeutic agents. Many reports show that Hsp90 inhibitors enhance the effect of DNA damaging agents, and some of them indicate that Hsp90 inhibitor decreases DNA damage repair through inhibiting ATM dependent DNA damage repair.
     In this report Hsp90 inhibitors GDM and its analogue CNDG were found to enhance the anticancer effect of LDM. Our results suggested that GDM pretreatment potentiated the cytotoxity of LDM by decreasing the LDM-induced DNA damage repair. To our knowledge, it is the first report about Hsp90 inhibitor enhancing potent cytotoxicity of LDM or enediyne antitumor antibiotic.
     1. LDM effects on SKOV-3 cells
     By MTT assay, LDM inhibited the proliferation of SKOV-3 cells. Examined with Annexin V-FITC/PI double staining flow cytometry, the apoptotic rates in 0.01 nM and 0.05 nM LDM treated cells were 10.92% and 44.78% respectively; at 0.05 nM, LDM dramatically increased the comet tail; and 0.1 nM of LDM inhibited the phosphorylation of HER2. In the hypoxia condition, LDM reduced the level of HIF-1α.
     2. GDM and its analogue sensitize SKOV-3, MCF-7 and Bel-7402 cells to LDM
     It has been demonstrated that inhibition of Hsp90 could potentiate the efficacy of ionization radiation, antimetabolites and alkylating agents. So we investigated whether inhibition of Hsp90 could potentiate the cytotoxicity of LDM. MTT assays were used on different cancer cells. Cells were exposed to LDM for 24 h after 16 h preincubated with GDM. Under our experimental conditions, low concentrations of LDM (0.01 and 0.05 nM) did not show extensive growth inhibition of SKOV-3 cells. In the following experiments, we chose 0.01 and 0.05 nM LDM for SKOV-3 as appropriate doses to detect the potentiation effects of Hsp90 inhibitor.
     By MTT assay, LDM markedly inhibited cell growth by inhibiting Hsp90. Combined with 100 and 500 nM GDM,0.01 and 0.05 nM LDM induced more growth inhibition than LDM alone. We compared the cytotoxicity of the GDM-LDM combination to the effect of the two agents alone using the median effect method, which determines whether the cytotoxicity for the combination is greater than (CI<1), equal to (CI=1), or less than (CI>1) the additive effect of the individual agents. Under the indicated doses, most of the CI values were less than 1 and some of them were less than 0.7. These results suggested that the combination of LDM and GDM showed a synergistic effect in SKOV-3 cells. Notably,17-(6-cinnamamido-hexylamino)-17-demethoxygel- danamycin (CNDG), a derivate of GDM synthesized in our laboratory had similar protentiation effect on LDM cytotoxicity to cancer cells.
     Under the same condition, we also chose Bel-7402, A549 and MCF-7 cells to check this combination method, and the results were quite similar.
     3. GDMpotentiats the apoptosis-inducing effect of LDM
     Western blot analysis was used to detect the cleavage of PARP, an indicator of caspase-mediated apoptosis. The 89-kDa cleaved fragment of PARP increased dose-dependently in SKOV-3 cells exposed to LDM. Enhanced levels of cleaved PARP appeared after combination treatment.
     To confirm the augmentation of LDM-induced apoptosis by GDM, we performed annexin V/PI staining assay. Consistent with above results, LDM in combination with GDM pretreatment induced more apoptosis than LDM alone. The apoptosis cell ratios were increasing from 14% to 32.2%.
     4. GDM enhances LDM-induced DNA damage
     To examine the effects on DNA damage induced by LDM in the absence and presence of GDM, the phosphorylation of Ser-139 of histone H2AX was compared by immunoflurorescence and Western blot. The treatment of LDM or GDM alone did not increase yH2AX protein, however, the combination of GDM and LDM markedly enhanced yH2AX levels. The figure of yH2AX foci by immunoflurorescence also confirmed the result.
     The alkaline comet assay could provide a direct measure of DSBs. There was no significant DNA damage in SKOV-3 cells exposed to either LDM or GDM alone. Significantly more DNA damage in combination-treated cells versus either LDM or GDM alone treated cells.
     5. GDM inhibits LDM-induced DNA damage repair
     yH2AX levels after were determined 1 h and 6 h LDM treatment. LDM increased the number of yH2AX positive cells. However, the number of yH2AX positive cells was reduced dramatically after 6 h treatment as compared with 1 h treatment, indicating that damaged DNA was partially repaired. The combination of GDM and LDM did not seriously affect the yH2AX positive cell number compared with LDM alone at 1 h treatment. Notably, after 6 h treatment, the percentage of yH2AX positive cells was significantly increased in the combination-treated cells as compared with LDM alone. Time course analysis confirmed that GDM reduced DNA damage repair in LDM-treated cells.
     6. GDM abrogates LDM-induced G2/M arrest and phosphorylations of Chkl and Chk2
     Most of DNA damage agents including LDM induce G2/M phase arrest. Treated by 0.05 nM LDM cells at G2/M phase were increased from 15.28%(control) to 76.79%. At the same condition,200 nM GDM did not induce G2/M arrest significantly. However, the pretreatment of GDM down-regulated the number of G2/M phase cells, and the percent of G2/M phase in LDM-treated cells was decreased from 76.79% to 29.33%. By Western blot analysis the phosphorylations of Chkl and Chk2 induced by LDM were decreased by pretreatment of GDM.
     7. GDM compromised LDM-induced ATM activity, the protein level of ATRIP and DNA damage related proteins
     To address the molecular mechanism involved in GDM mediated inhibition of DSB repair, we focused on LDM-induced ATM and ATR activity. As shown, ATM phosphorylation was increased within 1 h of LDM treatment. By contrast, cells pretreated with GDM had a decrease of phospho-ATM in LDM treated cells. These suggest that GDM interferes with the activation of ATM-mediated DSB repair in response to LDM.
     ATR is also an important kinase for LDM induced DNA damamge response. Previous studies showed that both ATR and ATRIP played an important role in this pathway, and ATRIP could regulate ATR levels. Thus, we checked ATR and ATRIP level of cells treated with GDM. The results showed that ATRIP dramatically decreased after 16 h GDM-treatment, while ATR had no change. We then examined the states of ATM, ATR and ATRIP in SKOV-3 cells treated with 200 nM GDM for 40 h,0.05 nM LDM for 24 h or with combination, that is, LDM was added after 16 h of GDM pretreatment. The total proteins of ATM and ATR remained unchanged after 16 h GDM. After 40 h GDM pretreatment, the total levels of ATM and ATR were decreased observablely.Then the influence of GDM on ATRIP expression was examined for cells in the presence or absence of proteasome inhibitors. For this study, SKOV-3 cells were incubated for 16 h with 200 nM GDM along with 10μM proteasome inhibitor MG132. The cells were lysed and analyzed for ATRIP protein levels. Evidently, incubation of cells with MG132 abolished the decrease of ATRIP protein in SKOV-3 cells induced by GDM treatment. Also, GDM could decrease other proteins that are related with DNA damage response.
     In summary, we found that Hsp90 inhibitors could enhance the potent cytotoxicity of LDM. The mechanism was dependent on, at least in part, increasing DNA damage and depressing cell response to DNA damage. Finally, our data suggests that the combination strategy of Hsp90 inhibitors and LDM may represent a novel and promising approach for ovarian cancer treatment.
引文
[1]Zhang SH, Chen J, Jiang M, Zhen YS. [Lidamycin induces apoptosis of human gastric carcinoma BGC823 cells and inhibits xenograft growth in nude mice]. Yao Xue Xue Bao 2008;43:601-4.
    [2]Huang YH, Shang BY, Zhen YS. Antitumor efficacy of lidamycin on hepatoma and active moiety of its molecule. World J Gastroenterol 2005;11:3980-4.
    [3]Chen J, Ouyang ZG, Zhang SH, Zhen YS. Down-regulation of the nuclear factor-kappaB by lidamycin in association with inducing apoptosis in human pancreatic cancer cells and inhibiting xenograft growth. Oncol Rep 2007;17:1445-51.
    [4]Liu X, Bian C, Ren K, Jin H, Li B, Shao RG. Lidamycin induces marked G2 cell cycle arrest in human colon carcinoma HT-29 cells through activation of p38 MAPK pathway. Oncol Rep 2007;17:597-603.
    [5]Nicolaou KC, Smith AL, Yue EW. Chemistry and biology of natural and designed enediynes. Proc Natl Acad Sci U S A 1993;90:5881-8.
    [6]Hu JL, Xue YC, Xie MY, Zhang R, Otani T, Minami Y, et al. A new macromolecular antitumor antibiotic, C-1027. I. Discovery, taxonomy of producing organism, fermentation and biological activity. J Antibiot (Tokyo) 1988;41:1575-9.
    [7]Otani T, Minami Y, Marunaka T, Zhang R, Xie MY. A new macromolecular antitumor antibiotic, C-1027. II. Isolation and physico-chemical properties. J Antibiot (Tokyo) 1988;41:1580-5.
    [8]Xu YJ, Zhen YS, Goldberg IH. C1027 chromophore, a potent new enediyne antitumor antibiotic, induces sequence-specific double-strand DNA cleavage. Biochemistry 1994;33:5947-54.
    [9]Xu YJ, Xi Z, Zhen YS, Goldberg IH. A single binding mode of activated enediyne C1027 generates two types of double-strand DNA lesions:deuterium isotope-induced shuttling between adjacent nucleotide target sites. Biochemistry 1995;34:12451-60.
    [10]Xu YJ, Xi Z, Zhen YS, Goldberg IH. Mechanism of formation of novel covalent drug.DNA interstrand cross-links and monoadducts by enediyne antitumor antibiotics. Biochemistry 1997;36:14975-84.
    [11]Liu YP, Li QS, Huang YR, Zhou MJ, Liu CX. Pharmacokinetics of C-1027 in mice as determined by TCA-RA method. World J Gastroenterol 2005; 11:717-20.
    [12]Liu XJ, Dai Y, Shang Y, Li Y, Zhen YS. [Inhibitory effect of lidamycin on growth of colon carcinoma 26 and hepatic metastasis in mice]. Ai Zheng 2005;24:641-5.
    [13]Miao Q, Shang B, Ouyang Z, Liu X, Zhen Y. Generation and antitumor effects of an engineered and energized fusion protein VL-LDP-AE composed of single-domain antibody and lidamycin. Sci China C Life Sci 2007;50:447-56.
    [14]Iwamoto T, Hiraku Y, Kojima M, Kawanishi S. Amplification of C1027-induced DNA cleavage and apoptosis by a quinacrine-netropsin hybrid molecule in tumor cell lines. Arch Biochem Biophys 2005;434:232-40.
    [15]Donnelly A, Blagg BS. Novobiocin and additional inhibitors of the Hsp90 C-terminal nucleotide-binding pocket. Curr Med Chem 2008;15:2702-17.
    [16]Peng B, Chang Q, Wang L, Hu Q, Wang Y, Tang J, et al. Suppression of human ovarian SKOV-3 cancer cell growth by Duchesnea phenolic fraction is associated with cell cycle arrest and apoptosis. Gynecol Oncol 2008; 108:173-81.
    [17]Taldone T, Gozman A, Maharaj R, Chiosis G. Targeting Hsp90: small-molecule inhibitors and their clinical development. Curr Opin Pharmacol 2008;8:370-4.
    [18]Powers MV, Workman P. Targeting of multiple signalling pathways by heat shock protein 90 molecular chaperone inhibitors. Endocr Relat Cancer 2006; 13 Suppl 1:S125-35.
    [19]Bae J, Mitsiades C, Tai YT, Bertheau R, Shammas M, Batchu RB, et al. Phenotypic and functional effects of heat shock protein 90 inhibition on dendritic cell. J Immunol 2007; 178:7730-7.
    [20]Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev Cancer 2005;5:761-72.
    [21]Blagosklonny MV. Hsp-90-associated oncoproteins:multiple targets of geldanamycin and its analogs. Leukemia 2002; 16:455-62.
    [22]Wu LX, Xu JH, Zhang KZ, Lin Q, Huang XW, Wen CX, et al. Disruption of the Bcr-Abl/Hsp90 protein complex:a possible mechanism to inhibit Bcr-Abl-positive human leukemic blasts by novobiocin. Leukemia 2008;22:1402-9.
    [23]Solit DB, Basso AD, Olshen AB, Scher HI, Rosen N. Inhibition of heat shock protein 90 function down-regulates Akt kinase and sensitizes tumors to Taxol. Cancer Res 2003;63:2139-44.
    [24]Sawai A, Chandarlapaty S, Greulich H, Gonen M, Ye Q, Arteaga CL, et al. Inhibition of Hsp90 down-regulates mutant epidermal growth factor receptor (EGFR) expression and sensitizes EGFR mutant tumors to paclitaxel. Cancer Res 2008;68:589-96.
    [25]Chandarlapaty S, Sawai A, Ye Q, Scott A, Silinski M, Huang K, et al. SNX2112, a synthetic heat shock protein 90 inhibitor, has potent antitumor activity against HER kinase-dependent cancers. Clin Cancer Res 2008;14:240-8.
    [26]Pedersen NM, Madshus IH, Haslekas C, Stang E. Geldanamycin-induced down-regulation of ErbB2 from the plasma membrane is clathrin dependent but proteasomal activity independent. Mol Cancer Res 2008;6:491-500.
    [27]Xu W, Yuan X, Beebe K, Xiang Z, Neckers L. Loss of Hsp90 association up-regulates Src-dependent ErbB2 activity. Mol Cell Biol 2007;27:220-8.
    [28]Georgakis GV, Li Y, Rassidakis GZ, Martinez-Valdez H, Medeiros LJ, Younes A. Inhibition of heat shock protein 90 function by 17-allylamino-17-demethoxy-geldanamycin in Hodgkin's lymphoma cells down-regulates Akt kinase, dephosphorylates extracellular signal-regulated kinase, and induces cell cycle arrest and cell death. Clin Cancer Res 2006;12:584-90.
    [29]Cao X, Bloomston M, Zhang T, Frankel WL, Jia G, Wang B, et al. Synergistic antipancreatic tumor effect by simultaneously targeting hypoxic cancer cells with HSP90 inhibitor and glycolysis inhibitor. Clin Cancer Res 2008;14:1831-9.
    [30]Kim WY, Oh SH, Woo JK, Hong WK, Lee HY. Targeting heat shock protein 90 overrides the resistance of lung cancer cells by blocking radiation-induced stabilization of hypoxia-inducible factor-lalpha. Cancer Res 2009;69:1624-32.
    [31]Park MA, Zhang G, Mitchell C, Rahmani M, Hamed H, Hagan MP, et al. Mitogen-activated protein kinase kinase 1/2 inhibitors and 17-allylamino-17-demethoxygeldanamycin synergize to kill human gastrointestinal tumor cells in vitro via suppression of c-FLIP-s levels and activation of CD95. Mol Cancer Ther 2008;7:2633-48.
    [32]Kim MS, Kwak HJ, Lee JW, Kim HJ, Park MJ, Park JB, et al. 17-Allylamino-17-demethoxygeldanamycin down-regulates hyaluronic acid-induced glioma invasion by blocking matrix metalloproteinase-9 secretion. Mol Cancer Res2008;6:1657-65.
    [33]Chen J, Wu SY, Ou-Yang ZG, Zhen YS. Synergy of gemcitabine and lidamycin associated with NF-kappaB downregulation in pancreatic carcinoma cells.Acta Pharmacol Sin 2008;29:614-9.
    [34]Gazitt Y, Kolaparthi V, Moncada K, Thomas C, Freeman J. Targeted therapy of human osteosarcoma with 17AAG or rapamycin:Characterization of induced apoptosis and inhibition of mTOR and Akt/MAPK/Wnt pathways. Int J Oncol 2009;34:551-61.
    [35]Schwock J, Dhani N, Cao MP, Zheng J, Clarkson R, Radulovich N, et al. Targeting focal adhesion kinase with dominant-negative FRNK or Hsp90 inhibitor 17-DMAG suppresses tumor growth and metastasis of SiHa cervical xenografts. Cancer Res 2009;69:4750-9.
    [36]Tse AN, Klimstra DS, Gonen M, Shah M, Sheikh T, Sikorski R, et al. A phase 1 dose-escalation study of irinotecan in combination with 17-allylamino-17-demethoxygeldanamycin in patients with solid tumors. Clin Cancer Res2008;14:6704-11.
    [37]Goetz MP, Toft D, Reid J, Ames M, Stensgard B, Safgren S, et al. Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advanced cancer. J Clin Oncol 2005;23:1078-87.
    [38]Heath El, Hillman DW, Vaishampayan U, Sheng S, Sarkar F, Harper F, et al. A phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with hormone-refractory metastatic prostate cancer. Clin Cancer Res 2008; 14:7940-6.
    [39]Banerji U. Heat shock protein 90 as a drug target:some like it hot. Clin Cancer Res 2009;15:9-14.
    [40]Shimamura T, Shapiro GI. Heat shock protein 90 inhibition in lung cancer. J Thorac Oncol 2008;3:S152-9.
    [41]Wu X, Wanders A, Wardega P, Tinge B, Gedda L, Bergstrom S, et al. Hsp90 is expressed and represents a therapeutic target in human oesophageal cancer using the inhibitor 17-allylamino-17-demethoxygeldanamycin. BrJ Cancer 2009; 100:334-43.
    [42]Reck M, von Pawel J, Zatloukal P, Ramlau R, Gorbounova V, Hirsh V, et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer:AVAil. J Clin Oncol 2009;27:1227-34.
    [43]Koll TT, Feis SS, Wright MH, Teniola MM, Richardson MM, Robles AI, et al. HSP90 inhibitor, DMAG, synergizes with radiation of lung cancer cells by interfering with base excision and ATM-mediated DNA repair. Mol Cancer Ther 2008;7:1985-92.
    [44]Dote H, Burgan WE, Camphausen K, Tofilon PJ. Inhibition of hsp90 compromises the DNA damage response to radiation. Cancer Res 2006;66:9211-20.
    [45]Quanz M, Berthault N, Roulin C, Roy M, Herbette A, Agrario C, et al. Small-molecule drugs mimicking DNA damage:a new strategy for sensitizing tumors to radiotherapy. Clin Cancer Res 2009; 15:1308-16.
    [46]Dziegielewski J, Beerman TA. Cellular responses to the DNA strand-scission enediyne C-1027 can be independent of ATM, ATR, and DNA-PK kinases. J Biol Chem 2002;277:20549-54.
    [47]Kennedy DR, Beerman TA. The radiomimetic enediyne C-1027 induces unusual DNA damage responses to double-strand breaks. Biochemistry 2006;45:3747-54.
    [48]Kennedy DR, Ju J, Shen B, Beerman TA. Designer enediynes generate DNA breaks, interstrand cross-links, or both, with concomitant changes in the regulation of DNA damage responses. Proc Natl Acad Sci U S A 2007; 104:17632-7.
    [49]Wakeling AE, Guy SP, Woodburn JR, Ashton SE, Curry BJ, Barker AJ, et al. ZD1839 (Iressa):an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res 2002;62:5749-54.
    [50]Mai Z, Blackburn GL, Zhou JR. Soy phytochemicals synergistically enhance the preventive effect of tamoxifen on the growth of estrogen-dependent human breast carcinoma in mice. Carcinogenesis 2007;28:1217-23.
    [51]Romanelli S, Perego P, Pratesi G, Carenini N, Tortoreto M, Zunino F. In vitro and in vivo interaction between cisplatin and topotecan in ovarian carcinoma systems. Cancer Chemother Pharmacol 1998;41:385-90.
    [52]Duthie SJ, Collins AR, Duthie GG, Dobson VL. Quercetin and myricetin protect against hydrogen peroxide-induced DNA damage (strand breaks and oxidised pyrimidines) in human lymphocytes. Mutat Res 1997;393:223-31.
    [53]Collins AR, Dobson VL, Dusinska M, Kennedy G, Stetina R. The comet assay:what can it really tell us? Mutat Res 1997;375:183-93.
    [54]Collins A, Dusinska M, Franklin M, Somorovska M, Petrovska H, Duthie S, et al. Comet assay in human biomonitoring studies:reliability, validation, and applications. Environ Mol Mutagen 1997;30:139-46.
    [55]Zhang J, Willers H, Feng Z, Ghosh JC, Kim S, Weaver DT, et al. Chk2 phosphorylation of BRCA1 regulates DNA double-strand break repair. Mol Cell Biol 2004;24:708-18.
    [56]Liu X, He H, Feng Y, Zhang M, Ren K, Shao R. Difference of cell cycle arrests induced by lidamycin in human breast cancer cells. Anticancer Drugs 2006;17:173-9.
    [57]Clingen PH, Wu JY, Miller J, Mistry N, Chin F, Wynne P, et al. Histone H2AX phosphorylation as a molecular pharmacological marker for DNA interstrand crosslink cancer chemotherapy. Biochem Pharmacol 2008;76:19-27.
    [58]Economopoulou M, Langer HF, Celeste A, Orlova VV, Choi EY, Ma M, et al. Histone H2AX is integral to hypoxia-driven neovascularization. Nat Med 2009;15:553-8.
    [59]Giuliano P, De Cristofaro T, Affaitati A, Pizzulo GM, Feliciello A, Criscuolo C, et al. DNA damage induced by polyglutamine-expanded proteins. Hum Mol Genet 2003;12:2301-9.
    [60]Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem 1998;273:5858-68.
    [61]Kinner A, Wu W, Staudt C, Iliakis G. Gamma-H2AX in recognition and signaling of DNA double-strand breaks in the context of chromatin. Nucleic Acids Res 2008;36:5678-94.
    [62]Hurley PJ, Bunz F. ATM and ATR:components of an integrated circuit. Cell Cycle 2007;6:414-7.
    [63]Bakkenist CJ, Kastan MB. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 2003;421:499-506.
    [64]Takai H, Tominaga K, Motoyama N, Minamishima YA, Nagahama H, Tsukiyama T, et al. Aberrant cell cycle checkpoint function and early embryonic death in Chkl(-/-) mice. Genes Dev 2000; 14:1439-47.
    [65]Jin J, Ang XL, Ye X, Livingstone M, Harper JW. Differential roles for checkpoint kinases in DNA damage-dependent degradation of the Cdc25A protein phosphatase. J Biol Chem 2008;283:19322-8.
    [66]Yan Y, Black CP, Cao PT, Haferbier JL, Kolb RH, Spieker RS, et al. Gamma-irradiation-induced DNA damage checkpoint activation involves feedback regulation between extracellular signal-regulated kinase 1/2 and BRCA1. Cancer Res 2008;68:5113-21.
    [67]Zajac M, Moneo MV, Carnero A, Benitez J, Martinez-Delgado B. Mitotic catastrophe cell death induced by heat shock protein 90 inhibitor in BRCA1-deficient breast cancer cell lines. Mol Cancer Ther 2008;7:2358-66.
    [68]Wolf I, Golan T, Shani A, Aderka D. Cetuximab in metastatic colorectal cancer. Lancet Oncol 11:313-4; author reply 4.
    [69]Ibrahim EM, Zekri JM, Bin Sadiq BM. Cetuximab-based therapy for metastatic colorectal cancer:a meta-analysis of the effect of K-ras mutations. Int J Colorectal Dis.
    [70]Lilenbaum RC. The evolving role of cetuximab in non-small cell lung cancer. Clin Cancer Res 2006;12:4432s-5s.
    [71]Liu H, Li L, Li XQ, Liu XJ, Zhen YS. Enediyne lidamycin enhances the effect of epidermal growth factor receptor tyrosine kinase inhibitor, gefitinib, in epidermoid carcinoma A431 cells and lung carcinoma H460 cells. Anticancer Drugs 2009;20:41-9.
    [72]Beerman TA, Gawron LS, Shin S, Shen B, McHugh MM. C-1027, a radiomimetic enediyne anticancer drug, preferentially targets hypoxic cells. Cancer Res 2009;69:593-8.
    [73]Gudmundsdottir K, Ashworth A. The roles of BRCA1 and BRCA2 and associated proteins in the maintenance of genomic stability. Oncogene 2006;25:5864-74.
    [74]Smith E, Dejsuphong D, Balestrini A, Hampel M, Lenz C, Takeda S, et al. An ATM-and ATR-dependent checkpoint inactivates spindle assembly by targeting CEP63. Nat Cell Biol 2009; 11:278-85.
    [75]Garner E, Costanzo V. Studying the DNA damage response using in vitro model systems. DNA Repair (Amst) 2009;8:1025-37.
    [76]Moran DM, Gawlak G, Jayaprakash MS, Mayar S, Maki CG. Geldanamycin promotes premature mitotic entry and micronucleation in irradiated p53/p21 deficient colon carcinoma cells. Oncogene 2008.
    [77]Sugimoto K, Sasaki M, Isobe Y, Tsutsui M, Suto H, Ando J, et al. Hsp90-inhibitor geldanamycin abrogates G2 arrest in p53-negative leukemia cell lines through the depletion of Chkl. Oncogene 2008;27:3091-101.
    [78]Alcazar A, Cid C. High cytotoxic sensitivity of the oligodendrocyte precursor cells to HSP90 inhibitors in cell cultures. Exp Neurol 2009.
    [79]Kobayashi S, Nantz R, Kitamura T, Higashikubo R, Horikoshi N. Combined inhibition of extracellular signal-regulated kinases and HSP90 sensitizes human colon carcinoma cells to ionizing radiation. Oncogene 2005;24:3011-9.
    [80]Koopman G, Reutelingsperger CP, Kuijten GA, Keehnen RM, Pals ST, van Oers MH. Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood 1994;84:1415-20.
    [81]Li L, Liu H, Xu XD, Zhen YS. A novel derivative of geldanamycin and its antitumor activity. Chin Chem Lett 2009;15:391-2.
    [82]Arlander SJ, Eapen AK, Vroman BT, McDonald RJ, Toft DO, Karnitz LM. Hsp90 inhibition depletes Chkl and sensitizes tumor cells to replication stress. J Biol Chem 2003;278:52572-7.
    [83]Zou L, Elledge SJ. Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes. Science 2003;300:1542-8.
    [84]Fanning E, Klimovich V, Nager AR. A dynamic model for replication protein A (RPA) function in DNA processing pathways. Nucleic Acids Res 2006;34:4126-37.
    [85]Cimprich KA, Cortez D. ATR:an essential regulator of genome integrity. Nat Rev Mol Cell Biol 2008;9:616-27.
    [86]Kitagawa R, Bakkenist CJ, McKinnon PJ, Kastan MB. Phosphorylation of SMC1 is a critical downstream event in the ATM-NBS1-BRCA1 pathway. Genes Dev 2004;18:1423-38.
    [87]Uziel T, Lerenthal Y, Moyal L, Andegeko Y, Mittelman L, Shiloh Y Requirement of the MRN complex for ATM activation by DNA damage. EMBO J 2003;22:5612-21.
    [88]Carson CT, Schwartz RA, Stracker TH, Lilley CE, Lee DV, Weitzman MD. The Mrell complex is required for ATM activation and the G2/M checkpoint. EMBO J 2003;22:6610-20.
    [89]Tomimatsu N, Mukherjee B, Burma S. Distinct roles of ATR and DNA-PKcs in triggering DNA damage responses in ATM-deficient cells. EMBO Rep 2009;10:629-35.
    [90]Falck J, Coates J, Jackson SP. Conserved modes of recruitment of ATM, ATR and DNA-PKcs to sites of DNA damage. Nature 2005;434:605-11.
    [91]Williams RS, Williams JS, Tainer JA. Mrell-Rad50-Nbsl is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem Cell Biol 2007;85:509-20.
    [92]Arnaudeau C, Lundin C, Helleday T. DNA double-strand breaks associated with replication forks are predominantly repaired by homologous recombination involving an exchange mechanism in mammalian cells. J Mol Biol 2001;307:1235-45.
    [93]Olson E, Nievera CJ, Lee AY, Chen L, Wu X. The Mrell-Rad50-Nbsl complex acts both upstream and downstream of ataxia telangiectasia mutated and Rad3-related protein (ATR) to regulate the S-phase checkpoint following UV treatment. J Biol Chem 2007;282:22939-52.
    [94]Zhou J, Lim CU, Li JJ, Cai L, Zhang Y. The role of NBS1 in the modulation of PIKK family proteins ATM and ATR in the cellular response to DNA damage. Cancer Lett 2006;243:9-15.
    [95]Gatei M, Sloper K, Sorensen C, Syljuasen R, Falck J, Hobson K, et al. Ataxia-telangiectasia-mutated (ATM) and NBS1-dependent phosphorylation of Chkl on Ser-317 in response to ionizing radiation. J Biol Chem 2003;278:14806-11.
    [96]Lee JS, Collins KM, Brown AL, Lee CH, Chung JH. hCdsl-mediated phosphorylation of BRCA1 regulates the DNA damage response. Nature 2000;404:201-4.
    [97]Santarosa M, Del Col L, Tonin E, Caragnano A, Viel A, Maestro R. Premature senescence is a major response to DNA cross-linking agents in BRCA1-defective cells:implication for tailored treatments of BRCA1 mutation carriers. Mol Cancer Ther 2009;8:844-54.
    [98]McGuire V, Whittemore AS, Norris R, Oakley-Girvan 1. Survival in epithelial ovarian cancer patients with prior breast cancer. Am J Epidemiol 2000;152:528-32.
    [99]Umezu K, Sugawara N, Chen C, Haber JE, Kolodner RD. Genetic analysis of yeast RPA1 reveals its multiple functions in DNA metabolism. Genetics 1998;148:989-1005.
    [100]Lee SE, Moore JK, Holmes A, Umezu K, Kolodner RD, Haber JE. Saccharomyces Ku70, mre11/rad50 and RPA proteins regulate adaptation to G2/M arrest after DNA damage. Cell 1998;94:399-409.
    [101]Costanzo V, Shechter D, Lupardus PJ, Cimprich KA, Gottesman M, Gautier J. An ATR-and Cdc7-dependent DNA damage checkpoint that inhibits initiation of DNA replication. Mol Cell 2003;11:203-13.
    [102]McHugh MM, Yin X, Kuo SR, Liu JS, Melendy T, Beerman TA. The cellular response to DNA damage induced by the enediynes C-1027 and neocarzinostatin includes hyperphosphorylation and increased nuclear retention of replication protein a (RPA) and trans inhibition of DNA replication. Biochemistry 2001;40:4792-9.
    [103]Liu JS, Kuo SR, Yin X, Beerman TA, Melendy T. DNA damage by the enediyne C-1027 results in the inhibition of DNA replication by loss of replication protein A function and activation of DNA-dependent protein kinase. Biochemistry 2001;40:14661-8.
    [104]Mordes DA, Cortez D. Activation of ATR and related PIKKs. Cell Cycle 2008;7:2809-12.
    [105]Bomgarden RD, Yean D, Yee MC, Cimprich KA. A novel protein activity mediates DNA binding of an ATR-ATRIP complex. J Biol Chem 2004;279:13346-53.
    [106]Itakura E, Sawada I, Matsuura A. Dimerization of the ATRIP protein through the coiled-coil motif and its implication to the maintenance of stalled replication forks. Mol Biol Cell 2005; 16:5551-62.
    [107]Itakura E, Takai KK, Umeda K, Kimura M, Ohsumi M, Tamai K, et al. Amino-terminal domain of ATRIP contributes to intranuclear relocation of the ATR-ATRIP complex following DNA damage. FEBS Lett 2004;577:289-93.
    [108]Ball HL, Cortez D. ATRIP oligomerization is required for ATR-dependent checkpoint signaling. J Biol Chem 2005;280:31390-6.
    [109]Ball HL, Ehrhardt MR, Mordes DA, Glick GG, Chazin WJ, Cortez D. Function of a conserved checkpoint recruitment domain in ATRIP proteins. Mol Cell Biol 2007;27:3367-77.
    [110]Pabla N, Huang S, Mi QS, Daniel R, Dong Z. ATR-Chk2 signaling in p53 activation and DNA damage response during cisplatin-induced apoptosis. J Biol Chem 2008;283:6572-83.
    [111]Itakura E, Umeda K, Sekoguchi E, Takata H, Ohsumi M, Matsuura A. ATR-dependent phosphorylation of ATRIP in response to genotoxic stress. Biochem Biophys Res Commun 2004;323:1197-202.
    [112]Cortez D, Guntuku S, Qin J, Elledge SJ. ATR and ATRIP:partners in checkpoint signaling. Science 2001;294:1713-6.
    [113]Toledo LI, Murga M, Gutierrez-Martinez P, Soria R, Fernandez-Capetillo O. ATR signaling can drive cells into senescence in the absence of DNA breaks. Genes Dev 2008;22:297-302.
    [114]Mordes DA, Glick GG, Zhao R, Cortez D. TopBP1 activates ATR through ATRIP and a PIKK regulatory domain. Genes Dev 2008;22:1478-89.
    [115]Sokka M, Parkkinen S, Pospiech H, Syvaoja JE. Function of TopBPl in Genome Stability. Subcell Biochem 50:119-41.
    [116]Olayioye MA, Neve RM, Lane HA, Hynes NE. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J 2000; 19:3159-67.
    [117]Baselga J, Arteaga CL. Critical update and emerging trends in epidermal growth factor receptor targeting in cancer. J Clin Oncol 2005;23:2445-59.
    [118]Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol 2001;2:127-37.
    [119]Giannopoulou E, Antonacopoulou A, Floratou K, Papavassiliou AG, Kalofonos HP. Dual targeting of EGFR and HER-2 in colon cancer cell lines. Cancer Chemother Pharmacol 2009;63:973-81.
    [120]Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL, Human breast cancer:correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;235:177-82.
    [121]Tiwari RK, Borgen PI, Wong GY, Cordon-Cardo C, Osborne MP. HER-2/neu amplification and overexpression in primary human breast cancer is associated with early metastasis. Anticancer Res 1992; 12:419-25.
    [122]Carr JA, Havstad S, Zarbo RJ, Divine G, Mackowiak P, Velanovich V. The association of HER-2/neu amplification with breast cancer recurrence. Arch Surg 2000;135:1469-74.
    [123]Yunxia Z, Jun C, Guanshan Z, Yachao L, Xueke Z, Jin L. Mutations in epidermal growth factor receptor and K-ras in Chinese patients with colorectal cancer. BMC Med Genet 11:34.
    [124]Giovannetti E, Zucali PA, Peters GJ, Cortesi F, D'Incecco A, Smit EF, et al. Association of polymorphisms in AKT1 and EGFR with clinical outcome and toxicity in non-small cell lung cancer patients treated with gefitinib. Mol Cancer Ther 9:581-93.
    [1]Powers MV, Workman P. Targeting of multiple signalling pathways by heat shock protein 90 molecular chaperone inhibitors. Endocr Relat Cancer 2006; 13 Suppl 1:S125-35.
    [2]Taldone T, Gozman A, Maharaj R, Chiosis G. Targeting Hsp90:small-molecule inhibitors and their clinical development. Curr Opin Pharmacol 2008;8:370-4.
    [3]Chandarlapaty S, Sawai A, Ye Q, Scott A, Silinski M, Huang K, et al. SNX2112, a synthetic heat shock protein 90 inhibitor, has potent antitumor activity against HER kinase-dependent cancers. Clin Cancer Res 2008;14:240-8.
    [4]Okawa Y, Hideshima T, Steed P, Vallet S, Hall S, Huang K, et al. SNX-2112, a selective Hsp90 inhibitor, potently inhibits tumor cell growth, angiogenesis, and osteoclastogenesis in multiple myeloma and other hematological tumors by abrogating signaling via Akt and ERK. Blood 2008.
    [5]Soti C, Racz A, Csermely P. A Nucleotide-dependent molecular switch controls ATP binding at the C-terminal domain of Hsp90. N-terminal nucleotide binding unmasks a C-terminal binding pocket. J Biol Chem 2002;277:7066-75.
    [6]Bae J, Mitsiades C, Tai YT, Bertheau R, Shammas M, Batchu RB, et al. Phenotypic and functional effects of heat shock protein 90 inhibition on dendritic cell. J Immunol 2007; 178:7730-7.
    [7]Wu LX, Xu JH, Zhang KZ, Lin Q, Huang XW, Wen CX, et al. Disruption of the Bcr-Abl/Hsp90 protein complex:a possible mechanism to inhibit Bcr-Abl-positive human leukemic blasts by novobiocin. Leukemia 2008;22:1402-9.
    [8]Solit DB, Basso AD, Olshen AB, Scher HI, Rosen N. Inhibition of heat shock protein 90 function down-regulates Akt kinase and sensitizes tumors to Taxol. Cancer Res 2003;63:2139-44.
    [9]Sawai A, Chandarlapaty S, Greulich H, Gonen M, Ye Q, Arteaga CL, et al. Inhibition of Hsp90 down-regulates mutant epidermal growth factor receptor (EGFR) expression and sensitizes EGFR mutant tumors to paclitaxel. Cancer Res 2008;68:589-96.
    [10]Pedersen NM, Madshus IH, Haslekas C, Stang E. Geldanamycin-induced down-regulation of ErbB2 from the plasma membrane is clathrin dependent but proteasomal activity independent. Mol Cancer Res 2008;6:491-500.
    [11]Xu W, Yuan X, Beebe K, Xiang Z, Neckers L. Loss of Hsp90 association up-regulates Src-dependent ErbB2 activity. Mol Cell Biol 2007;27:220-8.
    [12]Georgakis GV, Li Y, Rassidakis GZ, Martinez-Valdez H, Medeiros LJ, Younes A. Inhibition of heat shock protein 90 function by 17-allylamino-17-demethoxy-geldanamycin in Hodgkin's lymphoma cells down-regulates Akt kinase, dephosphorylates extracellular signal-regulated kinase, and induces cell cycle arrest and cell death. Clin Cancer Res 2006;12:584-90.
    [13]Cao X, Bloomston M, Zhang T, Frankel WL, Jia G, Wang B, et al. Synergistic antipancreatic tumor effect by simultaneously targeting hypoxic cancer cells with HSP90 inhibitor and glycolysis inhibitor. Clin Cancer Res 2008;14:1831-9.
    [14]Kim WY, Oh SH, Woo JK, Hong WK, Lee HY. Targeting heat shock protein 90 overrides the resistance of lung cancer cells by blocking radiation-induced stabilization of hypoxia-inducible factor-1 alpha. Cancer Res 2009;69:1624-32.
    [15]Park MA, Zhang G, Mitchell C, Rahmani M, Hamed H, Hagan MP, et al. Mitogen-activated protein kinase kinase 1/2 inhibitors and 17-allylamino-17-demethoxygeldanamycin synergize to kill human gastrointestinal tumor cells in vitro via suppression of c-FLIP-s levels and activation of CD95. Mol Cancer Ther 2008;7:2633-48.
    [16]Kim MS, Kwak HJ, Lee JW, Kim HJ, Park MJ, Park JB, et al. 17-Allylamino-17-demethoxygeldanamycin down-regulates hyaluronic acid-induced glioma invasion by blocking matrix metalloproteinase-9 secretion. Mol Cancer Res 2008;6:1657-65.
    [17]Chen J, Wu SY, Ou-Yang ZG, Zhen YS. Synergy of gemcitabine and lidamycin associated with NF-kappaB downregulation in pancreatic carcinoma cells. Acta Pharmacol Sin 2008;29:614-9.
    [18]Gazitt Y, Kolaparthi V, Moncada K, Thomas C, Freeman J. Targeted therapy of human osteosarcoma with 17AAG or rapamycin:Characterization of induced apoptosis and inhibition of mTOR and Akt/MAPK/Wnt pathways. Int J Oncol 2009;34:551-61.
    [19]Schwock J, Dhani N, Cao MP, Zheng J, Clarkson R, Radulovich N, et al. Targeting focal adhesion kinase with dominant-negative FRNK or Hsp90 inhibitor 17-DMAG suppresses tumor growth and metastasis of SiHa cervical xenografts. Cancer Res 2009;69:4750-9.
    [20]Heath El, Hillman DW, Vaishampayan U, Sheng S, Sarkar F, Harper F, et al. A phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with hormone-refractory metastatic prostate cancer. Clin Cancer Res 2008; 14:7940-6.
    [21]Solit DB, Osman I, Polsky D, Panageas KS, Daud A, Goydos JS, et al. Phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with metastatic melanoma. Clin Cancer Res 2008;14:8302-7.
    [22]Dote H, Burgan WE, Camphausen K, Tofilon PJ. Inhibition of hsp90 compromises the DNA damage response to radiation. Cancer Res 2006;66:9211-20.
    [23]McCollum AK, Lukasiewicz KB, Teneyck CJ, Lingle WL, Toft DO, Erlichman C. Cisplatin abrogates the geldanamycin-induced heat shock response. Mol Cancer Ther 2008;7:3256-64.
    [24]Mesa RA, Loegering D, Powell HL, Flatten K, Arlander SJ, Dai NT, et al. Heat shock protein 90 inhibition sensitizes acute myelogenous leukemia cells to cytarabine. Blood 2005;106:318-27.
    [25]Wetzler M, Earp JC, Brady MT, Keng MK, Jusko WJ. Synergism between arsenic trioxide and heat shock protein 90 inhibitors on signal transducer and activator of transcription protein 3 activity-pharmacodynamic drug-drug interaction modeling. Clin Cancer Res 2007; 13:2261-70.
    [26]Pelicano H, Carew JS, McQueen TJ, Andreeff M, Plunkett W, Keating MJ, et al. Targeting Hsp90 by 17-AAG in leukemia cells:mechanisms for synergistic and antagonistic drug combinations with arsenic trioxide and Ara-C. Leukemia 2006;20:610-9.
    [27]Wu X, Wanders A, Wardega P, Tinge B, Gedda L, Bergstrom S, et al. Hsp90 is expressed and represents a therapeutic target in human oesophageal cancer using the inhibitor 17-allylamino-17-demethoxygeldanamycin. Br J Cancer 2009; 100:334-43.
    [28]Koll TT, Feis SS, Wright MH, Teniola MM, Richardson MM, Robles Al, et al. HSP90 inhibitor, DMAG, synergizes with radiation of lung cancer cells by interfering with base excision and ATM-mediated DNA repair. Mol Cancer Ther 2008;7:1985-92.
    [29]Kobayashi S, Nantz R, Kitamura T, Higashikubo R, Horikoshi N. Combined inhibition of extracellular signal-regulated kinases and HSP90 sensitizes human colon carcinoma cells to ionizing radiation. Oncogene 2005;24:3011-9.
    [30]McCollum AK, Teneyck CJ, Sauer BM, Toft DO, Erlichman C. Up-regulation of heat shock protein 27 induces resistance to 17-allylamino-demethoxygeldanamycin through a glutathione-mediated mechanism. Cancer Res 2006;66:10967-75.
    [31]Pabla N, Huang S, Mi QS, Daniel R, Dong Z. ATR-Chk2 signaling in p53 activation and DNA damage response during cisplatin-induced apoptosis. J Biol Chem 2008;283:6572-83.
    [32]Moran DM, Gawlak G, Jayaprakash MS, Mayar S, Maki CG. Geldanamycin promotes premature mitotic entry and micronucleation in irradiated p53/p21 deficient colon carcinoma cells. Oncogene 2008.
    [33]Sugimoto K, Sasaki M, Isobe Y, Tsutsui M, Suto H, Ando J, et al. Hsp90-inhibitor geldanamycin abrogates G2 arrest in p53-negative leukemia cell lines through the depletion of Chkl. Oncogene 2008;27:3091-101.
    [34]Alcazar A, Cid C. High cytotoxic sensitivity of the oligodendrocyte precursor cells to HSP90 inhibitors in cell cultures. Exp Neurol 2009.
    [1]Ikegami S, Taguchi T, Ohashi M, Oguro M, Nagano H, Mano Y. Aphidicolin prevents mitotic cell division by interfering with the activity of DNA polymerase-alpha. Nature 1978;275:458-60.
    [2]Bianchi V, Pontis E, Reichard P. Changes of deoxyribonucleoside triphosphate pools induced by hydroxyurea and their relation to DNA synthesis. J Biol Chem 1986;261:16037-42.
    [3]Lundin C, Erixon K, Arnaudeau C, Schultz N, Jenssen D, Meuth M, et al. Different roles for nonhomologous end joining and homologous recombination following replication arrest in mammalian cells. Mol Cell Biol 2002;22:5869-78.
    [4]Saintigny Y, Delacote F, Vares G, Petitot F, Lambert S, Averbeck D, et al. Characterization of homologous recombination induced by replication inhibition in mammalian cells. EMBO J 2001;20:3861-70.
    [5]Shiloh Y. ATM and related protein kinases:safeguarding genome integrity. Nat Rev Cancer 2003;3:155-68.
    [6]Kitagawa R, Bakkenist CJ, McKinnon PJ, Kastan MB. Phosphorylation of SMC1 is a critical downstream event in the ATM-NBS1-BRCA1 pathway. Genes Dev 2004;18:1423-38.
    [7]Uziel T, Lerenthal Y, Moyal L, Andegeko Y, Mittelman L, Shiloh Y. Requirement of the MRN complex for ATM activation by DNA damage. EMBO J 2003;22:5612-21.
    [8]Carson CT, Schwartz RA, Stracker TH, Lilley CE, Lee DV, Weitzman MD. The Mrell complex is required for ATM activation and the G2/M checkpoint. EMBO J 2003;22:6610-20.
    [9]Horejsi Z, Falck J, Bakkenist CJ, Kastan MB, Lukas J, Bartek J. Distinct functional domains of Nbsl modulate the timing and magnitude of ATM activation after low doses of ionizing radiation. Oncogene 2004;23:3122-7.
    [10]Cortez D, Guntuku S, Qin J, Elledge SJ. ATR and ATRIP:partners in checkpoint signaling. Science 2001;294:1713-6.
    [11]Brown EJ, Baltimore D. Essential and dispensable roles of ATR in cell cycle arrest and genome maintenance. Genes Dev 2003;17:615-28.
    [12]Brown EJ, Baltimore D. ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev 2000; 14:397-402.
    [13]O'Driscoll M, Ruiz-Perez VL, Woods CG, Jeggo PA, Goodship JA. A splicing mutation affecting expression of ataxia-telangiectasia and Rad3-related protein (ATR) results in Seckel syndrome. Nat Genet 2003;33:497-501.
    [14]Itakura E, Umeda K, Sekoguchi E, Takata H, Ohsumi M, Matsuura A. ATR-dependent phosphorylation of ATRIP in response to genotoxic stress. Biochem Biophys Res Commun 2004;323:1197-202.
    [15]Ball HL, Myers JS, Cortez D. ATRIP binding to replication protein A-single-stranded DNA promotes ATR-ATRIP localization but is dispensable for Chkl phosphorylation. Mol Biol Cell 2005; 16:2372-81.
    [16]Zou L, Elledge SJ. Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes. Science 2003;300:1542-8.
    [17]Venere M, Snyder A, Zgheib O, Halazonetis TD. Phosphorylation of ATR-interacting protein on Ser239 mediates an interaction with breast-ovarian cancer susceptibility 1 and checkpoint function. Cancer Res 2007;67:6100-5.
    [18]Myers JS, Zhao R, Xu X, Ham AJ, Cortez D. Cyclin-dependent kinase 2 dependent phosphorylation of ATRIP regulates the G2-M checkpoint response to DNA damage. Cancer Res 2007;67:6685-90.
    [19]Kerzendorfer C, O'Driscoll M. UVB and caffeine:inhibiting the DNA damage response to protect against the adverse effects of UVB. J Invest Dermatol 2009;129:1611-3.
    [20]Ward IM, Minn K, Chen J. UV-induced ataxia-telangiectasia-mutated and Rad3-related (ATR) activation requires replication stress. J Biol Chem 2004;279:9677-80.
    [21]Yan Y, Black CP, Cao PT, Haferbier JL, Kolb RH, Spieker RS, et al. Gamma-irradiation-induced DNA damage checkpoint activation involves feedback regulation between extracellular signal-regulated kinase 1/2 and BRCA1. Cancer Res 2008;68:5113-21.
    [22]Umezu K, Sugawara N, Chen C, Haber JE, Kolodner RD. Genetic analysis of yeast RPA1 reveals its multiple functions in DNA metabolism. Genetics 1998;148:989-1005.
    [23]Lee SE, Moore JK, Holmes A, Umezu K, Kolodner RD, Haber JE. Saccharomyces Ku70, mre11/rad50 and RPA proteins regulate adaptation to G2/M arrest after DNA damage. Cell 1998;94:399-409.
    [24]Costanzo V, Shechter D, Lupardus PJ, Cimprich KA, Gottesman M, Gautier J. An ATR-and Cdc7-dependent DNA damage checkpoint that inhibits initiation of DNA replication. Mol Cell 2003;11:203-13.
    [25]Dasso M, Newport JW. Completion of DNA replication is monitored by a feedback system that controls the initiation of mitosis in vitro:studies in Xenopus. Cell 1990;61:811-23.
    [26]Dasso M, Smythe C, Milarski K, Kornbluth S, Newport JW. DNA replication and progression through the cell cycle. Ciba Found Symp 1992; 170:161-80; discussion 80-6.
    [27]Newport J, Dasso M. On the coupling between DNA replication and mitosis. J Cell Sci Suppl 1989; 12:149-60.
    [28]Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 2001; 15:2177-96.
    [29]Alderton GK, Joenje H, Varon R, Borglum AD, Jeggo PA, O'Driscoll M. Seckel syndrome exhibits cellular features demonstrating defects in the ATR-signalling pathway. Hum Mol Genet 2004; 13:3127-38.
    [30]Shiloh Y. ATM and ATR:networking cellular responses to DNA damage. Curr Opin Genet Dev 2001; 11:71-7.
    [31]Costanzo V, Gautier J. Single-strand DNA gaps trigger an ATR-and Cdc7-dependent checkpoint. Cell Cycle 2003;2:17.
    [32]Lindsey-Boltz LA, Bermudez VP, Hurwitz J, Sancar A. Purification and characterization of human DNA damage checkpoint Rad complexes. Proc Natl Acad Sci U S A 2001;98:11236-41.
    [33]Zou L, Liu D, Elledge SJ. Replication protein A-mediated recruitment and activation of Rad17 complexes. Proc Natl Acad Sci U S A 2003;100:13827-32.
    [34]Delacroix S, Wagner JM, Kobayashi M, Yamamoto K, Karnitz LM. The Rad9-Husl-Radl (9-1-1) clamp activates checkpoint signaling via TopBP1. Genes Dev 2007;21:1472-7.
    [35]Lee J, Kumagai A, Dunphy WG. The Rad9-Husl-Radl checkpoint clamp regulates interaction of TopBP1 with ATR. J Biol Chem 2007;282:28036-44.
    [36]Kumagai A, Lee J, Yoo HY, Dunphy WG. TopBP1 activates the ATR-ATRIP complex. Cell 2006;124:943-55.
    [37]Toledo LI, Murga M, Gutierrez-Martinez P, Soria R, Fernandez-Capetillo O. ATR signaling can drive cells into senescence in the absence of DNA breaks. Genes Dev 2008;22:297-302.
    [38]Mordes DA, Glick GG, Zhao R, Cortez D. TopBP1 activates ATR through ATRIP and a PIKK regulatory domain. Genes Dev 2008;22:1478-89.
    [39]Sanchez Y, Wong C, Thoma RS, Richman R, Wu Z, Piwnica-Worms H, et al. Conservation of the Chkl checkpoint pathway in mammals:linkage of DNA damage to Cdk regulation through Cdc25. Science 1997;277:1497-501.
    [40]Furnari B, Rhind N, Russell P. Cdc25 mitotic inducer targeted by chkl DNA damage checkpoint kinase. Science 1997;277:1495-7.
    [41]Shechter D, Costanzo V, Gautier J. ATR and ATM regulate the timing of DNA replication origin firing. Nat Cell Biol 2004;6:648-55.
    [42]Kumagai A, Dunphy WG. Claspin, a novel protein required for the activation of Chkl during a DNA replication checkpoint response in Xenopus egg extracts. Mol Cell 2000;6:839-49.
    [43]Kumagai A, Dunphy WG. Repeated phosphopeptide motifs in Claspin mediate the regulated binding of Chkl. Nat Cell Biol 2003;5:161-5.
    [44]Lee J, Kumagai A, Dunphy WG. Claspin, a Chkl-regulatory protein, monitors DNA replication on chromatin independently of RPA, ATR, and Rad 17. Mol Cell 2003; 11:329-40.
    [45]Hashimoto Y, Tsujimura T, Sugino A, Takisawa H. The phosphorylated C-terminal domain of Xenopus Cut5 directly mediates ATR-dependent activation of Chk1. Genes Cells 2006;11:993-1007.
    [46]Rotman G, Shiloh Y. ATM:a mediator of multiple responses to genotoxic stress. Oncogene 1999;18:6135-44.
    [47]Bartek J, Lukas J. DNA repair:Damage alert. Nature 2003;421:486-8.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700