在微卫星不稳定性癌症中靶向具有合成致死性的维尔纳螺旋酶:有前景的治疗方法

Huanzhang Xie, Jing Zhang
{"title":"在微卫星不稳定性癌症中靶向具有合成致死性的维尔纳螺旋酶:有前景的治疗方法","authors":"Huanzhang Xie,&nbsp;Jing Zhang","doi":"10.1002/mog2.82","DOIUrl":null,"url":null,"abstract":"<p>Two recent companion papers published in Nature have reported two promising drug candidates, HRO761<span><sup>1</sup></span> and VVD-133214,<span><sup>2</sup></span> for microsatellite instability (MSI) cancers targeting the werner syndrome RecQ helicase (WRN), a synthetic lethal target in cancer cells with MSI. Currently, both candidates are undergoing clinical trials to evaluate their safety, tolerability, and preliminary antitumor activity in MSI patients.</p><p>Microsatellites, also known as short tandem repeats, are susceptible to slippage errors during replication, rendering them heavily reliant on the DNA mismatch repair (MMR) system. MMR deficiency results in widespread MSI by failing to correct replication errors, thus initiating cancer via aberrant tumor suppressor gene function. The prevalence of MSI ranges from 10% to 30% across multiple cancer types, such as colorectal, endometrial, ovarian, and gastric cancers.<span><sup>1-3</sup></span> In MSI tumors, deficiencies in MMR mechanisms heighten genomic instability, prompting the activation of alternative DNA repair pathways, including those implicating WRN. Inhibitors targeting WRN in MSI cancer cells, which already possess compromised DNA repair mechanisms, may induce synthetic lethality, thereby triggering DNA damage and subsequent cancer cell death. This targeted approach is ineffective against normal or microsatellite instability (MSS) cells, as their MMR mechanisms remain intact. Hence, WRN inhibitors emerge as a highly promising synthetic lethal agent, with the potential to selectively eradicate tumor cells while sparing normal cells (Figure 1A).</p><p>Novartis researchers reported HRO761,<span><sup>1</sup></span> a novel WRN helicase inhibitor (Figure 1B) which targets the ATPase of WRN as a noncovalent inhibitor. Cocrystal structures of HRO761 with WRN helicase revealed its binding to a nonconserved site at the D1–D2 interface, immobilizing WRN in an inactive conformation with an approximate 180° rotation relative to the adenosine triphosphate (ATP)-bound conformation (Figure 1C). Despite its 702 Da molecular weight, HRO761 displayed favorable physicochemical properties and pharmacokinetics (PK), with a clean off-target profile. In vitro cellular assays showed that HRO761 exhibits an IC<sub>50</sub> of 100 nM in ATPase assays at high ATP concentration, effectively impairing the viability of MSI cancer cells, while showing no effect in MSS cells. Furthermore, characterization of HRO761 treatment effects on MSI cells revealed time- and dose-dependent cell cycle arrest and DNA damage, regardless of p53 mutation status. In the SW48 cell-derived xenografts (CDX) model, oral administration of 15–60 mg/kg HRO761 resulted in significant tumor regression without observed toxicity. Additionally, combination therapy involving HRO761 with other antitumor drugs may enhance treatment efficacy and reduce side effects and resistance. In vivo studies show complete tumor regression with combination of HRO761 and irinotecan, without affecting body weight. Consequently, clinical trial NCT05838768 is presently underway to explore the synergistic effects of HRO761 when combined with either irinotecan or tislelizumab.</p><p>On the same day, Vividion Therapeutics researchers reported another WRN inhibitor, VVD-133214 (also known as RO7589831).<span><sup>2</sup></span> Unlike HRO761, VVD-133214 acts as an irreversible covalent inhibitor. Cocrystal analysis revealed that VVD-133214 can bind to the ATPase domain simultaneously with adenosine diphosphate (ADP), forming a covalent bond with the sulfur atom of C727 (Figure 1C). VVD-133214 demonstrates ADP cooperativity, wherein preincubation with ADP reduces its IC<sub>50</sub> to 0.1 μM, compared to 3 μM in the absence of ADP. In vitro studies showed that VVD-133214 exhibited sensitivity in nearly 80% of MSI-high (MSI-H) cell lines (11 out of 14), while MSS cell lines showed no responsiveness. Oral administration of VVD-133214 at ≥5 mg/kg once daily achieved nearly complete (approximately 95%) tumor target engagement. Remarkably, VVD-133214 exhibited efficacy in decreasing tumor burden in six out of seven distinct MSI-H colorectal cancer patient-derived xenograft (PDX) models, including those with p53 mutations.</p><p>Synthetic lethality is a significant advancement in cancer treatment, offering a novel strategy, particularly for addressing previously challenging mutations. Unlike traditional therapies that target rapidly dividing cells indiscriminately, synthetic lethality exploits genetic differences to selectively eliminate tumor cells with specific mutations while sparing normal cells. This approach minimizes side effects and damage to healthy tissues, providing more precise and effective treatment options for cancer patients.</p><p>In 2014, AstraZeneca's PARP inhibitor olaparib became the first FDA-approved synthetic lethality inhibitor for treating breast cancer susceptibilty gene mutated ovarian cancer, marking the beginning of synthetic lethality therapy for cancer. In 2019, a series of independent studies revealed a therapeutic potential synthetic lethality relationship between WRN and MSI tumors, sparking interest in targeted drug development against WRN, and attracted attention from global pharmaceutical giants such as Roche, Novartis, and GSK. Novartis possesses patents for several WRN inhibitors, notably HRO761. Additionally, Vividion, a Bayer subsidiary, collaborated with Roche to develop VVD-133214 in 2020, while GSK acquired IDEAYA's WRN project for developing candidate drugs targeting WRN. Figure 1B lists some active WRN inhibitors, among which the Vividion and GSK<span><sup>4</sup></span> series are known as covalent inhibitors. Their diverse scaffolds suggest considerable potential for further exploration in developing WRN-targeting inhibitors.</p><p>However, identifying inhibitors targeting WRN has presented challenges due to the abundance of false positives, such as protein interference. Previously published compounds like NSC617145, NSC19630, and ML216 have undergone rigorous validation and have been proven not to be specific WRN inhibitors.<span><sup>5</sup></span> Moreover, targeting the ATPase domain poses challenges due to the conservative nature of the domain, prompting exploration of allosteric inhibitors as a promising strategy for reducing drug toxicities. Both HRO761 and VVD-133214 bind to the allosteric site near C727, inducing conformational changes in WRN, thereby enhancing specificity in regulating the target protein's function. Furthermore, the selectivity of WRN inhibitors towards other members of the RECQ helicase family is crucial, as these enzymes play significant roles in cellular functions such as senescence and DNA repair. Off-target effects may lead to severe toxic side effects. HRO761 shows excellent selectivity, with an IC<sub>50</sub> of 0.1 μM for WRN and &gt;100 μM for other RECQ helicases, indicating promising development prospects.</p><p>In summary, recent reports in Nature highlight the growing interest in WRN synthetic lethality in MSI tumor therapy. Both HRO761 and VVD-133214 exhibit specificity for MSI cancer cells, inducing cancer cell death while preserving healthy cells. Preclinical studies demonstrate their efficacy in reducing tumor burden and inducing regression in MSI cancer models. Currently, both compounds are undergoing Phase I clinical trials to evaluate their safety and tolerability. These discoveries highlight the significant potential of WRN inhibitors in the treatment of MSI cancers, and advancing precision medicine strategies in oncology.</p><p><b>Huanzhang Xie</b>: Visualization (lead); writing—original draft (lead). <b>Jing Zhang</b>: Conceptualization (lead); writing—review and editing (equal). Both authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>The authors have nothing to report.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 2","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2024-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.82","citationCount":"0","resultStr":"{\"title\":\"Targeting werner helicase with synthetic lethality in microsatellite instability cancers: Promising therapeutic approaches\",\"authors\":\"Huanzhang Xie,&nbsp;Jing Zhang\",\"doi\":\"10.1002/mog2.82\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Two recent companion papers published in Nature have reported two promising drug candidates, HRO761<span><sup>1</sup></span> and VVD-133214,<span><sup>2</sup></span> for microsatellite instability (MSI) cancers targeting the werner syndrome RecQ helicase (WRN), a synthetic lethal target in cancer cells with MSI. Currently, both candidates are undergoing clinical trials to evaluate their safety, tolerability, and preliminary antitumor activity in MSI patients.</p><p>Microsatellites, also known as short tandem repeats, are susceptible to slippage errors during replication, rendering them heavily reliant on the DNA mismatch repair (MMR) system. MMR deficiency results in widespread MSI by failing to correct replication errors, thus initiating cancer via aberrant tumor suppressor gene function. The prevalence of MSI ranges from 10% to 30% across multiple cancer types, such as colorectal, endometrial, ovarian, and gastric cancers.<span><sup>1-3</sup></span> In MSI tumors, deficiencies in MMR mechanisms heighten genomic instability, prompting the activation of alternative DNA repair pathways, including those implicating WRN. Inhibitors targeting WRN in MSI cancer cells, which already possess compromised DNA repair mechanisms, may induce synthetic lethality, thereby triggering DNA damage and subsequent cancer cell death. This targeted approach is ineffective against normal or microsatellite instability (MSS) cells, as their MMR mechanisms remain intact. Hence, WRN inhibitors emerge as a highly promising synthetic lethal agent, with the potential to selectively eradicate tumor cells while sparing normal cells (Figure 1A).</p><p>Novartis researchers reported HRO761,<span><sup>1</sup></span> a novel WRN helicase inhibitor (Figure 1B) which targets the ATPase of WRN as a noncovalent inhibitor. Cocrystal structures of HRO761 with WRN helicase revealed its binding to a nonconserved site at the D1–D2 interface, immobilizing WRN in an inactive conformation with an approximate 180° rotation relative to the adenosine triphosphate (ATP)-bound conformation (Figure 1C). Despite its 702 Da molecular weight, HRO761 displayed favorable physicochemical properties and pharmacokinetics (PK), with a clean off-target profile. In vitro cellular assays showed that HRO761 exhibits an IC<sub>50</sub> of 100 nM in ATPase assays at high ATP concentration, effectively impairing the viability of MSI cancer cells, while showing no effect in MSS cells. Furthermore, characterization of HRO761 treatment effects on MSI cells revealed time- and dose-dependent cell cycle arrest and DNA damage, regardless of p53 mutation status. In the SW48 cell-derived xenografts (CDX) model, oral administration of 15–60 mg/kg HRO761 resulted in significant tumor regression without observed toxicity. Additionally, combination therapy involving HRO761 with other antitumor drugs may enhance treatment efficacy and reduce side effects and resistance. In vivo studies show complete tumor regression with combination of HRO761 and irinotecan, without affecting body weight. Consequently, clinical trial NCT05838768 is presently underway to explore the synergistic effects of HRO761 when combined with either irinotecan or tislelizumab.</p><p>On the same day, Vividion Therapeutics researchers reported another WRN inhibitor, VVD-133214 (also known as RO7589831).<span><sup>2</sup></span> Unlike HRO761, VVD-133214 acts as an irreversible covalent inhibitor. Cocrystal analysis revealed that VVD-133214 can bind to the ATPase domain simultaneously with adenosine diphosphate (ADP), forming a covalent bond with the sulfur atom of C727 (Figure 1C). VVD-133214 demonstrates ADP cooperativity, wherein preincubation with ADP reduces its IC<sub>50</sub> to 0.1 μM, compared to 3 μM in the absence of ADP. In vitro studies showed that VVD-133214 exhibited sensitivity in nearly 80% of MSI-high (MSI-H) cell lines (11 out of 14), while MSS cell lines showed no responsiveness. Oral administration of VVD-133214 at ≥5 mg/kg once daily achieved nearly complete (approximately 95%) tumor target engagement. Remarkably, VVD-133214 exhibited efficacy in decreasing tumor burden in six out of seven distinct MSI-H colorectal cancer patient-derived xenograft (PDX) models, including those with p53 mutations.</p><p>Synthetic lethality is a significant advancement in cancer treatment, offering a novel strategy, particularly for addressing previously challenging mutations. Unlike traditional therapies that target rapidly dividing cells indiscriminately, synthetic lethality exploits genetic differences to selectively eliminate tumor cells with specific mutations while sparing normal cells. This approach minimizes side effects and damage to healthy tissues, providing more precise and effective treatment options for cancer patients.</p><p>In 2014, AstraZeneca's PARP inhibitor olaparib became the first FDA-approved synthetic lethality inhibitor for treating breast cancer susceptibilty gene mutated ovarian cancer, marking the beginning of synthetic lethality therapy for cancer. In 2019, a series of independent studies revealed a therapeutic potential synthetic lethality relationship between WRN and MSI tumors, sparking interest in targeted drug development against WRN, and attracted attention from global pharmaceutical giants such as Roche, Novartis, and GSK. Novartis possesses patents for several WRN inhibitors, notably HRO761. Additionally, Vividion, a Bayer subsidiary, collaborated with Roche to develop VVD-133214 in 2020, while GSK acquired IDEAYA's WRN project for developing candidate drugs targeting WRN. Figure 1B lists some active WRN inhibitors, among which the Vividion and GSK<span><sup>4</sup></span> series are known as covalent inhibitors. Their diverse scaffolds suggest considerable potential for further exploration in developing WRN-targeting inhibitors.</p><p>However, identifying inhibitors targeting WRN has presented challenges due to the abundance of false positives, such as protein interference. Previously published compounds like NSC617145, NSC19630, and ML216 have undergone rigorous validation and have been proven not to be specific WRN inhibitors.<span><sup>5</sup></span> Moreover, targeting the ATPase domain poses challenges due to the conservative nature of the domain, prompting exploration of allosteric inhibitors as a promising strategy for reducing drug toxicities. Both HRO761 and VVD-133214 bind to the allosteric site near C727, inducing conformational changes in WRN, thereby enhancing specificity in regulating the target protein's function. Furthermore, the selectivity of WRN inhibitors towards other members of the RECQ helicase family is crucial, as these enzymes play significant roles in cellular functions such as senescence and DNA repair. Off-target effects may lead to severe toxic side effects. HRO761 shows excellent selectivity, with an IC<sub>50</sub> of 0.1 μM for WRN and &gt;100 μM for other RECQ helicases, indicating promising development prospects.</p><p>In summary, recent reports in Nature highlight the growing interest in WRN synthetic lethality in MSI tumor therapy. Both HRO761 and VVD-133214 exhibit specificity for MSI cancer cells, inducing cancer cell death while preserving healthy cells. Preclinical studies demonstrate their efficacy in reducing tumor burden and inducing regression in MSI cancer models. Currently, both compounds are undergoing Phase I clinical trials to evaluate their safety and tolerability. These discoveries highlight the significant potential of WRN inhibitors in the treatment of MSI cancers, and advancing precision medicine strategies in oncology.</p><p><b>Huanzhang Xie</b>: Visualization (lead); writing—original draft (lead). <b>Jing Zhang</b>: Conceptualization (lead); writing—review and editing (equal). Both authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>The authors have nothing to report.</p>\",\"PeriodicalId\":100902,\"journal\":{\"name\":\"MedComm – Oncology\",\"volume\":\"3 2\",\"pages\":\"\"},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-06-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.82\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm – Oncology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mog2.82\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm – Oncology","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mog2.82","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

最近发表在《自然》(Nature)杂志上的两篇论文报告了两种很有前景的候选药物:HRO7611 和 VVD-1332142 ,这两种药物针对的是沃纳综合征 RecQ 螺旋酶(WRN),这是 MSI 癌细胞中的一个合成致死靶点,用于治疗微卫星不稳定(MSI)癌症。目前,这两种候选药物都在进行临床试验,以评估它们在 MSI 患者中的安全性、耐受性和初步抗肿瘤活性。微卫星又称短串联重复序列,在复制过程中容易出现滑动错误,因此严重依赖 DNA 错配修复(MMR)系统。MMR 缺乏会导致无法纠正复制错误,从而导致广泛的 MSI,进而通过肿瘤抑制基因的异常功能引发癌症。在多种癌症类型中,如结直肠癌、子宫内膜癌、卵巢癌和胃癌,MSI 的发病率从 10% 到 30% 不等。1-3 在 MSI 肿瘤中,MMR 机制的缺陷增加了基因组的不稳定性,促使替代 DNA 修复途径(包括与 WRN 有关的途径)被激活。MSI 癌细胞的 DNA 修复机制已经受损,针对 WRN 的抑制剂可能会诱导合成致死,从而引发 DNA 损伤并导致癌细胞死亡。这种靶向方法对正常细胞或微卫星不稳定(MSS)细胞无效,因为它们的 MMR 机制仍然完好。因此,WRN 抑制剂成为一种极具前景的合成致死剂,它有可能选择性地消灭肿瘤细胞,而放过正常细胞(图 1A)。诺华公司的研究人员报告了一种新型 WRN 螺旋酶抑制剂 HRO7611(图 1B),它以 WRN 的 ATP 酶为靶点,是一种非共价抑制剂。HRO761 与 WRN 螺旋酶的共晶体结构显示,HRO761 与 D1-D2 界面的一个非保留位点结合,将 WRN 固定在一个非活性构象中,相对于三磷酸腺苷(ATP)结合构象旋转了大约 180°(图 1C)。尽管 HRO761 的分子量为 702 Da,但它显示出良好的理化特性和药代动力学(PK),而且没有脱靶现象。体外细胞实验显示,在高浓度 ATP 的 ATPase 实验中,HRO761 的 IC50 值为 100 nM,可有效降低 MSI 癌细胞的活力,而对 MSS 细胞则无影响。此外,HRO761 对 MSI 细胞的处理效果表征显示,细胞周期停滞和 DNA 损伤与时间和剂量有关,与 p53 突变状态无关。在 SW48 细胞衍生异种移植(CDX)模型中,口服 15-60 mg/kg HRO761 可使肿瘤显著消退,且无明显毒性。此外,HRO761与其他抗肿瘤药物的联合治疗可提高疗效,减少副作用和耐药性。体内研究显示,HRO761 与伊立替康联合治疗可使肿瘤完全消退,且不影响体重。因此,目前正在进行 NCT05838768 临床试验,以探索 HRO761 与伊立替康或替舒利珠单抗联用时的协同效应。2 与 HRO761 不同,VVD-133214 是一种不可逆的共价抑制剂。2 与 HRO761 不同,VVD-133214 是一种不可逆的共价抑制剂。共晶分析表明,VVD-133214 可与二磷酸腺苷(ADP)同时结合到 ATPase 结构域,与 C727 的硫原子形成共价键(图 1C)。VVD-133214 具有 ADP 协同作用,与 ADP 预孵育时的 IC50 值为 3 μM,而无 ADP 时的 IC50 值为 0.1 μM。体外研究显示,VVD-133214 对近 80% 的 MSI-高(MSI-H)细胞系(14 个细胞系中的 11 个)具有敏感性,而 MSS 细胞系则没有反应。口服 VVD-133214 的剂量≥5 毫克/千克,每天一次,可实现几乎完全(约 95%)的肿瘤靶点参与。值得注意的是,VVD-133214 在七种不同的 MSI-H 结直肠癌患者衍生异种移植(PDX)模型中的六种都显示出了降低肿瘤负荷的疗效,包括那些 p53 突变的模型。与不加区分地针对快速分裂细胞的传统疗法不同,合成致死疗法利用基因差异,选择性地消灭具有特定突变的肿瘤细胞,同时保留正常细胞。2014 年,阿斯利康的 PARP 抑制剂 olaparib 成为首个获得 FDA 批准的合成致死抑制剂,用于治疗乳腺癌易感基因突变的卵巢癌,标志着合成致死疗法开始用于癌症治疗。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Targeting werner helicase with synthetic lethality in microsatellite instability cancers: Promising therapeutic approaches

Two recent companion papers published in Nature have reported two promising drug candidates, HRO7611 and VVD-133214,2 for microsatellite instability (MSI) cancers targeting the werner syndrome RecQ helicase (WRN), a synthetic lethal target in cancer cells with MSI. Currently, both candidates are undergoing clinical trials to evaluate their safety, tolerability, and preliminary antitumor activity in MSI patients.

Microsatellites, also known as short tandem repeats, are susceptible to slippage errors during replication, rendering them heavily reliant on the DNA mismatch repair (MMR) system. MMR deficiency results in widespread MSI by failing to correct replication errors, thus initiating cancer via aberrant tumor suppressor gene function. The prevalence of MSI ranges from 10% to 30% across multiple cancer types, such as colorectal, endometrial, ovarian, and gastric cancers.1-3 In MSI tumors, deficiencies in MMR mechanisms heighten genomic instability, prompting the activation of alternative DNA repair pathways, including those implicating WRN. Inhibitors targeting WRN in MSI cancer cells, which already possess compromised DNA repair mechanisms, may induce synthetic lethality, thereby triggering DNA damage and subsequent cancer cell death. This targeted approach is ineffective against normal or microsatellite instability (MSS) cells, as their MMR mechanisms remain intact. Hence, WRN inhibitors emerge as a highly promising synthetic lethal agent, with the potential to selectively eradicate tumor cells while sparing normal cells (Figure 1A).

Novartis researchers reported HRO761,1 a novel WRN helicase inhibitor (Figure 1B) which targets the ATPase of WRN as a noncovalent inhibitor. Cocrystal structures of HRO761 with WRN helicase revealed its binding to a nonconserved site at the D1–D2 interface, immobilizing WRN in an inactive conformation with an approximate 180° rotation relative to the adenosine triphosphate (ATP)-bound conformation (Figure 1C). Despite its 702 Da molecular weight, HRO761 displayed favorable physicochemical properties and pharmacokinetics (PK), with a clean off-target profile. In vitro cellular assays showed that HRO761 exhibits an IC50 of 100 nM in ATPase assays at high ATP concentration, effectively impairing the viability of MSI cancer cells, while showing no effect in MSS cells. Furthermore, characterization of HRO761 treatment effects on MSI cells revealed time- and dose-dependent cell cycle arrest and DNA damage, regardless of p53 mutation status. In the SW48 cell-derived xenografts (CDX) model, oral administration of 15–60 mg/kg HRO761 resulted in significant tumor regression without observed toxicity. Additionally, combination therapy involving HRO761 with other antitumor drugs may enhance treatment efficacy and reduce side effects and resistance. In vivo studies show complete tumor regression with combination of HRO761 and irinotecan, without affecting body weight. Consequently, clinical trial NCT05838768 is presently underway to explore the synergistic effects of HRO761 when combined with either irinotecan or tislelizumab.

On the same day, Vividion Therapeutics researchers reported another WRN inhibitor, VVD-133214 (also known as RO7589831).2 Unlike HRO761, VVD-133214 acts as an irreversible covalent inhibitor. Cocrystal analysis revealed that VVD-133214 can bind to the ATPase domain simultaneously with adenosine diphosphate (ADP), forming a covalent bond with the sulfur atom of C727 (Figure 1C). VVD-133214 demonstrates ADP cooperativity, wherein preincubation with ADP reduces its IC50 to 0.1 μM, compared to 3 μM in the absence of ADP. In vitro studies showed that VVD-133214 exhibited sensitivity in nearly 80% of MSI-high (MSI-H) cell lines (11 out of 14), while MSS cell lines showed no responsiveness. Oral administration of VVD-133214 at ≥5 mg/kg once daily achieved nearly complete (approximately 95%) tumor target engagement. Remarkably, VVD-133214 exhibited efficacy in decreasing tumor burden in six out of seven distinct MSI-H colorectal cancer patient-derived xenograft (PDX) models, including those with p53 mutations.

Synthetic lethality is a significant advancement in cancer treatment, offering a novel strategy, particularly for addressing previously challenging mutations. Unlike traditional therapies that target rapidly dividing cells indiscriminately, synthetic lethality exploits genetic differences to selectively eliminate tumor cells with specific mutations while sparing normal cells. This approach minimizes side effects and damage to healthy tissues, providing more precise and effective treatment options for cancer patients.

In 2014, AstraZeneca's PARP inhibitor olaparib became the first FDA-approved synthetic lethality inhibitor for treating breast cancer susceptibilty gene mutated ovarian cancer, marking the beginning of synthetic lethality therapy for cancer. In 2019, a series of independent studies revealed a therapeutic potential synthetic lethality relationship between WRN and MSI tumors, sparking interest in targeted drug development against WRN, and attracted attention from global pharmaceutical giants such as Roche, Novartis, and GSK. Novartis possesses patents for several WRN inhibitors, notably HRO761. Additionally, Vividion, a Bayer subsidiary, collaborated with Roche to develop VVD-133214 in 2020, while GSK acquired IDEAYA's WRN project for developing candidate drugs targeting WRN. Figure 1B lists some active WRN inhibitors, among which the Vividion and GSK4 series are known as covalent inhibitors. Their diverse scaffolds suggest considerable potential for further exploration in developing WRN-targeting inhibitors.

However, identifying inhibitors targeting WRN has presented challenges due to the abundance of false positives, such as protein interference. Previously published compounds like NSC617145, NSC19630, and ML216 have undergone rigorous validation and have been proven not to be specific WRN inhibitors.5 Moreover, targeting the ATPase domain poses challenges due to the conservative nature of the domain, prompting exploration of allosteric inhibitors as a promising strategy for reducing drug toxicities. Both HRO761 and VVD-133214 bind to the allosteric site near C727, inducing conformational changes in WRN, thereby enhancing specificity in regulating the target protein's function. Furthermore, the selectivity of WRN inhibitors towards other members of the RECQ helicase family is crucial, as these enzymes play significant roles in cellular functions such as senescence and DNA repair. Off-target effects may lead to severe toxic side effects. HRO761 shows excellent selectivity, with an IC50 of 0.1 μM for WRN and >100 μM for other RECQ helicases, indicating promising development prospects.

In summary, recent reports in Nature highlight the growing interest in WRN synthetic lethality in MSI tumor therapy. Both HRO761 and VVD-133214 exhibit specificity for MSI cancer cells, inducing cancer cell death while preserving healthy cells. Preclinical studies demonstrate their efficacy in reducing tumor burden and inducing regression in MSI cancer models. Currently, both compounds are undergoing Phase I clinical trials to evaluate their safety and tolerability. These discoveries highlight the significant potential of WRN inhibitors in the treatment of MSI cancers, and advancing precision medicine strategies in oncology.

Huanzhang Xie: Visualization (lead); writing—original draft (lead). Jing Zhang: Conceptualization (lead); writing—review and editing (equal). Both authors have read and approved the final manuscript.

The authors declare no conflict of interest.

The authors have nothing to report.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Macrophage-Mediated Myelin Recycling Promotes Malignant Development of Glioblastoma Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention Lomitapide: Targeting METTL3 to Overcome Osimertinib Resistance in NSCLC Through Autophagy Activation Ephrin A1 ligand-based CAR-T cells for immunotherapy of EphA2-positive cancer Analysis of reoperational reason of patients with thyroid cancer and strategies for its diagnosis and treatment: A 6-year single-center retrospective study
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
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
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
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