首页 > 最新文献

Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response最新文献

英文 中文
Electrochemical behavior of Methotrexate upon binding to the DNA of different cell lines 甲氨蝶呤与不同细胞系DNA结合后的电化学行为
Douglas Vieira Thomaz, P.H.N. Santos
{"title":"Electrochemical behavior of Methotrexate upon binding to the DNA of different cell lines","authors":"Douglas Vieira Thomaz, P.H.N. Santos","doi":"10.3390/iecc2021-09215","DOIUrl":"https://doi.org/10.3390/iecc2021-09215","url":null,"abstract":"","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89483837","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Exploiting DNA Repair Defect in Triple Negative Brest Cancer Using CDK Inhibition Strategy 利用CDK抑制策略研究三阴性乳腺癌DNA修复缺陷
C. Velázquez, Esin Orhan, I. Tabet, Lise Fenou, Laura Boudarel, C. Theillet
Triple-negative breast cancer (TNBC), representing 15% of breast carcinomas, is an aggressive breast cancer subtype with a high probability of metastasis and limited treatment options. Noticeably, BRCA-deficiency occurs in 25% of the TNBCs and results in deficient homologous recombination (HR) repair. Interestingly, PARP inhibitors (PARPi) have shown synthetic lethality in a BRCA-deficient context, however, their efficacy is frequently hampered by intrinsic or acquired resistance mechanisms involving restoration of the HR. In that regard, the role of some CDKs proven to regulate key HR actors was of interest to us.We aim at understanding the rewiring pathways determining resistance to PARPi in BRCA-deficient cancers and assessing the role of transcriptional regulating CDKs such as CDK7, CDK9 or CDK12 in the transcriptional regulation of key HR genes. Our ultimate goal is to determine whether and which CDK inhibitors could be effective approaches to repress HR gene expression and induce pharmacological HR-deficiency. As such, these CDK-inhibitors could be molecules of choice allowing sensitization of tumors that would otherwise respond poorly to DNA damaging treatment. With this purpose, we use in vitro and in vivo (PDX) models of TNBC and study the attenuation of HR response in tumor cells and PDX models treated with CDK-inhibitors. Our final aim is to determine the most efficient combination CDK-I + PARP-I. Our HR read outs are RAD51 and BRCA1 foci formation upon PARP-I treatment. We also measure the modification of RNA and protein expression levels induced by CDK-I treatment on a series of diagnostic HR genes (BRCA2, PALB2, ATR, FAND2), as a measure of HR repression. We will present data comparing the relative efficiency of 3 CDK-I, dinaciclib, NVP-2 and SR-4835, which have different specificities and inhibit different CDKs with variable efficacy.
三阴性乳腺癌(TNBC)占乳腺癌的15%,是一种侵袭性乳腺癌亚型,转移概率高,治疗选择有限。值得注意的是,brca缺陷发生在25%的tnbc中,并导致同源重组(HR)修复缺陷。有趣的是,PARP抑制剂(PARPi)在brca缺乏的情况下显示出合成致死性,然而,它们的效力经常受到涉及HR恢复的内在或获得性耐药机制的阻碍。在这方面,我们对一些CDKs的作用感兴趣,这些CDKs被证明可以调节关键的人力资源参与者。我们的目标是了解brca缺陷癌症中决定PARPi耐药性的重布线途径,并评估转录调节cdk(如CDK7、CDK9或CDK12)在关键HR基因转录调节中的作用。我们的最终目标是确定CDK抑制剂是否以及哪些可以有效抑制HR基因表达并诱导药理学上的HR缺乏。因此,这些cdk抑制剂可能是使肿瘤敏感化的首选分子,否则这些肿瘤对DNA损伤治疗反应不佳。为此,我们使用TNBC的体外和体内(PDX)模型,研究cdk抑制剂治疗肿瘤细胞和PDX模型中HR反应的衰减。我们的最终目标是确定CDK-I + PARP-I的最有效组合。我们的HR读数是PARP-I治疗后RAD51和BRCA1灶的形成。我们还测量了CDK-I治疗对一系列HR诊断基因(BRCA2, PALB2, ATR, FAND2)诱导的RNA和蛋白质表达水平的修饰,作为HR抑制的测量。我们将提供数据比较3种CDK-I、dinaciclib、NVP-2和SR-4835的相对效率,它们具有不同的特异性,抑制不同CDKs的效果也不同。
{"title":"Exploiting DNA Repair Defect in Triple Negative Brest Cancer Using CDK Inhibition Strategy","authors":"C. Velázquez, Esin Orhan, I. Tabet, Lise Fenou, Laura Boudarel, C. Theillet","doi":"10.3390/iecc2021-09212","DOIUrl":"https://doi.org/10.3390/iecc2021-09212","url":null,"abstract":"Triple-negative breast cancer (TNBC), representing 15% of breast carcinomas, is an aggressive breast cancer subtype with a high probability of metastasis and limited treatment options. Noticeably, BRCA-deficiency occurs in 25% of the TNBCs and results in deficient homologous recombination (HR) repair. Interestingly, PARP inhibitors (PARPi) have shown synthetic lethality in a BRCA-deficient context, however, their efficacy is frequently hampered by intrinsic or acquired resistance mechanisms involving restoration of the HR. In that regard, the role of some CDKs proven to regulate key HR actors was of interest to us.We aim at understanding the rewiring pathways determining resistance to PARPi in BRCA-deficient cancers and assessing the role of transcriptional regulating CDKs such as CDK7, CDK9 or CDK12 in the transcriptional regulation of key HR genes. Our ultimate goal is to determine whether and which CDK inhibitors could be effective approaches to repress HR gene expression and induce pharmacological HR-deficiency. As such, these CDK-inhibitors could be molecules of choice allowing sensitization of tumors that would otherwise respond poorly to DNA damaging treatment. With this purpose, we use in vitro and in vivo (PDX) models of TNBC and study the attenuation of HR response in tumor cells and PDX models treated with CDK-inhibitors. Our final aim is to determine the most efficient combination CDK-I + PARP-I. Our HR read outs are RAD51 and BRCA1 foci formation upon PARP-I treatment. We also measure the modification of RNA and protein expression levels induced by CDK-I treatment on a series of diagnostic HR genes (BRCA2, PALB2, ATR, FAND2), as a measure of HR repression. We will present data comparing the relative efficiency of 3 CDK-I, dinaciclib, NVP-2 and SR-4835, which have different specificities and inhibit different CDKs with variable efficacy.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"81044586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The roles of cell cycle and BRCA1 in the DNA damage response 细胞周期和BRCA1在DNA损伤反应中的作用
Shihori Tanabe, S. Quader, R. Ono, H. Cabral, K. Aoyagi, A. Hirose, H. Yokozaki, H. Sasaki
Molecular network activation states alter dynamically in biology and diseases. In cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT) networks play an important role to acquisition of the drug resistance and cancer malignant feature. To reveal the network pathways in EMT and CSCs, gene expression in diffuse- and intestinal-type gastric cancer (GC) have been analyzed. The several canonical pathways have been found to be altered in diffuse- and intestinal-type GC. Canonical pathway on Cell Cycle: G1/S Checkpoint Regulation was activated in diffuse-type GC, and Cyclins and Cell Cycle Regulation was activated in intestinal-type GC. In Cell Cycle: G1/S Checkpoint Regulation, DNA damage induces p53, which was predicted to be activated in diffuse-type GC. Canonical pathway related to Role of BRCA1 in DNA Damage Response was activated in intestinal-type GC, where BRCA1 which is related to G1/S phase transition was up-regulated. Cell cycle regulation may be altered in EMT condition in diffuse-type GC.
分子网络激活状态在生物学和疾病中是动态变化的。在肿瘤干细胞(CSCs)中,上皮-间质转化(EMT)网络在耐药和肿瘤恶性特征的获得中起着重要作用。为了揭示EMT和CSCs的网络通路,我们分析了弥漫型和肠型胃癌(GC)中的基因表达。已经发现弥漫性和肠型GC的几个典型途径发生了改变。细胞周期的典型途径:弥漫性GC激活G1/S检查点调节,肠型GC激活Cyclins和细胞周期调节。在细胞周期:G1/S检查点调节中,DNA损伤诱导p53,预计在弥漫性GC中p53被激活。在肠型GC中,与BRCA1在DNA损伤反应中作用相关的典型通路被激活,与G1/S相变相关的BRCA1上调。弥漫性GC在EMT条件下可能改变细胞周期调控。
{"title":"The roles of cell cycle and BRCA1 in the DNA damage response","authors":"Shihori Tanabe, S. Quader, R. Ono, H. Cabral, K. Aoyagi, A. Hirose, H. Yokozaki, H. Sasaki","doi":"10.3390/iecc2021-09193","DOIUrl":"https://doi.org/10.3390/iecc2021-09193","url":null,"abstract":"Molecular network activation states alter dynamically in biology and diseases. In cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT) networks play an important role to acquisition of the drug resistance and cancer malignant feature. To reveal the network pathways in EMT and CSCs, gene expression in diffuse- and intestinal-type gastric cancer (GC) have been analyzed. The several canonical pathways have been found to be altered in diffuse- and intestinal-type GC. Canonical pathway on Cell Cycle: G1/S Checkpoint Regulation was activated in diffuse-type GC, and Cyclins and Cell Cycle Regulation was activated in intestinal-type GC. In Cell Cycle: G1/S Checkpoint Regulation, DNA damage induces p53, which was predicted to be activated in diffuse-type GC. Canonical pathway related to Role of BRCA1 in DNA Damage Response was activated in intestinal-type GC, where BRCA1 which is related to G1/S phase transition was up-regulated. Cell cycle regulation may be altered in EMT condition in diffuse-type GC.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87848464","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pre-clinical investigation of inhibition of the DNA damage response as a targetted therapy in myeloproliferative neoplasms shows synergism of ATR inhibitors with standard-of-care treatment. 抑制DNA损伤反应作为骨髓增殖性肿瘤靶向治疗的临床前研究显示,ATR抑制剂与标准治疗具有协同作用。
Aleksander Ślusarczyk, H. Bryant, E. Chen, I. Hitchcock, M. Zeidler, A. Chantry, Sally Thomas
Introduction Myeloproliferative neoplasms (MPNs) are a group of haematological malignancies arising from haematopoietic stem cells with acquired driver mutations in JAK2, MPL and CALR. Increased replication stress is seen in the presence of JAK2V617F. Genes involved in the DNA double-strand break (DSB) repair pathways – BRCA-dependent homologous recombination repair (HRR) and DNA-dependent protein kinase-mediated non-homologous end-joining (D-NHEJ) are upregulated in MPN cells expressing mutated JAK2. Aims Using JAK2V617F and CALR (del 52) mutant cell lines: Determine the effect of single-agent DNA damage repair (DDR) inhibitors on cell viability and apoptosis. Evaluate the efficacy of DDR inhibitors in combination with hydroxyurea or ruxolitinib. Materials and Methods Cell lines expressing JAK2 (V617F)- HEL and CALR (del52)- MARIMO were treated with a drug panel comprising hydroxyurea, ruxolitinib, methotrexate, AZD6738 (ATRi), NU7441 (DNA-PKi), Olaparib (PARPi) and VE-821 (ATRi). AlamarBlue assay for cell proliferation and annexin V/ propidium iodide staining for flow cytometry were used to evaluate the toxicity. Results In JAK2 and CALR mutated cell lines, ATR inhibition by AZD6738 or VE-821, DNA-PKs inhibition by NU7441 and hydroxyurea each reduced viability, whereas PARP inhibition by olaparib had a minimal effect. The combination of ATR inhibition and hydroxyurea demonstrated high synergism in both apoptosis induction and proliferation arrest. Ruxolitinib alone had a modest effect in the presence of JAK2V617F and a minimal effect in CALR (del 52) mutated cells. Synergistic toxicity was observed for ruxolitinib and AZD6738/ VE-821 combination in JAK2 mutated cell line. Conclusions DDR inhibition reduces viability in cells expressing the driver mutations seen in MPNs. Most notably, ATR kinase inhibitors have a synergistic effect with the current standard-of-care treatment hydroxyurea. This study provides preliminary evidence that ATR inhibitors combined with standard therapies may be exploited in MPNs harbouring JAK2 and CALR mutations.
骨髓增生性肿瘤(mpn)是一组由造血干细胞引起的血液系统恶性肿瘤,这些造血干细胞具有JAK2、MPL和CALR的获得性驱动突变。JAK2V617F的存在增加了复制应激。参与DNA双链断裂(DSB)修复途径的基因——brca依赖性同源重组修复(HRR)和DNA依赖性蛋白激酶介导的非同源末端连接(D-NHEJ)在表达突变JAK2的MPN细胞中上调。目的利用JAK2V617F和CALR (del 52)突变细胞系,研究单剂DNA损伤修复(DDR)抑制剂对细胞活力和凋亡的影响。评价DDR抑制剂与羟基脲或鲁索利替尼联合使用的疗效。材料和方法用羟基脲、鲁索替尼、甲氨蝶呤、AZD6738 (ATRi)、NU7441 (DNA-PKi)、奥拉帕尼(PARPi)和VE-821 (ATRi)组成的药物组处理表达JAK2 (V617F)- HEL和CALR (del52)- MARIMO的细胞株。用AlamarBlue法检测细胞增殖,用流式细胞术检测膜联蛋白V/碘化丙啶染色。结果在JAK2和CALR突变细胞系中,AZD6738或VE-821对ATR的抑制、NU7441和羟基脲对DNA-PKs的抑制均降低了细胞活力,而奥拉帕尼对PARP的抑制作用最小。ATR抑制与羟基脲联合使用在诱导细胞凋亡和抑制细胞增殖方面均表现出较高的协同作用。单独Ruxolitinib在JAK2V617F存在时具有适度的作用,在CALR (del 52)突变细胞中具有最小的作用。ruxolitinib与AZD6738/ VE-821联合使用对JAK2突变细胞系有协同毒性作用。结论DDR抑制降低了mpn中表达驱动突变的细胞的活力。最值得注意的是,ATR激酶抑制剂与目前的标准治疗羟基脲具有协同作用。这项研究提供了初步证据,表明ATR抑制剂联合标准疗法可能用于携带JAK2和CALR突变的mpn。
{"title":"Pre-clinical investigation of inhibition of the DNA damage response as a targetted therapy in myeloproliferative neoplasms shows synergism of ATR inhibitors with standard-of-care treatment.","authors":"Aleksander Ślusarczyk, H. Bryant, E. Chen, I. Hitchcock, M. Zeidler, A. Chantry, Sally Thomas","doi":"10.3390/iecc2021-09209","DOIUrl":"https://doi.org/10.3390/iecc2021-09209","url":null,"abstract":"Introduction \u0000Myeloproliferative neoplasms (MPNs) are a group of haematological malignancies arising from haematopoietic stem cells with acquired driver mutations in JAK2, MPL and CALR. Increased replication stress is seen in the presence of JAK2V617F. Genes involved in the DNA double-strand break (DSB) repair pathways – BRCA-dependent homologous recombination repair (HRR) and DNA-dependent protein kinase-mediated non-homologous end-joining (D-NHEJ) are upregulated in MPN cells expressing mutated JAK2. \u0000Aims \u0000Using JAK2V617F and CALR (del 52) mutant cell lines: \u0000 \u0000Determine the effect of single-agent DNA damage repair (DDR) inhibitors on cell viability and apoptosis. \u0000Evaluate the efficacy of DDR inhibitors in combination with hydroxyurea or ruxolitinib. \u0000 \u0000Materials and Methods \u0000Cell lines expressing JAK2 (V617F)- HEL and CALR (del52)- MARIMO were treated with a drug panel comprising hydroxyurea, ruxolitinib, methotrexate, AZD6738 (ATRi), NU7441 (DNA-PKi), Olaparib (PARPi) and VE-821 (ATRi). AlamarBlue assay for cell proliferation and annexin V/ propidium iodide staining for flow cytometry were used to evaluate the toxicity. \u0000Results \u0000In JAK2 and CALR mutated cell lines, ATR inhibition by AZD6738 or VE-821, DNA-PKs inhibition by NU7441 and hydroxyurea each reduced viability, whereas PARP inhibition by olaparib had a minimal effect. The combination of ATR inhibition and hydroxyurea demonstrated high synergism in both apoptosis induction and proliferation arrest. Ruxolitinib alone had a modest effect in the presence of JAK2V617F and a minimal effect in CALR (del 52) mutated cells. Synergistic toxicity was observed for ruxolitinib and AZD6738/ VE-821 combination in JAK2 mutated cell line. \u0000Conclusions \u0000DDR inhibition reduces viability in cells expressing the driver mutations seen in MPNs. Most notably, ATR kinase inhibitors have a synergistic effect with the current standard-of-care treatment hydroxyurea. This study provides preliminary evidence that ATR inhibitors combined with standard therapies may be exploited in MPNs harbouring JAK2 and CALR mutations.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78057014","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
G-quadruplexes and DNA damage in colorectal cancer tumorigenesis 结直肠癌肿瘤发生中的g -四联体与DNA损伤
V. Sánchez-Martín
{"title":"G-quadruplexes and DNA damage in colorectal cancer tumorigenesis","authors":"V. Sánchez-Martín","doi":"10.3390/iecc2021-09198","DOIUrl":"https://doi.org/10.3390/iecc2021-09198","url":null,"abstract":"","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88808227","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of genetic determinants of DNA mismatch repair loss that predict response to immune checkpoint blockade 鉴定DNA错配修复损失的遗传决定因素,预测对免疫检查点封锁的反应
Charlotte Smith, D. Cucchi, Amy Gibson, Kirsten Brooksbank, S. Martin
Despite showing great clinical promise, response rates to immune checkpoint blockade (ICB) vary greatly and biomarkers of response are lacking. A recent Phase II clinical trial in patients with deficiency in the DNA mismatch repair (MMR) pathway indicated that MMR status predicted clinical benefit with the PD-1 inhibitor, pembrolizumab. These findings have led to the first tissue-agnostic approval for anti PD-1 therapy for unresectable or metastatic solid tumours with MMR deficiency. However, it is becoming increasingly clear that many MMR-deficient tumours fail to respond to ICBs with ~50% refractory to treatment. Furthermore, there is a wide diversity of clinical benefit among responders. However why this is the case and how this can be clinically translated remains largely unknown. Our exciting preliminary data suggest that loss of specific MMR genes results in a differential increased expression of the immune checkpoint molecule, PD-L1. Significantly, we observed an upregulation of PD-L1 expression in cells silenced for the MMR genes, MLH1, MSH2, PMS2 and MSH3, as expected. However, we did not observe an increased expression of PD-L1 upon MSH6 loss. This differential expression amongst MMR gene loss was further validated at both RNA and cell surface level. Upon investigation of the molecular mechanism regulating PD-L1 expression after MMR loss, we observed that that phosphorylation of STAT1 positively correlates with PD-L1 expression whilst STAT3 phosphorylation was negatively correlated, such that increased STAT1 phosphorylation was observed upon MLH1 and PMS2 loss and not in MSH6-deficient cells whilst STAT3 phosphorylation was only observed upon MSH6 loss. Significantly, inhibition of STAT3, both pharmacologically and genetically, reinstated PD-L1 expression in MSH6-deficient cells. Therefore, we have evidence that loss of specific MMR genes can trigger differential expression of PD-L1 through a STAT1/STAT3 mediated pathway and we hypothesize that it is this differential expression that may in part determine sensitivity to treatment with ICB.
尽管具有很大的临床前景,但免疫检查点阻断(ICB)的应答率差异很大,而且缺乏应答的生物标志物。最近一项针对DNA错配修复(MMR)途径缺陷患者的II期临床试验表明,MMR状态预示着PD-1抑制剂派姆单抗的临床益处。这些发现导致了抗PD-1治疗MMR缺乏症不可切除或转移性实体瘤的首次组织不可知批准。然而,越来越清楚的是,许多mmr缺陷肿瘤对ICBs没有反应,约50%的肿瘤难以治疗。此外,应答者的临床获益差异很大。然而,为什么会出现这种情况,以及如何将其临床转化,在很大程度上仍然未知。我们激动人心的初步数据表明,特异性MMR基因的缺失导致免疫检查点分子PD-L1表达的差异增加。值得注意的是,我们观察到在MMR基因MLH1、MSH2、PMS2和MSH3沉默的细胞中PD-L1表达上调,正如预期的那样。然而,我们没有观察到MSH6缺失后PD-L1的表达增加。在RNA和细胞表面水平上进一步验证了MMR基因缺失之间的差异表达。通过研究MMR缺失后调控PD-L1表达的分子机制,我们发现STAT1的磷酸化与PD-L1的表达呈正相关,而STAT3的磷酸化与PD-L1的表达呈负相关,因此在MLH1和PMS2缺失的细胞中,STAT1的磷酸化增加,而在MSH6缺失的细胞中没有,而STAT3的磷酸化仅在MSH6缺失的细胞中观察到。值得注意的是,在药理学和遗传学上,抑制STAT3可以恢复msh6缺陷细胞中PD-L1的表达。因此,我们有证据表明特异性MMR基因的缺失可以通过STAT1/STAT3介导的途径触发PD-L1的差异表达,我们假设正是这种差异表达可能在一定程度上决定了对ICB治疗的敏感性。
{"title":"Identification of genetic determinants of DNA mismatch repair loss that predict response to immune checkpoint blockade","authors":"Charlotte Smith, D. Cucchi, Amy Gibson, Kirsten Brooksbank, S. Martin","doi":"10.3390/iecc2021-09195","DOIUrl":"https://doi.org/10.3390/iecc2021-09195","url":null,"abstract":"Despite showing great clinical promise, response rates to immune checkpoint blockade (ICB) vary greatly and biomarkers of response are lacking. A recent Phase II clinical trial in patients with deficiency in the DNA mismatch repair (MMR) pathway indicated that MMR status predicted clinical benefit with the PD-1 inhibitor, pembrolizumab. These findings have led to the first tissue-agnostic approval for anti PD-1 therapy for unresectable or metastatic solid tumours with MMR deficiency. However, it is becoming increasingly clear that many MMR-deficient tumours fail to respond to ICBs with ~50% refractory to treatment. Furthermore, there is a wide diversity of clinical benefit among responders. However why this is the case and how this can be clinically translated remains largely unknown. \u0000Our exciting preliminary data suggest that loss of specific MMR genes results in a differential increased expression of the immune checkpoint molecule, PD-L1. Significantly, we observed an upregulation of PD-L1 expression in cells silenced for the MMR genes, MLH1, MSH2, PMS2 and MSH3, as expected. However, we did not observe an increased expression of PD-L1 upon MSH6 loss. This differential expression amongst MMR gene loss was further validated at both RNA and cell surface level. \u0000Upon investigation of the molecular mechanism regulating PD-L1 expression after MMR loss, we observed that that phosphorylation of STAT1 positively correlates with PD-L1 expression whilst STAT3 phosphorylation was negatively correlated, such that increased STAT1 phosphorylation was observed upon MLH1 and PMS2 loss and not in MSH6-deficient cells whilst STAT3 phosphorylation was only observed upon MSH6 loss. Significantly, inhibition of STAT3, both pharmacologically and genetically, reinstated PD-L1 expression in MSH6-deficient cells. \u0000Therefore, we have evidence that loss of specific MMR genes can trigger differential expression of PD-L1 through a STAT1/STAT3 mediated pathway and we hypothesize that it is this differential expression that may in part determine sensitivity to treatment with ICB.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82857984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An Integrated Structure-based Approach for the Development of MDM2 inhibitors 基于集成结构的MDM2抑制剂开发方法
Y. Sheng
MDM2 is an oncogenic E3 ligase found to be overexpressed in a number of human cancers, leading to poor prognosis. MDM2 overexpression inhibits the function of the tumour suppressor p53, which plays a critical role in safeguarding the integrity of the genome. MDM2 ubiquitinates p53 and tags it for proteasomal degradation. MDM2 also exhibits p53-independent oncogenic activities through targeting other tumour suppressor proteins, such as Foxo3a and Rb. Thus, aberrant regulation of Mdm2 is a key factor in promotion of tumor formation and progression and represents an important cancer therapeutic target. Here, we describe an integrated structure- based approach to develop potential lead compounds for inhibition of MDM2 oncogenic activity. We established a structure-based virtual screening strategy and used the crystal structure of the MDM2:MDMX RING domain heterodimer to predict the potential “druggable” pocket on Mdm2. An in silico screening of a small molecule compound library was employed to identify the candidate compounds that could interact with the MDM2:MDMX heterodimer RING domain. Additionally, using biochemical and cellular assays, the candidate compounds were examined for their ability to inhibit MDM2 E3 ligase activity, to induce apoptosis and to inhibit cell proliferation in cancer cell lines. This study reveals that inhibition of the MDM2:MDMX RING heterodimer could be a plausible approach for the development of MDM2 inhibitors as potential anti-cancer therapeutic agents.
MDM2是一种致癌E3连接酶,在许多人类癌症中被发现过表达,导致预后不良。MDM2过表达会抑制肿瘤抑制因子p53的功能,而p53在保护基因组完整性方面起着关键作用。MDM2泛素化p53并标记它进行蛋白酶体降解。MDM2还通过靶向其他肿瘤抑制蛋白(如Foxo3a和Rb)表现出p53独立的致癌活性。因此,Mdm2的异常调控是促进肿瘤形成和进展的关键因素,是重要的肿瘤治疗靶点。在这里,我们描述了一种基于综合结构的方法来开发抑制MDM2致癌活性的潜在先导化合物。我们建立了基于结构的虚拟筛选策略,利用MDM2:MDMX环域异二聚体的晶体结构预测MDM2上潜在的“可药物”口袋。通过对一个小分子化合物文库进行计算机筛选,确定了可能与MDM2:MDMX异二聚体RING结构域相互作用的候选化合物。此外,通过生化和细胞分析,研究了候选化合物抑制MDM2 E3连接酶活性、诱导细胞凋亡和抑制癌细胞增殖的能力。本研究表明,抑制MDM2:MDMX RING异源二聚体可能是开发MDM2抑制剂作为潜在抗癌治疗剂的可行途径。
{"title":"An Integrated Structure-based Approach for the Development of MDM2 inhibitors","authors":"Y. Sheng","doi":"10.3390/iecc2021-09208","DOIUrl":"https://doi.org/10.3390/iecc2021-09208","url":null,"abstract":"MDM2 is an oncogenic E3 ligase found to be overexpressed in a number of human cancers, leading to poor prognosis. MDM2 overexpression inhibits the function of the tumour suppressor p53, which plays a critical role in safeguarding the integrity of the genome. MDM2 ubiquitinates p53 and tags it for proteasomal degradation. MDM2 also exhibits p53-independent oncogenic activities through targeting other tumour suppressor proteins, such as Foxo3a and Rb. Thus, aberrant regulation of Mdm2 is a key factor in promotion of tumor formation and progression and represents an important cancer therapeutic target. Here, we describe an integrated structure- based approach to develop potential lead compounds for inhibition of MDM2 oncogenic activity. We established a structure-based virtual screening strategy and used the crystal \u0000structure of the MDM2:MDMX RING domain heterodimer to predict the potential “druggable” \u0000pocket on Mdm2. An in silico screening of a small molecule compound library was employed to identify the candidate compounds that could interact with the MDM2:MDMX heterodimer RING domain. Additionally, using biochemical and cellular assays, the candidate compounds were examined for their ability to inhibit MDM2 E3 ligase activity, to induce apoptosis and to inhibit cell proliferation in cancer cell lines. This study reveals that inhibition of the MDM2:MDMX RING heterodimer could be a plausible approach for the development of MDM2 inhibitors as potential anti-cancer therapeutic agents.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87946951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Responsive signatures established by pharmaco-transcriptomic correlation analysis identifies subsets for PARP-targeted therapy and reveals potential synergistic interactors 通过药物-转录组学相关分析建立的应答特征确定了parp靶向治疗的亚群,并揭示了潜在的协同相互作用
Hai-xia Yang, Beibei Sun, S. Hall, Ke Xu, Liang Zhao, R. Schmid, R. Peng, F. Yao
(1) Background: Poly (ADP-ribose) polymerases (PARPs) have pleiotropic roles including canonical DNA-damage response (DDR) pathways. PARP inhibition is initially proposed as a synthetic lethal interactor with cancer harboring homologous recombination deficiency (HRD), thus becoming a key therapeutic option for genetically-defined subsets of patients. Recently, there has been increasing evidence supporting the expansion of PARP-targeted therapy beyond HRD. Besides, synthetic lethality pathways for PARP-targeted therapy are being studied extensively due to the rapidly developed resistance to PARP inhibitors (PARPi). (2) Methods: We perform integrative pharmaco-transcriptomic analyses by correlating the drug response profiles of clinically-approved PARPi olaparib with the transcriptomes of solid cancer cell lines (n=659) to establish PARPis responsive gene signatures, which are then evaluated for their reliability using independent drug response datasets, and applied to identify tumor subsets primed for PARPi and potential targets synergistically interacting with PARPi. (3) Results: Based on the pharmaco-transcriptomic correlation analysis, we delineate gene signatures to predict the sensitivity and resistance to olaparib in pan-solid cancer cells, which is confirmed by independent drug response datasets. In further exploring the PARPi sensitivity signature, we identify IDH1/2 (isocitrate dehydrogenase 1/2)-mutated low-grade glioma (LGG) and NEUROD1-driven small cell lung cancer (SCLC) as potential subsets for prioritized PARPi, highlighting relaunching PARPi as a promising and innovative strategy to target these malignancy subtypes. Interestingly, the PARPi responsive signatures display a high degree of heterogeneity in the correlation with the curated HRD signatures across TCGA pan-solid cancer cohort, suggesting that these signatures predictive of PARPi responsiveness are HRD-independent. With the PARPi resistance signature, we identified several potentially synthetic lethal interactors with PARPi, e.g. dasatinib, EGFR, or MEK inhibitors. (4) Conclusions: The established PARPi responsive (sensitive/resistant) signatures in solid tumors exhibit robustness in identifying cancer subtypes that are highly primed for PARP-targeted therapy, and combined targets that synergistically augment the efficacy of PARPi.
(1)背景:聚(adp -核糖)聚合酶(PARPs)具有多种作用,包括典型的dna损伤反应(DDR)途径。PARP抑制最初被认为是一种与携带同源重组缺陷(HRD)的癌症的合成致死相互作用,因此成为基因定义的患者亚群的关键治疗选择。最近,越来越多的证据支持将parp靶向治疗扩展到HRD之外。此外,由于PARP抑制剂(PARPi)耐药性的迅速发展,PARP靶向治疗的合成致死性途径正在被广泛研究。(2)方法:通过将临床批准的PARPi奥拉帕尼的药物反应谱与实体癌细胞(n=659)的转录组相关联,进行综合药物-转录组学分析,建立PARPi反应基因特征,然后使用独立的药物反应数据集评估其可靠性,并应用于鉴定PARPi的肿瘤亚群和与PARPi协同作用的潜在靶点。(3)结果:基于药物-转录组学相关分析,我们描绘了基因特征来预测泛实体癌细胞对奥拉帕尼的敏感性和耐药性,并通过独立的药物反应数据集证实了这一点。在进一步探索PARPi的敏感性特征时,我们发现IDH1/2(异柠檬酸脱氢酶1/2)突变的低级别胶质瘤(LGG)和neurod1驱动的小细胞肺癌(SCLC)是优先PARPi的潜在亚群,强调重新启动PARPi是一种有前途的创新策略,可以靶向这些恶性肿瘤亚型。有趣的是,在TCGA泛实体癌队列中,PARPi反应性特征与策划的HRD特征的相关性显示出高度的异质性,这表明这些预测PARPi反应性的特征与HRD无关。根据PARPi的耐药特征,我们确定了几种可能与PARPi合成的致命相互作用物,例如达沙替尼、EGFR或MEK抑制剂。(4)结论:在实体瘤中建立的PARPi应答(敏感/耐药)特征在识别parp靶向治疗的高度启动的癌症亚型以及协同增强PARPi疗效的联合靶点方面具有稳健性。
{"title":"Responsive signatures established by pharmaco-transcriptomic correlation analysis identifies subsets for PARP-targeted therapy and reveals potential synergistic interactors","authors":"Hai-xia Yang, Beibei Sun, S. Hall, Ke Xu, Liang Zhao, R. Schmid, R. Peng, F. Yao","doi":"10.3390/iecc2021-09207","DOIUrl":"https://doi.org/10.3390/iecc2021-09207","url":null,"abstract":"(1) Background: Poly (ADP-ribose) polymerases (PARPs) have pleiotropic roles including canonical DNA-damage response (DDR) pathways. PARP inhibition is initially proposed as a synthetic lethal interactor with cancer harboring homologous recombination deficiency (HRD), thus becoming a key therapeutic option for genetically-defined subsets of patients. Recently, there has been increasing evidence supporting the expansion of PARP-targeted therapy beyond HRD. Besides, synthetic lethality pathways for PARP-targeted therapy are being studied extensively due to the rapidly developed resistance to PARP inhibitors (PARPi). \u0000(2) Methods: We perform integrative pharmaco-transcriptomic analyses by correlating the drug response profiles of clinically-approved PARPi olaparib with the transcriptomes of solid cancer cell lines (n=659) to establish PARPis responsive gene signatures, which are then evaluated for their reliability using independent drug response datasets, and applied to identify tumor subsets primed for PARPi and potential targets synergistically interacting with PARPi. \u0000(3) Results: Based on the pharmaco-transcriptomic correlation analysis, we delineate gene signatures to predict the sensitivity and resistance to olaparib in pan-solid cancer cells, which is confirmed by independent drug response datasets. In further exploring the PARPi sensitivity signature, we identify IDH1/2 (isocitrate dehydrogenase 1/2)-mutated low-grade glioma (LGG) and NEUROD1-driven small cell lung cancer (SCLC) as potential subsets for prioritized PARPi, highlighting relaunching PARPi as a promising and innovative strategy to target these malignancy subtypes. Interestingly, the PARPi responsive signatures display a high degree of heterogeneity in the correlation with the curated HRD signatures across TCGA pan-solid cancer cohort, suggesting that these signatures predictive of PARPi responsiveness are HRD-independent. With the PARPi resistance signature, we identified several potentially synthetic lethal interactors with PARPi, e.g. dasatinib, EGFR, or MEK inhibitors. \u0000(4) Conclusions: The established PARPi responsive (sensitive/resistant) signatures in solid tumors exhibit robustness in identifying cancer subtypes that are highly primed for PARP-targeted therapy, and combined targets that synergistically augment the efficacy of PARPi.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86892091","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting the DNA repair enzyme APE1 in cancer therapy 靶向DNA修复酶APE1在癌症治疗中的应用
Debanu Das, P. Pellicena, M. Duncton, David Wilson, M. Georgiadis, A. Deacon
Cancer cells respond to increases in DNA damage by upregulating their DNA damage response (DDR). The base excision repair (BER) pathway corrects damage to single DNA bases through the action of multiple enzymes, including the central protagonist, apurinic/apyrimidinic endonuclease 1 (APE1). Numerous studies have shown association between increased APE1 levels and enhanced growth, migration, and drug resistance in human tumor cells, as well as with decreased patient survival. APE1 has been implicated in over 20 human cancers, making this an attractive target for developing anticancer therapies. Despite intensive effort, there are currently no clinical endonuclease inhibitors of APE1. We have used a newly developed high-throughput protein X-ray crystallography-based fragment screen to obtain starting points for the design of molecules to block APE1 function. Starting with a proprietary fragment library, we obtained high quality fragment-bound crystal structures showing diversity of chemical matter and hit location, representing the first experimental 3D structures of APE1 bound to drug-like molecules, thereby resolving a primary bottleneck in the path to inhibitor development. The implementation of this unique lead discovery campaign has facilitated three independent strategies toward the development of APE1 inhibitors, including (i) fragment growing and elaboration of hits bound at the endonuclease site; (ii) linking of fragments bound to distinct but proximally located sites, and (iii) use of fragments for the design of hooks to use in targeted protein degradation (TPD) strategies. We are using a combination of computational and medicinal chemistry, structural biology, and biochemical and biophysical studies and will discuss our progress towards these goals.
癌细胞通过上调DNA损伤反应(DDR)来应对DNA损伤的增加。碱基切除修复(BER)途径通过多种酶的作用来纠正单个DNA碱基的损伤,包括中心主角,无嘌呤/无嘧啶内切酶1 (APE1)。大量研究表明,APE1水平升高与人类肿瘤细胞生长、迁移和耐药增强以及患者生存率降低有关。APE1与20多种人类癌症有关,这使得它成为开发抗癌疗法的一个有吸引力的靶点。尽管付出了巨大的努力,但目前还没有临床的APE1核酸内切酶抑制剂。我们使用新开发的基于高通量蛋白质x射线晶体学的片段筛选来获得阻断APE1功能的分子设计的起点。从专有的片段库开始,我们获得了高质量的片段结合晶体结构,显示了化学物质和命中位置的多样性,代表了APE1与药物样分子结合的第一个实验3D结构,从而解决了抑制剂开发路径中的主要瓶颈。这种独特的先导发现活动的实施促进了开发APE1抑制剂的三种独立策略,包括(i)片段生长和在核酸内切酶位点结合的命中的细化;(ii)将结合到不同但近端位置的片段连接起来,以及(iii)使用片段设计钩子,用于靶向蛋白质降解(TPD)策略。我们正在结合计算化学和药物化学、结构生物学、生化和生物物理学研究,并将讨论我们在实现这些目标方面的进展。
{"title":"Targeting the DNA repair enzyme APE1 in cancer therapy","authors":"Debanu Das, P. Pellicena, M. Duncton, David Wilson, M. Georgiadis, A. Deacon","doi":"10.3390/iecc2021-09201","DOIUrl":"https://doi.org/10.3390/iecc2021-09201","url":null,"abstract":"Cancer cells respond to increases in DNA damage by upregulating their DNA damage response (DDR). The base excision repair (BER) pathway corrects damage to single DNA bases through the action of multiple enzymes, including the central protagonist, apurinic/apyrimidinic endonuclease 1 (APE1). Numerous studies have shown association between increased APE1 levels and enhanced growth, migration, and drug resistance in human tumor cells, as well as with decreased patient survival. APE1 has been implicated in over 20 human cancers, making this an attractive target for developing anticancer therapies. Despite intensive effort, there are currently no clinical endonuclease inhibitors of APE1. We have used a newly developed high-throughput protein X-ray crystallography-based fragment screen to obtain starting points for the design of molecules to block APE1 function. Starting with a proprietary fragment library, we obtained high quality fragment-bound crystal structures showing diversity of chemical matter and hit location, representing the first experimental 3D structures of APE1 bound to drug-like molecules, thereby resolving a primary bottleneck in the path to inhibitor development. The implementation of this unique lead discovery campaign has facilitated three independent strategies toward the development of APE1 inhibitors, including (i) fragment growing and elaboration of hits bound at the endonuclease site; (ii) linking of fragments bound to distinct but proximally located sites, and (iii) use of fragments for the design of hooks to use in targeted protein degradation (TPD) strategies. We are using a combination of computational and medicinal chemistry, structural biology, and biochemical and biophysical studies and will discuss our progress towards these goals.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83578919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract IA15: PARP inhibitor resistance and acquired vulnerability in breast cancer 摘要:乳腺癌中PARP抑制剂的耐药与获得性易感性
A. D’Andrea
Large-scale genomic studies have demonstrated that some breast cancers, especially triple-negative breast cancers (TNBCs), harbor genetic and epigenetic alterations in homologous recombination repair (HRR) pathway genes. The most commonly altered HRR genes are BRCA1 and BRCA2, followed by other Fanconi anemia genes including FANCN/PALB2, FANCO/RAD51, FANCJ/BRIP, and FANCA. Loss of HRR causes genomic instability, hyperdependence on alternative DNA repair mechanisms, and enhanced sensitivity to platinum analogues, topoisomerase inhibitors, and PARP inhibitors (PARPi). The synthetic lethal interaction with PARPi is being exploited therapeutically in diverse clinical contexts and most notably in ovarian cancer where the PARPi olaparib is FDA approved for use in patients with germline BRCA1/2 mutations. PARP inhibitor resistance has already emerged as a vexing clinical problem for the treatment of BRCA1/2 deficient tumors. The most prevalent mechanism of PARPi resistance is secondary events that cancel the original HRR alteration and restore HRR proficiency. However, PARPi resistance may still develop without restoration of HRR proficiency via disruption of multiple proteins, such as PTIP or CHD4, that leads to replication fork (RF) stabilization. Importantly, this latter mechanism—namely, the restoration of RF stability—appears to be a highly prevalent mechanism of PARP inhibitor resistance in vitro and in vivo, particularly in tumor cells with an underlying BRCA2 deficiency. Due to their underlying deficiency in BRCA2 and inability to generate RAD51 nucleofilaments, these tumor cells are unable to restore HRR mechanisms. Instead, these cells acquire PARP inhibitor resistance by limiting the nucleolytic degradation of their stalled replication forks. We have recently made the surprising observation that BRCA2-deficient tumors can become resistant to PARPi by downregulating the expression of the polycomb repressive complex PRC2, a methyltransferase complex containing EZH2, SUZ12, EED, and RbAp48. Importantly, downregulation of PRC2 results in the reduced recruitment of the nuclease MUS81 to the RF, thereby providing a novel mechanism of RF protection and PARPi resistance. A molecular understanding of PARP inhibitor resistance mechanisms may allow the generation of a new class of drugs, or a repurposing of existing drugs, which may reverse this resistance and extend the use of PARP inhibitors to more tumor types. Citation Format: Alan D. D’Andrea. PARP inhibitor resistance and acquired vulnerability in breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr IA15.
大规模基因组研究表明,一些乳腺癌,特别是三阴性乳腺癌(tnbc),同源重组修复(HRR)途径基因存在遗传和表观遗传改变。最常见的HRR改变基因是BRCA1和BRCA2,其次是其他范可尼贫血基因,包括FANCN/PALB2、FANCO/RAD51、FANCJ/BRIP和FANCA。HRR的缺失导致基因组不稳定,对替代DNA修复机制的高度依赖,以及对铂类似物、拓扑异构酶抑制剂和PARP抑制剂(PARPi)的敏感性增强。与PARPi的合成致死性相互作用正被用于多种临床环境的治疗,最值得注意的是卵巢癌,PARPi奥拉帕尼已被FDA批准用于生殖系BRCA1/2突变患者。PARP抑制剂耐药性已经成为治疗BRCA1/2缺陷肿瘤的一个令人烦恼的临床问题。PARPi耐药最普遍的机制是次要事件,这些事件取消了原始HRR改变并恢复HRR熟练程度。然而,PARPi耐药性仍然可能通过破坏多种蛋白质(如PTIP或CHD4)而导致复制叉(RF)稳定,而没有恢复HRR的熟练程度。重要的是,后一种机制——即RF稳定性的恢复——似乎是体外和体内PARP抑制剂耐药性的一种高度普遍的机制,特别是在潜在BRCA2缺乏的肿瘤细胞中。由于其潜在的BRCA2缺失和无法产生RAD51核丝,这些肿瘤细胞无法恢复HRR机制。相反,这些细胞通过限制其停滞的复制叉的核分解降解获得PARP抑制剂抗性。我们最近进行了令人惊讶的观察,发现brca2缺陷的肿瘤可以通过下调多梳抑制复合物PRC2(一种含有EZH2、SUZ12、EED和RbAp48的甲基转移酶复合物)的表达而对PARPi产生耐药性。重要的是,PRC2的下调导致核酸酶MUS81向RF的募集减少,从而提供了一种新的RF保护和PARPi抗性机制。对PARP抑制剂耐药机制的分子理解可能会产生一类新的药物,或者对现有药物进行重新利用,这可能会逆转这种耐药性,并将PARP抑制剂的使用范围扩大到更多的肿瘤类型。引用格式:Alan D. D 'Andrea。乳腺癌中PARP抑制剂的耐药与获得性易感性[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;中华肿瘤杂志,2018;16(8):1 - 5。
{"title":"Abstract IA15: PARP inhibitor resistance and acquired vulnerability in breast cancer","authors":"A. D’Andrea","doi":"10.1158/1557-3125.ADVBC17-IA15","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-IA15","url":null,"abstract":"Large-scale genomic studies have demonstrated that some breast cancers, especially triple-negative breast cancers (TNBCs), harbor genetic and epigenetic alterations in homologous recombination repair (HRR) pathway genes. The most commonly altered HRR genes are BRCA1 and BRCA2, followed by other Fanconi anemia genes including FANCN/PALB2, FANCO/RAD51, FANCJ/BRIP, and FANCA. Loss of HRR causes genomic instability, hyperdependence on alternative DNA repair mechanisms, and enhanced sensitivity to platinum analogues, topoisomerase inhibitors, and PARP inhibitors (PARPi). The synthetic lethal interaction with PARPi is being exploited therapeutically in diverse clinical contexts and most notably in ovarian cancer where the PARPi olaparib is FDA approved for use in patients with germline BRCA1/2 mutations. PARP inhibitor resistance has already emerged as a vexing clinical problem for the treatment of BRCA1/2 deficient tumors. The most prevalent mechanism of PARPi resistance is secondary events that cancel the original HRR alteration and restore HRR proficiency. However, PARPi resistance may still develop without restoration of HRR proficiency via disruption of multiple proteins, such as PTIP or CHD4, that leads to replication fork (RF) stabilization. Importantly, this latter mechanism—namely, the restoration of RF stability—appears to be a highly prevalent mechanism of PARP inhibitor resistance in vitro and in vivo, particularly in tumor cells with an underlying BRCA2 deficiency. Due to their underlying deficiency in BRCA2 and inability to generate RAD51 nucleofilaments, these tumor cells are unable to restore HRR mechanisms. Instead, these cells acquire PARP inhibitor resistance by limiting the nucleolytic degradation of their stalled replication forks. We have recently made the surprising observation that BRCA2-deficient tumors can become resistant to PARPi by downregulating the expression of the polycomb repressive complex PRC2, a methyltransferase complex containing EZH2, SUZ12, EED, and RbAp48. Importantly, downregulation of PRC2 results in the reduced recruitment of the nuclease MUS81 to the RF, thereby providing a novel mechanism of RF protection and PARPi resistance. A molecular understanding of PARP inhibitor resistance mechanisms may allow the generation of a new class of drugs, or a repurposing of existing drugs, which may reverse this resistance and extend the use of PARP inhibitors to more tumor types. Citation Format: Alan D. D’Andrea. PARP inhibitor resistance and acquired vulnerability in breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr IA15.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80061716","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1