首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
Targeting PDHK1 by DCA to restore NK cell function in hepatocellular carcinoma. 通过 DCA 靶向 PDHK1 恢复肝细胞癌中 NK 细胞的功能
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-20 DOI: 10.1158/1535-7163.MCT-24-0222
Xinyi Yang, Yuanyuan Liu, Peng Wang, Min Li, Tong Xiang, Songzuo Xie, Minxing Li, Yan Wang, Desheng Weng, Jingjing Zhao

Pyruvate dehydrogenase complex is a crucial enzyme involved in the oxidation of glucose. It is regulated by pyruvate dehydrogenase kinase and pyruvate dehydrogenase phosphatase. Studies have demonstrated that pyruvate dehydrogenase kinase 1 (PDHK1), a key enzyme in glucose metabolism, behaves like oncogenes. It is highly expressed in tumors and is associated with poor patient prognosis. However, there is limited research on how PDHK1 affects immune cell function. We have established a model of natural killer (NK) cell exhaustion to investigate the impact of dichloroacetate (DCA) on NK cell function. The production of Granzyme B, IFN-γ, TNF-α, and CD107a by NK cells was explored by flow cytometry. The real-time live cell imaging system was used to monitor the ability of NK cells against tumor cells. The Seahorse analyzer was utilized to measure the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) of NK cells. The mouse model was used to investigate the potential of combining DCA with adjuvant NK cell infusion. Our study demonstrated that the hepatocellular carcinoma (HCC) microenvironment mediated NK cellular exhaustion, high expression of PDHK1 and reduced cytokine secretion. We discovered that the PDHK1 inhibitor DCA enhances the activity and function of exhausted NK cells infiltrating the tumor microenvironment. Furthermore, in a subcutaneous HCC mouse model, DCA combined with NK cell treatment resulted in retarding cancer progression. This study indicates the potential of DCA in rescuing NK cell exhaustion and eliciting anti-tumor immunity.

丙酮酸脱氢酶复合物是参与葡萄糖氧化的一种重要酶。它受丙酮酸脱氢酶激酶和丙酮酸脱氢酶磷酸酶的调控。研究表明,丙酮酸脱氢酶激酶 1(PDHK1)是葡萄糖代谢中的一个关键酶,其行为类似于癌基因。它在肿瘤中高表达,与患者预后不良有关。然而,关于 PDHK1 如何影响免疫细胞功能的研究还很有限。我们建立了一个自然杀伤(NK)细胞衰竭模型,研究二氯乙酸(DCA)对NK细胞功能的影响。我们用流式细胞仪检测了 NK 细胞产生的 Granzyme B、IFN-γ、TNF-α 和 CD107a。实时活细胞成像系统用于监测 NK 细胞对抗肿瘤细胞的能力。海马分析仪用于测量 NK 细胞的耗氧率(OCR)和细胞外酸化率(ECAR)。我们利用小鼠模型研究了DCA与NK细胞辅助输注相结合的潜力。我们的研究表明,肝细胞癌(HCC)微环境介导了 NK 细胞衰竭、PDHK1 高表达和细胞因子分泌减少。我们发现,PDHK1 抑制剂 DCA 能增强浸润肿瘤微环境的衰竭 NK 细胞的活性和功能。此外,在皮下 HCC 小鼠模型中,DCA 与 NK 细胞联合治疗可延缓癌症进展。这项研究表明,DCA 在挽救 NK 细胞衰竭和激发抗肿瘤免疫方面具有潜力。
{"title":"Targeting PDHK1 by DCA to restore NK cell function in hepatocellular carcinoma.","authors":"Xinyi Yang, Yuanyuan Liu, Peng Wang, Min Li, Tong Xiang, Songzuo Xie, Minxing Li, Yan Wang, Desheng Weng, Jingjing Zhao","doi":"10.1158/1535-7163.MCT-24-0222","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0222","url":null,"abstract":"<p><p>Pyruvate dehydrogenase complex is a crucial enzyme involved in the oxidation of glucose. It is regulated by pyruvate dehydrogenase kinase and pyruvate dehydrogenase phosphatase. Studies have demonstrated that pyruvate dehydrogenase kinase 1 (PDHK1), a key enzyme in glucose metabolism, behaves like oncogenes. It is highly expressed in tumors and is associated with poor patient prognosis. However, there is limited research on how PDHK1 affects immune cell function. We have established a model of natural killer (NK) cell exhaustion to investigate the impact of dichloroacetate (DCA) on NK cell function. The production of Granzyme B, IFN-γ, TNF-α, and CD107a by NK cells was explored by flow cytometry. The real-time live cell imaging system was used to monitor the ability of NK cells against tumor cells. The Seahorse analyzer was utilized to measure the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) of NK cells. The mouse model was used to investigate the potential of combining DCA with adjuvant NK cell infusion. Our study demonstrated that the hepatocellular carcinoma (HCC) microenvironment mediated NK cellular exhaustion, high expression of PDHK1 and reduced cytokine secretion. We discovered that the PDHK1 inhibitor DCA enhances the activity and function of exhausted NK cells infiltrating the tumor microenvironment. Furthermore, in a subcutaneous HCC mouse model, DCA combined with NK cell treatment resulted in retarding cancer progression. This study indicates the potential of DCA in rescuing NK cell exhaustion and eliciting anti-tumor immunity.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142004820","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Separable cell cycle arrest and immune response elicited through pharmacological CDK4/6 and MEK inhibition in RASmut disease models. 在 RASmut 疾病模型中通过药理 CDK4/6 和 MEK 抑制引起可分离的细胞周期停滞和免疫反应。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-16 DOI: 10.1158/1535-7163.MCT-24-0369
Jin Wu, Jianxin Wang, Thomas N O'Connor, Stephanie L Tzetzo, Katerina V Gurova, Erik S Knudsen, Agnieszka K Witkiewicz

The combination of CDK4/6 and MEK inhibition as a therapeutic strategy has shown promise in various cancer models, particularly in those harboring RAS mutations. An initial high-throughput drug screen identified a high synergy between the CDK4/6 inhibitor palbociclib and the MEK inhibitor trametinib when used in combination in soft tissue sarcomas. In RAS mutant models, combination treatment with palbociclib and trametinib induced significant G1 cell cycle arrest, resulting in a marked reduction in cell proliferation and growth. CRISPR-mediated RB1 depletion resulted in a decreased response to CDK4/6 and MEK inhibition, which was validated in both cell culture and xenograft models. Beyond its cell cycle inhibitory effects, pathway enrichment analysis revealed the robust activation of interferon pathways upon CDK4/6 and MEK inhibition. This induction of gene expression was associated with the upregulation of retroviral elements. The TBK1(TANK-binding kinase 1) inhibitor GSK8612 selectively blocked the induction of interferon-related genes induced by palbociclib and trametinib treatment, and highlighted the separable epigenetic responses elicited by combined CDK4/6 and MEK inhibition. Together, these findings provide key mechanistic insights into the therapeutic potential of CDK4/6 and MEK inhibition in soft tissue sarcoma.

CDK4/6和MEK抑制剂联合作为一种治疗策略已在各种癌症模型中显示出前景,尤其是在携带RAS突变的癌症模型中。最初的高通量药物筛选发现,CDK4/6抑制剂palbociclib和MEK抑制剂曲美替尼联用治疗软组织肉瘤具有高度协同作用。在RAS突变模型中,palbociclib和曲美替尼的联合治疗可诱导G1细胞周期显著停滞,从而明显减少细胞的增殖和生长。CRISPR 介导的 RB1 缺失导致对 CDK4/6 和 MEK 抑制的反应减弱,这在细胞培养和异种移植模型中都得到了验证。除了细胞周期抑制作用外,通路富集分析还显示,CDK4/6和MEK抑制可显著激活干扰素通路。这种基因表达的诱导与逆转录病毒元件的上调有关。TBK1(TANK结合激酶1)抑制剂GSK8612选择性地阻断了palbociclib和曲美替尼治疗诱导的干扰素相关基因的诱导,并强调了CDK4/6和MEK联合抑制引起的可分离的表观遗传学反应。总之,这些发现为CDK4/6和MEK抑制在软组织肉瘤中的治疗潜力提供了重要的机理启示。
{"title":"Separable cell cycle arrest and immune response elicited through pharmacological CDK4/6 and MEK inhibition in RASmut disease models.","authors":"Jin Wu, Jianxin Wang, Thomas N O'Connor, Stephanie L Tzetzo, Katerina V Gurova, Erik S Knudsen, Agnieszka K Witkiewicz","doi":"10.1158/1535-7163.MCT-24-0369","DOIUrl":"10.1158/1535-7163.MCT-24-0369","url":null,"abstract":"<p><p>The combination of CDK4/6 and MEK inhibition as a therapeutic strategy has shown promise in various cancer models, particularly in those harboring RAS mutations. An initial high-throughput drug screen identified a high synergy between the CDK4/6 inhibitor palbociclib and the MEK inhibitor trametinib when used in combination in soft tissue sarcomas. In RAS mutant models, combination treatment with palbociclib and trametinib induced significant G1 cell cycle arrest, resulting in a marked reduction in cell proliferation and growth. CRISPR-mediated RB1 depletion resulted in a decreased response to CDK4/6 and MEK inhibition, which was validated in both cell culture and xenograft models. Beyond its cell cycle inhibitory effects, pathway enrichment analysis revealed the robust activation of interferon pathways upon CDK4/6 and MEK inhibition. This induction of gene expression was associated with the upregulation of retroviral elements. The TBK1(TANK-binding kinase 1) inhibitor GSK8612 selectively blocked the induction of interferon-related genes induced by palbociclib and trametinib treatment, and highlighted the separable epigenetic responses elicited by combined CDK4/6 and MEK inhibition. Together, these findings provide key mechanistic insights into the therapeutic potential of CDK4/6 and MEK inhibition in soft tissue sarcoma.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141988386","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Harnessing the Potential of FAP-IL-12mut TMEkine™ for Targeted and Enhanced Anti-tumor Responses. 利用 FAP-IL-12mut TMEkine™ 的潜力,有针对性地增强抗肿瘤反应。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-16 DOI: 10.1158/1535-7163.MCT-24-0125
Dahea Lee, Dongsu Kim, Donggeon Kim, Jisu Kang, Kiram Lee, Hyunji Lee, Yujin Yoon, Youngin Lee, Nahmju Kim, Byoung Chul Cho, Jihoon Chang, Byoung Chul Lee

While cancer immunotherapy has yielded encouraging outcomes in hematological malignancies, it has faced challenges in achieving the same level of effectiveness in numerous solid tumors, primarily because of the presence of immune-suppressive tumor microenvironments (TMEs). The immunosuppressive qualities of the TME have generated considerable interest, making it a focal point for treatments aimed at enhancing immune responses and inhibiting tumor progression. Fibroblast activation protein (FAP), an attractive candidate for targeted immunotherapy, is prominently expressed in the TME of various solid tumors. Interleukin-12 (IL-12), recognized as a key mediator of immune responses, has been explored as a potential candidate for cancer treatment. Nevertheless, initial efforts to administer IL-12 systemically demonstrated limited efficacy and notable side effects, emphasizing the necessity for innovation. To address these concerns, our molecules incorporated specific IL-12 mutations, called IL-12mut, which reduced toxicity. This study explored the therapeutic potential of the FAP-IL-12mut TMEkine™-a novel immunotherapeutic agent selectively engineered to target FAP-expressing cells in preclinical cancer models. Our preclinical results, conducted across diverse murine cancer models, demonstrated that FAP-IL-12mut significantly inhibits tumor growth, enhances immune cell infiltration, and promotes a shift toward a cytotoxic immune activation profile. These findings suggest that FAP-IL-12mut could offer effective cancer treatment strategies.

虽然癌症免疫疗法在血液恶性肿瘤中取得了令人鼓舞的成果,但在众多实体瘤中取得同样的疗效却面临着挑战,这主要是因为存在免疫抑制性肿瘤微环境(TME)。肿瘤微环境的免疫抑制特性引起了人们的极大兴趣,使其成为旨在增强免疫反应和抑制肿瘤进展的治疗方法的焦点。成纤维细胞活化蛋白(FAP)是一种有吸引力的候选靶向免疫疗法,在各种实体瘤的 TME 中都有显著表达。白细胞介素-12(IL-12)被认为是免疫反应的关键介质,一直被视为癌症治疗的潜在候选药物。然而,最初对 IL-12 进行系统给药的努力显示出有限的疗效和显著的副作用,这强调了创新的必要性。为了解决这些问题,我们的分子加入了特定的 IL-12 突变,称为 IL-12mut,从而降低了毒性。本研究探索了 FAP-IL-12mut TMEkine™ 的治疗潜力--这是一种新型免疫治疗剂,经过选择性设计,可在临床前癌症模型中靶向 FAP 表达细胞。我们在各种小鼠癌症模型中进行的临床前研究结果表明,FAP-IL-12mut 能显著抑制肿瘤生长,增强免疫细胞浸润,并促进向细胞毒性免疫激活模式转变。这些研究结果表明,FAP-IL-12mut 可提供有效的癌症治疗策略。
{"title":"Harnessing the Potential of FAP-IL-12mut TMEkine™ for Targeted and Enhanced Anti-tumor Responses.","authors":"Dahea Lee, Dongsu Kim, Donggeon Kim, Jisu Kang, Kiram Lee, Hyunji Lee, Yujin Yoon, Youngin Lee, Nahmju Kim, Byoung Chul Cho, Jihoon Chang, Byoung Chul Lee","doi":"10.1158/1535-7163.MCT-24-0125","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0125","url":null,"abstract":"<p><p>While cancer immunotherapy has yielded encouraging outcomes in hematological malignancies, it has faced challenges in achieving the same level of effectiveness in numerous solid tumors, primarily because of the presence of immune-suppressive tumor microenvironments (TMEs). The immunosuppressive qualities of the TME have generated considerable interest, making it a focal point for treatments aimed at enhancing immune responses and inhibiting tumor progression. Fibroblast activation protein (FAP), an attractive candidate for targeted immunotherapy, is prominently expressed in the TME of various solid tumors. Interleukin-12 (IL-12), recognized as a key mediator of immune responses, has been explored as a potential candidate for cancer treatment. Nevertheless, initial efforts to administer IL-12 systemically demonstrated limited efficacy and notable side effects, emphasizing the necessity for innovation. To address these concerns, our molecules incorporated specific IL-12 mutations, called IL-12mut, which reduced toxicity. This study explored the therapeutic potential of the FAP-IL-12mut TMEkine™-a novel immunotherapeutic agent selectively engineered to target FAP-expressing cells in preclinical cancer models. Our preclinical results, conducted across diverse murine cancer models, demonstrated that FAP-IL-12mut significantly inhibits tumor growth, enhances immune cell infiltration, and promotes a shift toward a cytotoxic immune activation profile. These findings suggest that FAP-IL-12mut could offer effective cancer treatment strategies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141988384","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting CD33+ Acute Myeloid Leukemia with GLK-33, a Lintuzumab-Auristatin Conjugate with a Wide Therapeutic Window. 用GLK-33靶向CD33+急性髓性白血病,GLK-33是一种具有宽治疗窗口期的林妥珠单抗-阿瑞斯汀共轭物。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/1535-7163.MCT-23-0720
Tero Satomaa, Henna Pynnönen, Olli Aitio, Jukka O Hiltunen, Virve Pitkänen, Tuula Lähteenmäki, Titta Kotiranta, Annamari Heiskanen, Anna-Liisa Hänninen, Ritva Niemelä, Jari Helin, Heikki Kuusanmäki, Ida Vänttinen, Ramji Rathod, Anni I Nieminen, Emrah Yatkin, Caroline A Heckman, Mika Kontro, Juhani Saarinen

CD33 (Siglec-3) is a cell surface receptor expressed in approximately 90% of acute myeloid leukemia (AML) blasts, making it an attractive target for therapy of AML. Although previous CD33-targeting antibody-drug conjugates (ADC) like gemtuzumab ozogamicin (GO, Mylotarg) have shown efficacy in AML treatment, they have suffered from toxicity and narrow therapeutic window. This study aimed to develop a novelADCwith improved tolerability and a wider therapeutic window. GLK-33 consists of the anti-CD33 antibody lintuzumab and eight mavg-MMAU auristatin linkerpayloads per antibody. The experimental methods included testing in cell cultures, patient-derived samples, mouse xenograft models, and rat toxicology studies. GLK-33 exhibited remarkable efficacy in reducing cell viability within CD33-positive leukemia cell lines and primary AML samples. Notably, GLK-33 demonstrated antitumor activity at single dose as low as 300 mg/kg in mice, while maintaining tolerability at single dose of 20 to 30 mg/kg in rats. In contrast with both GO and lintuzumab vedotin, GLK-33 exhibited a wide therapeutic window and activity against multidrug-resistant cells. The development of GLK-33 addresses the limitations of previous ADCs, offering a wider therapeutic window, improved tolerability, and activity against drug-resistant leukemia cells. These findings encourage further exploration of GLK-33 in AML through clinical trials.

CD33(Siglec-3)是一种细胞表面受体,在大约 90% 的急性髓性白血病(AML)血块中都有表达,因此成为治疗急性髓性白血病(AML)的一个极具吸引力的靶点。虽然之前的CD33靶向抗体-药物共轭物(ADCs),如吉妥珠单抗-奥佐米星(GO,Mylotarg)在急性髓性白血病治疗中显示出了疗效,但它们也存在毒性和治疗窗口狭窄的问题。这项研究旨在开发一种耐受性更好、治疗窗口期更宽的新型 ADC。GLK-33 由抗 CD33 抗体林妥珠单抗和每个抗体的 8 个 mavg-MMAU auristatin 连接子负载组成。实验方法包括在细胞培养物、患者样本、小鼠异种移植模型和大鼠毒理学研究中进行测试。GLK-33 在降低 CD33 阳性白血病细胞系和原发性急性髓细胞白血病样本中的细胞活力方面表现出显著的功效。值得注意的是,GLK-33 在小鼠体内的单次剂量低至 300 微克/千克时就表现出抗肿瘤活性,而在大鼠体内的单次剂量为 20-30 毫克/千克时也能保持耐受性。与 GO 和 Lintuzumab vedotin 相比,GLK-33 的治疗窗口期较宽,而且对耐多药细胞具有活性。GLK-33 的开发解决了以往 ADC 的局限性,提供了更宽的治疗窗口期、更好的耐受性以及对耐药白血病细胞的活性。这些发现鼓励人们通过临床试验进一步探索 GLK-33 在急性髓细胞白血病中的应用。
{"title":"Targeting CD33+ Acute Myeloid Leukemia with GLK-33, a Lintuzumab-Auristatin Conjugate with a Wide Therapeutic Window.","authors":"Tero Satomaa, Henna Pynnönen, Olli Aitio, Jukka O Hiltunen, Virve Pitkänen, Tuula Lähteenmäki, Titta Kotiranta, Annamari Heiskanen, Anna-Liisa Hänninen, Ritva Niemelä, Jari Helin, Heikki Kuusanmäki, Ida Vänttinen, Ramji Rathod, Anni I Nieminen, Emrah Yatkin, Caroline A Heckman, Mika Kontro, Juhani Saarinen","doi":"10.1158/1535-7163.MCT-23-0720","DOIUrl":"10.1158/1535-7163.MCT-23-0720","url":null,"abstract":"<p><p>CD33 (Siglec-3) is a cell surface receptor expressed in approximately 90% of acute myeloid leukemia (AML) blasts, making it an attractive target for therapy of AML. Although previous CD33-targeting antibody-drug conjugates (ADC) like gemtuzumab ozogamicin (GO, Mylotarg) have shown efficacy in AML treatment, they have suffered from toxicity and narrow therapeutic window. This study aimed to develop a novelADCwith improved tolerability and a wider therapeutic window. GLK-33 consists of the anti-CD33 antibody lintuzumab and eight mavg-MMAU auristatin linkerpayloads per antibody. The experimental methods included testing in cell cultures, patient-derived samples, mouse xenograft models, and rat toxicology studies. GLK-33 exhibited remarkable efficacy in reducing cell viability within CD33-positive leukemia cell lines and primary AML samples. Notably, GLK-33 demonstrated antitumor activity at single dose as low as 300 mg/kg in mice, while maintaining tolerability at single dose of 20 to 30 mg/kg in rats. In contrast with both GO and lintuzumab vedotin, GLK-33 exhibited a wide therapeutic window and activity against multidrug-resistant cells. The development of GLK-33 addresses the limitations of previous ADCs, offering a wider therapeutic window, improved tolerability, and activity against drug-resistant leukemia cells. These findings encourage further exploration of GLK-33 in AML through clinical trials.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1073-1083"},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140336249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of RGT-018: a Potent, Selective and Orally Bioavailable SOS1 Inhibitor for KRAS-driven Cancers. 发现 RGT-018:一种针对 KRAS 驱动型癌症的强效、选择性和口服生物可用性 SOS1 抑制剂。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/1535-7163.MCT-24-0049
Fei Xiao, Kailiang Wang, Xinjuan Wang, Huijuan Li, Zhilong Hu, Xiaoming Ren, Wei Huang, Teng Feng, Lili Yao, Jing Lin, Chunlai Li, Zhuanzhuan Zhang, Liufeng Mei, Xiaotian Zhu, Wenge Zhong, Zhi Xie

KRAS is the most frequently dysregulated oncogene with high prevalence in NSCLC, colorectal cancer, and pancreatic cancer. FDA-approved sotorasib and adagrasib provide breakthrough therapies for cancer patients with KRASG12C mutation. However, there is still high unmet medical need for new agents targeting broader KRAS-driven tumors. An emerging and promising opportunity is to develop a pan KRAS inhibitor by suppressing the upstream protein SOS1. SOS1 is a key activator of KRAS and facilitates the conversion of GDP-bound KRAS state to GTP-bound KRAS state. Binding to its catalytic domain, small molecule SOS1 inhibitor has demonstrated the ability to suppress KRAS activation and cancer cell proliferation. RGT-018, a potent and selective SOS1 inhibitor, was identified with optimal drug-like properties. In vitro, RGT-018 blocked the interaction of KRAS:SOS1 with single digit nM potency and is highly selective against SOS2. RGT-018 inhibited KRAS signaling and the proliferation of a broad spectrum of KRAS-driven cancer cells as a single agent in vitro. Further enhanced anti-proliferation activity was observed when RGT-018 was combined with MEK, KRASG12C, EGFR or CDK4/6 inhibitors. Oral administration of RGT-018 inhibited tumor growth and suppressed KRAS signaling in tumor xenografts in vivo. Combination with MEK or KRASG12C inhibitors led to significant tumor regression. Furthermore, RGT-018 overcame the resistance to the approved KRASG12C inhibitors caused by clinically acquired KRAS mutations either as a single agent or in combination. RGT-018 displayed promising pharmacological properties for combination with targeted agents to treat a broader KRAS-driven patient population.

KRAS 是最常见的失调癌基因,在 NSCLC、结直肠癌和胰腺癌中发病率很高。美国 FDA 批准的 sotorasib 和 adagrasib 为 KRASG12C 突变的癌症患者提供了突破性疗法。然而,针对更广泛的 KRAS 驱动肿瘤的新药仍有大量医疗需求未得到满足。通过抑制上游蛋白 SOS1 开发泛 KRAS 抑制剂是一个新兴且前景广阔的机会。SOS1 是 KRAS 的关键激活剂,能促进 GDP 结合的 KRAS 状态向 GTP 结合的 KRAS 状态转化。小分子 SOS1 抑制剂与它的催化结构域结合,具有抑制 KRAS 活化和癌细胞增殖的能力。RGT-018 是一种强效的选择性 SOS1 抑制剂,具有最佳的类药物特性。在体外,RGT-018 以个位数 nM 的效力阻断 KRAS 与 SOS1 的相互作用,并对 SOS2 具有高度选择性。在体外,RGT-018 作为一种单药抑制了 KRAS 信号传导和多种 KRAS 驱动的癌细胞的增殖。当 RGT-018 与 MEK、KRASG12C、表皮生长因子受体(EGFR)或 CDK4/6 抑制剂联用时,抗增殖活性进一步增强。口服 RGT-018 可抑制肿瘤生长并抑制体内肿瘤异种移植物的 KRAS 信号传导。与 MEK 或 KRASG12C 抑制剂联用可显著抑制肿瘤生长。此外,RGT-018 作为单药或联合用药都能克服临床获得性 KRAS 突变导致的对已批准的 KRASG12C 抑制剂的耐药性。RGT-018显示出与靶向药物联合治疗更广泛的KRAS驱动患者群体的良好药理特性。
{"title":"Discovery of RGT-018: a Potent, Selective and Orally Bioavailable SOS1 Inhibitor for KRAS-driven Cancers.","authors":"Fei Xiao, Kailiang Wang, Xinjuan Wang, Huijuan Li, Zhilong Hu, Xiaoming Ren, Wei Huang, Teng Feng, Lili Yao, Jing Lin, Chunlai Li, Zhuanzhuan Zhang, Liufeng Mei, Xiaotian Zhu, Wenge Zhong, Zhi Xie","doi":"10.1158/1535-7163.MCT-24-0049","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0049","url":null,"abstract":"<p><p>KRAS is the most frequently dysregulated oncogene with high prevalence in NSCLC, colorectal cancer, and pancreatic cancer. FDA-approved sotorasib and adagrasib provide breakthrough therapies for cancer patients with KRASG12C mutation. However, there is still high unmet medical need for new agents targeting broader KRAS-driven tumors. An emerging and promising opportunity is to develop a pan KRAS inhibitor by suppressing the upstream protein SOS1. SOS1 is a key activator of KRAS and facilitates the conversion of GDP-bound KRAS state to GTP-bound KRAS state. Binding to its catalytic domain, small molecule SOS1 inhibitor has demonstrated the ability to suppress KRAS activation and cancer cell proliferation. RGT-018, a potent and selective SOS1 inhibitor, was identified with optimal drug-like properties. In vitro, RGT-018 blocked the interaction of KRAS:SOS1 with single digit nM potency and is highly selective against SOS2. RGT-018 inhibited KRAS signaling and the proliferation of a broad spectrum of KRAS-driven cancer cells as a single agent in vitro. Further enhanced anti-proliferation activity was observed when RGT-018 was combined with MEK, KRASG12C, EGFR or CDK4/6 inhibitors. Oral administration of RGT-018 inhibited tumor growth and suppressed KRAS signaling in tumor xenografts in vivo. Combination with MEK or KRASG12C inhibitors led to significant tumor regression. Furthermore, RGT-018 overcame the resistance to the approved KRASG12C inhibitors caused by clinically acquired KRAS mutations either as a single agent or in combination. RGT-018 displayed promising pharmacological properties for combination with targeted agents to treat a broader KRAS-driven patient population.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141860321","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of Monovalent Direct Degraders of BRD4 that Act via the Recruitment of DCAF11. 发现通过招募 DCAF11 起作用的 BRD4 单价直接降解剂
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-05 DOI: 10.1158/1535-7163.MCT-24-0219
Gregory S Parker, Julia I Toth, Sarah Fish, Gabrielle Blanco, Taylor Kampert, Xiaoming Li, Linette Yang, Craig R Stumpf, Kenneth Steadman, Aleksandar Jamborcic, Stephen Chien, Elizabeth Daniele, Alejandro Dearie, Geoffray Leriche, Simon Bailey, Peggy A Thompson

Targeted protein degradation (TPD) using the ubiquitin proteasome system (UPS) is a rapidly growing drug discovery modality to eliminate pathogenic proteins. Strategies for TPD have focused on heterobifunctional degraders that often suffer from poor drug-like properties, and molecular glues that rely on serendipitous discovery. Monovalent "direct" degraders represent an alternative approach, in which small molecules bind to a target protein and induce degradation of that protein through the recruitment of an E3 ligase complex. Using an ultra-high throughput cell-based screening platform, degraders of the bromodomain extraterminal protein BRD4 were identified and optimized to yield a lead compound, PLX-3618. In this paper, we demonstrate that PLX-3618 elicited UPS-mediated selective degradation of BRD4, resulting in potent antitumor activity in vitro and in vivo. Characterization of the degradation mechanism identified DCAF11 as the E3 ligase required for PLX-3618-mediated degradation of BRD4. Protein-protein interaction studies verified a BRD4:PLX-3618:DCAF11 ternary complex, and mutational studies provided further insights into the DCAF11-mediated degradation mechanism. Collectively, these results demonstrate the discovery and characterization of a novel small molecule that selectively degrades BRD4 through the recruitment of the E3 substrate receptor, DCAF11, and promotes potent antitumor activity in vivo.

利用泛素蛋白酶体系统(UPS)进行靶向蛋白质降解(TPD)是一种快速发展的药物发现方式,可用于消除致病蛋白质。TPD的策略主要集中在异功能降解剂(通常具有较差的类药物特性)和依赖偶然发现的分子粘合剂上。单价 "直接 "降解剂代表了另一种方法,即小分子与目标蛋白质结合,通过招募 E3 连接酶复合物诱导该蛋白质降解。利用基于细胞的超高通量筛选平台,我们鉴定并优化了溴基链外端蛋白 BRD4 的降解剂,从而获得了先导化合物 PLX-3618。在本文中,我们证明了 PLX-3618 可诱导 UPS 介导的 BRD4 选择性降解,从而在体外和体内产生强效抗肿瘤活性。对降解机制的鉴定发现,DCAF11是PLX-3618介导的BRD4降解所需的E3连接酶。蛋白-蛋白相互作用研究验证了BRD4:PLX-3618:DCAF11三元复合物,突变研究进一步揭示了DCAF11介导的降解机制。总之,这些研究结果表明发现并鉴定了一种新型小分子,它能通过招募 E3 底物受体 DCAF11 选择性地降解 BRD4,并在体内促进有效的抗肿瘤活性。
{"title":"Discovery of Monovalent Direct Degraders of BRD4 that Act via the Recruitment of DCAF11.","authors":"Gregory S Parker, Julia I Toth, Sarah Fish, Gabrielle Blanco, Taylor Kampert, Xiaoming Li, Linette Yang, Craig R Stumpf, Kenneth Steadman, Aleksandar Jamborcic, Stephen Chien, Elizabeth Daniele, Alejandro Dearie, Geoffray Leriche, Simon Bailey, Peggy A Thompson","doi":"10.1158/1535-7163.MCT-24-0219","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0219","url":null,"abstract":"<p><p>Targeted protein degradation (TPD) using the ubiquitin proteasome system (UPS) is a rapidly growing drug discovery modality to eliminate pathogenic proteins. Strategies for TPD have focused on heterobifunctional degraders that often suffer from poor drug-like properties, and molecular glues that rely on serendipitous discovery. Monovalent \"direct\" degraders represent an alternative approach, in which small molecules bind to a target protein and induce degradation of that protein through the recruitment of an E3 ligase complex. Using an ultra-high throughput cell-based screening platform, degraders of the bromodomain extraterminal protein BRD4 were identified and optimized to yield a lead compound, PLX-3618. In this paper, we demonstrate that PLX-3618 elicited UPS-mediated selective degradation of BRD4, resulting in potent antitumor activity in vitro and in vivo. Characterization of the degradation mechanism identified DCAF11 as the E3 ligase required for PLX-3618-mediated degradation of BRD4. Protein-protein interaction studies verified a BRD4:PLX-3618:DCAF11 ternary complex, and mutational studies provided further insights into the DCAF11-mediated degradation mechanism. Collectively, these results demonstrate the discovery and characterization of a novel small molecule that selectively degrades BRD4 through the recruitment of the E3 substrate receptor, DCAF11, and promotes potent antitumor activity in vivo.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF13"},"PeriodicalIF":5.3,"publicationDate":"2024-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141534847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pretargeted Radioimmunotherapy with the Novel Anti-oxMIF/HSG Bispecific Antibody ON105 Results in Significant Tumor Regression in Murine Models of Cancer. 新型抗oxMIF/HSG双特异性抗体ON105的预靶向放射免疫疗法可使小鼠癌症模型中的肿瘤显著消退
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-04 DOI: 10.1158/1535-7163.MCT-24-0083
Alejandro A Puchol Tarazona, Alexander Schinagl, Irina Mirkina, Gregor Rossmueller, Randolf J Kerschbaumer, Friedmund Bachmann, Michael Thiele

Radioimmunotherapy (RIT) uses monoclonal antibodies to deliver radionuclides to cancer cells or the tumor microenvironment and has shown promise in treating localized and diffuse tumors. Although RIT agents have gained FDA/EMA approval for certain hematologic malignancies, effectiveness of RIT in treating solid tumors remains limited. In this study, we present PreTarg-it®, a novel approach for pretargeted RIT, providing optimized delivery of payloads in a two-step regimen. The effectiveness of PreTarg-it® is demonstrated by a powerful combination of ON105, a novel bispecific antibody against both oxidized macrophage migration inhibitory factor (oxMIF) and the histamine-succinyl-glycyl (HSG) hapten, as the first component and the radioactively labeled DOTA-di-HSG peptide as the second component in murine models of cancer. Mice bearing either subcutaneous mouse colorectal CT26 or human pancreatic CFPAC-1 tumors received an i.v. injection of ON105. After ON105 had accumulated in the tumor and cleared from circulation to approximately 1% to 3% of its peak concentration, 177Lu-DOTA-di-HSG peptide was administered. A single PreTarg-it® treatment cycle resulted in tumor regression when mice bearing CT26 tumors were given the highest treatment dose with a pretargeting delay of 3 days. Administered with a 5-day interval, the highest dose arrested tumor growth in both CT26 syngrafts and CFPAC-1 xenografts. In all cases, the highest treatment dose resulted in 100% survival at the study endpoint, whereas the control cohorts showed 0% and 60% survival in the CT26 and CFPAC-1 models, respectively. Therefore, PreTarg-it® holds potential as a novel and potent therapy for patients with hard-to-treat solid tumors, such as pancreatic cancer, as well as those with late-stage malignancies.

放射免疫疗法(RIT)使用单克隆抗体向癌细胞或肿瘤微环境释放放射性核素,在治疗局部和弥漫性肿瘤方面前景看好。虽然 RIT 药剂已获得 FDA/EMA 批准用于治疗某些血液系统恶性肿瘤,但 RIT 治疗实体瘤的效果仍然有限。在本研究中,我们介绍了一种用于预靶向 RIT 的新方法 PreTarg-it®,它通过两步疗法优化了有效载荷的递送。在小鼠癌症模型中,以 ON105(一种针对氧化巨噬细胞迁移抑制因子(oxMIF)和组胺-琥珀酰-甘氨酰(HSG)合肽的新型双特异性抗体)为第一成分,以放射性标记的 DOTA-di-HSG 肽为第二成分的强效组合证明了 PreTarg-it® 的有效性。皮下注射ON105给携带小鼠结直肠CT26肿瘤或人胰腺CFPAC-1肿瘤的小鼠。ON105在肿瘤中蓄积并从血液循环中清除至其峰值浓度的约1%至3%后,再注射177Lu-DOTA-di-HSG肽。对携带 CT26 肿瘤的小鼠施用最高剂量的 PreTarg-it® 并延迟 3 天进行预靶向治疗,一个 PreTarg-it® 治疗周期就能使肿瘤消退。在间隔 5 天的情况下,最高剂量可阻止 CT26 系统移植物和 CFPAC-1 异种移植物的肿瘤生长。在所有情况下,最高治疗剂量都能使研究终点的存活率达到 100%,而 CT26 和 CFPAC-1 模型的对照组存活率分别为 0% 和 60%。因此,PreTarg-it®有望成为胰腺癌等难以治疗的实体瘤患者以及晚期恶性肿瘤患者的一种新型强效疗法。
{"title":"Pretargeted Radioimmunotherapy with the Novel Anti-oxMIF/HSG Bispecific Antibody ON105 Results in Significant Tumor Regression in Murine Models of Cancer.","authors":"Alejandro A Puchol Tarazona, Alexander Schinagl, Irina Mirkina, Gregor Rossmueller, Randolf J Kerschbaumer, Friedmund Bachmann, Michael Thiele","doi":"10.1158/1535-7163.MCT-24-0083","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0083","url":null,"abstract":"<p><p>Radioimmunotherapy (RIT) uses monoclonal antibodies to deliver radionuclides to cancer cells or the tumor microenvironment and has shown promise in treating localized and diffuse tumors. Although RIT agents have gained FDA/EMA approval for certain hematologic malignancies, effectiveness of RIT in treating solid tumors remains limited. In this study, we present PreTarg-it®, a novel approach for pretargeted RIT, providing optimized delivery of payloads in a two-step regimen. The effectiveness of PreTarg-it® is demonstrated by a powerful combination of ON105, a novel bispecific antibody against both oxidized macrophage migration inhibitory factor (oxMIF) and the histamine-succinyl-glycyl (HSG) hapten, as the first component and the radioactively labeled DOTA-di-HSG peptide as the second component in murine models of cancer. Mice bearing either subcutaneous mouse colorectal CT26 or human pancreatic CFPAC-1 tumors received an i.v. injection of ON105. After ON105 had accumulated in the tumor and cleared from circulation to approximately 1% to 3% of its peak concentration, 177Lu-DOTA-di-HSG peptide was administered. A single PreTarg-it® treatment cycle resulted in tumor regression when mice bearing CT26 tumors were given the highest treatment dose with a pretargeting delay of 3 days. Administered with a 5-day interval, the highest dose arrested tumor growth in both CT26 syngrafts and CFPAC-1 xenografts. In all cases, the highest treatment dose resulted in 100% survival at the study endpoint, whereas the control cohorts showed 0% and 60% survival in the CT26 and CFPAC-1 models, respectively. Therefore, PreTarg-it® holds potential as a novel and potent therapy for patients with hard-to-treat solid tumors, such as pancreatic cancer, as well as those with late-stage malignancies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF11"},"PeriodicalIF":5.3,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141498501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ultrasensitive Response Explains the Benefit of Combination Chemotherapy Despite Drug Antagonism. 超敏反应解释了联合化疗在药物拮抗作用下仍能获益的原因。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.1158/1535-7163.MCT-23-0642
Sarah C Patterson, Amy E Pomeroy, Adam C Palmer

Most aggressive lymphomas are treated with combination chemotherapy, commonly as multiple cycles of concurrent drug administration. Concurrent administration is in theory optimal when combination therapies have synergistic (more than additive) drug interactions. We investigated pharmacodynamic interactions in the standard 4-drug "CHOP" regimen in peripheral T-cell lymphoma (PTCL) cell lines and found that CHOP consistently exhibits antagonism and not synergy. We tested whether staggered treatment schedules could improve tumor cell kill by avoiding antagonism, using in vitro models of concurrent or staggered treatments. Surprisingly, we observed that tumor cell kill is maximized by concurrent drug administration despite antagonistic drug-drug interactions. We propose that an ultrasensitive dose response, as described in radiology by the linear-quadratic (LQ) model, can reconcile these seemingly contradictory experimental observations. The LQ model describes the relationship between cell survival and dose, and in radiology has identified scenarios favoring hypofractionated radiotherapy-the administration of fewer large doses rather than multiple smaller doses. Specifically, hypofractionated treatment can be favored when cells require an accumulation of DNA damage, rather than a "single hit," to die. By adapting the LQ model to combination chemotherapy and accounting for tumor heterogeneity, we find that tumor cell kill is maximized by concurrent administration of multiple drugs, even when chemotherapies have antagonistic interactions. Thus, our study identifies a new mechanism by which combination chemotherapy can be clinically beneficial that is not contingent on positive drug-drug interactions.

大多数侵袭性淋巴瘤都采用联合化疗,通常是多个周期同时用药。理论上,当联合疗法具有协同(大于相加)的药物相互作用时,同时给药是最佳选择。我们研究了外周 T 细胞淋巴瘤(PTCL)细胞系中标准四药 "CHOP "方案的药效学相互作用,发现 CHOP 始终表现出拮抗作用而非协同作用。我们利用同时或交错治疗的体外模型,测试了交错治疗计划是否能避免拮抗作用,从而提高对肿瘤细胞的杀伤力。令人惊讶的是,我们观察到,尽管药物与药物之间存在拮抗作用,但同时给药能最大限度地杀伤肿瘤细胞。我们提出,放射学中的线性-四次方(LQ)模型所描述的超灵敏剂量反应可以调和这些看似矛盾的实验观察结果。LQ 模型描述了细胞存活与剂量之间的关系,并在放射学中确定了有利于低分次放射治疗的方案--施用较少的大剂量而不是多个较小剂量。具体来说,当细胞需要DNA损伤的累积而不是 "单次打击 "才能死亡时,低分次治疗就会受到青睐。通过将 LQ 模型应用于联合化疗并考虑肿瘤的异质性,我们发现,即使化疗药物之间存在拮抗作用,同时使用多种药物也能最大限度地杀死肿瘤细胞。因此,我们的研究发现了联合化疗对临床有益的新机制,而这种机制并不取决于药物间的正相互作用。
{"title":"Ultrasensitive Response Explains the Benefit of Combination Chemotherapy Despite Drug Antagonism.","authors":"Sarah C Patterson, Amy E Pomeroy, Adam C Palmer","doi":"10.1158/1535-7163.MCT-23-0642","DOIUrl":"10.1158/1535-7163.MCT-23-0642","url":null,"abstract":"<p><p>Most aggressive lymphomas are treated with combination chemotherapy, commonly as multiple cycles of concurrent drug administration. Concurrent administration is in theory optimal when combination therapies have synergistic (more than additive) drug interactions. We investigated pharmacodynamic interactions in the standard 4-drug \"CHOP\" regimen in peripheral T-cell lymphoma (PTCL) cell lines and found that CHOP consistently exhibits antagonism and not synergy. We tested whether staggered treatment schedules could improve tumor cell kill by avoiding antagonism, using in vitro models of concurrent or staggered treatments. Surprisingly, we observed that tumor cell kill is maximized by concurrent drug administration despite antagonistic drug-drug interactions. We propose that an ultrasensitive dose response, as described in radiology by the linear-quadratic (LQ) model, can reconcile these seemingly contradictory experimental observations. The LQ model describes the relationship between cell survival and dose, and in radiology has identified scenarios favoring hypofractionated radiotherapy-the administration of fewer large doses rather than multiple smaller doses. Specifically, hypofractionated treatment can be favored when cells require an accumulation of DNA damage, rather than a \"single hit,\" to die. By adapting the LQ model to combination chemotherapy and accounting for tumor heterogeneity, we find that tumor cell kill is maximized by concurrent administration of multiple drugs, even when chemotherapies have antagonistic interactions. Thus, our study identifies a new mechanism by which combination chemotherapy can be clinically beneficial that is not contingent on positive drug-drug interactions.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"995-1009"},"PeriodicalIF":5.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11219261/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140288567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Novel ATR Inhibitor M1774 Induces Replication Protein Overexpression and Broad Synergy with DNA-targeted Anticancer Drugs. 新型 ATR 抑制剂 M1774 可诱导复制蛋白过表达,并与 DNA 靶向抗癌药物产生广泛的协同作用。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.1158/1535-7163.MCT-23-0402
Ukhyun Jo, Yasuhiro Arakawa, Astrid Zimmermann, Daiki Taniyama, Makito Mizunuma, Lisa M Jenkins, Tapan Maity, Suresh Kumar, Frank T Zenke, Naoko Takebe, Yves Pommier

Ataxia telangiectasia and Rad3-related (ATR) checkpoint kinase inhibitors are in clinical trials. Here we explored the molecular pharmacology and therapeutic combination strategies of the oral ATR inhibitor M1774 (Tuvusertib) with DNA-damaging agents (DDA). As single agent, M1774 suppressed cancer cell viability at nanomolar concentrations, showing greater activity than ceralasertib and berzosertib, but less potency than gartisertib and elimusertib in the small cell lung cancer H146, H82, and DMS114 cell lines. M1774 also efficiently blocked the activation of the ATR-CHK1 checkpoint pathway caused by replication stress induced by TOP1 inhibitors. Combination with non-toxic dose of M1774 enhanced TOP1 inhibitor-induced cancer cell death by enabling unscheduled replication upon replicative damage, thereby increasing genome instability. Tandem mass tag-based quantitative proteomics uncovered that M1774, in the presence of DDA, forces the expression of proteins activating replication (CDC45) and G2-M progression (PLK1 and CCNB1). In particular, the fork protection complex proteins (TIMELESS and TIPIN) were enriched. Low dose of M1774 was found highly synergistic with a broad spectrum of clinical DDAs including TOP1 inhibitors (SN-38/irinotecan, topotecan, exatecan, and exatecan), the TOP2 inhibitor etoposide, cisplatin, the RNA polymerase II inhibitor lurbinectedin, and the PARP inhibitor talazoparib in various models including cancer cell lines, patient-derived organoids, and mouse xenograft models. Furthermore, we demonstrate that M1774 reverses chemoresistance to anticancer DDAs in cancer cells lacking SLFN11 expression, suggesting that SLFN11 can be utilized for patient selection in upcoming clinical trials.

共济失调性端粒增生症和Rad3相关(ATR)检查点激酶抑制剂正处于临床试验阶段。在这里,我们探索了口服ATR抑制剂M1774(Tuvusertib)与DNA损伤剂(DDAs)的分子药理学和治疗组合策略。在小细胞肺癌 H146、H82 和 DMS114 细胞系中,作为单药,M1774 在纳摩尔浓度下抑制癌细胞活力,其活性高于 ceralasertib 和 berzosertib,但低于 gartisertib 和 elimusertib。M1774 还能有效阻断 TOP1 抑制剂诱导的复制压力引起的 ATR-CHK1 检查点通路的激活。与无毒性剂量的 M1774 结合使用,可以在复制损伤时实现非计划复制,从而增加基因组的不稳定性,从而增强 TOP1 抑制剂诱导的癌细胞死亡。基于串联质量标签(TMT)的定量蛋白质组学发现,M1774 在 DDA 存在的情况下,会迫使激活复制(CDC45)和 G2/M 进展(PLK1 和 CCNB1)的蛋白质表达。叉保护复合体蛋白(TIMELESS 和 TIPIN)的表达尤其丰富。在各种模型(包括癌细胞系、患者衍生的器官组织和小鼠异种移植模型)中,低剂量 M1774 与多种临床 DDAs(包括 TOP1 抑制剂(SN-38/irinotecan、topotecan、exatecan 和 exatecan)、TOP2 抑制剂依托泊苷、顺铂、RNA 聚合酶 II 抑制剂 lurbinectedin 和 PARP 抑制剂 talazoparib)具有高度协同作用。此外,我们还证明 M1774 可逆转缺乏 SLFN11 表达的癌细胞对抗癌 DDAs 的化疗耐药性,这表明 SLFN11 可用于即将开展的临床试验中的患者选择。
{"title":"The Novel ATR Inhibitor M1774 Induces Replication Protein Overexpression and Broad Synergy with DNA-targeted Anticancer Drugs.","authors":"Ukhyun Jo, Yasuhiro Arakawa, Astrid Zimmermann, Daiki Taniyama, Makito Mizunuma, Lisa M Jenkins, Tapan Maity, Suresh Kumar, Frank T Zenke, Naoko Takebe, Yves Pommier","doi":"10.1158/1535-7163.MCT-23-0402","DOIUrl":"10.1158/1535-7163.MCT-23-0402","url":null,"abstract":"<p><p>Ataxia telangiectasia and Rad3-related (ATR) checkpoint kinase inhibitors are in clinical trials. Here we explored the molecular pharmacology and therapeutic combination strategies of the oral ATR inhibitor M1774 (Tuvusertib) with DNA-damaging agents (DDA). As single agent, M1774 suppressed cancer cell viability at nanomolar concentrations, showing greater activity than ceralasertib and berzosertib, but less potency than gartisertib and elimusertib in the small cell lung cancer H146, H82, and DMS114 cell lines. M1774 also efficiently blocked the activation of the ATR-CHK1 checkpoint pathway caused by replication stress induced by TOP1 inhibitors. Combination with non-toxic dose of M1774 enhanced TOP1 inhibitor-induced cancer cell death by enabling unscheduled replication upon replicative damage, thereby increasing genome instability. Tandem mass tag-based quantitative proteomics uncovered that M1774, in the presence of DDA, forces the expression of proteins activating replication (CDC45) and G2-M progression (PLK1 and CCNB1). In particular, the fork protection complex proteins (TIMELESS and TIPIN) were enriched. Low dose of M1774 was found highly synergistic with a broad spectrum of clinical DDAs including TOP1 inhibitors (SN-38/irinotecan, topotecan, exatecan, and exatecan), the TOP2 inhibitor etoposide, cisplatin, the RNA polymerase II inhibitor lurbinectedin, and the PARP inhibitor talazoparib in various models including cancer cell lines, patient-derived organoids, and mouse xenograft models. Furthermore, we demonstrate that M1774 reverses chemoresistance to anticancer DDAs in cancer cells lacking SLFN11 expression, suggesting that SLFN11 can be utilized for patient selection in upcoming clinical trials.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"911-923"},"PeriodicalIF":5.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555614/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140102031","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Characteristics of Carcinoembryonic Antigen-Related Cell Adhesion Molecules and Their Relationship to Cancer. 癌胚抗原相关细胞粘附分子的特征及其与癌症的关系。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.1158/1535-7163.MCT-23-0461
Ru-Xue Ma, Jian-Rui Wei, Yan-Wei Hu

Carcinoembryonic antigen-related cell adhesion molecules (CEACAM), such as carcinoembryonic antigen (CEA) and the oncofetal glycoprotein family, are tumor markers. The CEACAMs consist of 12 different human CEACAMs and 5 different murine CEACAMs. The CEACAM family of proteins participates in multiple biological processes that include the immune response, angiogenesis, and cancer. CEACAMs play a significant role in cancer initiation and development. Increasing evidence suggests that family members may be new cancer biomarkers and targets in that CEACEAMs tend to be aberrantly expressed and therefore may have potential diagnostic and therapeutic importance. This review systematically summarizes the biogenesis, biological properties, and functions of CEACAMs, with a focus on their relationship with cancer and potential clinical application. As our knowledge of the relationships among CEACAMs and cancer increases, and as our understanding of the involved molecular mechanisms improves, new therapeutic strategies will evolve for cancer prevention and treatment of patients with cancer.

癌胚抗原相关细胞粘附分子(CEACAMs),如癌胚抗原(CEA)和胎盘糖蛋白家族,是肿瘤标志物。CEACAM 包括 12 种不同的人类 CEACAM 和 5 种不同的鼠类 CEACAM。CEACAM 蛋白家族参与多种生物过程,包括免疫反应、血管生成和癌症。CEACAM 在癌症的诱发和发展过程中发挥着重要作用。越来越多的证据表明,该家族成员可能是新的癌症生物标志物和靶标,因为 CEACEAMs 往往表达异常,因此可能具有潜在的诊断和治疗意义。本综述系统地总结了 CEACAMs 的生物发生、生物特性和功能,重点关注它们与癌症的关系以及潜在的临床应用。随着我们对 CEACAMs 与癌症之间关系的认识不断加深,以及对相关分子机制的理解不断提高,新的治疗策略将在癌症预防和癌症患者治疗方面得到发展。
{"title":"Characteristics of Carcinoembryonic Antigen-Related Cell Adhesion Molecules and Their Relationship to Cancer.","authors":"Ru-Xue Ma, Jian-Rui Wei, Yan-Wei Hu","doi":"10.1158/1535-7163.MCT-23-0461","DOIUrl":"10.1158/1535-7163.MCT-23-0461","url":null,"abstract":"<p><p>Carcinoembryonic antigen-related cell adhesion molecules (CEACAM), such as carcinoembryonic antigen (CEA) and the oncofetal glycoprotein family, are tumor markers. The CEACAMs consist of 12 different human CEACAMs and 5 different murine CEACAMs. The CEACAM family of proteins participates in multiple biological processes that include the immune response, angiogenesis, and cancer. CEACAMs play a significant role in cancer initiation and development. Increasing evidence suggests that family members may be new cancer biomarkers and targets in that CEACEAMs tend to be aberrantly expressed and therefore may have potential diagnostic and therapeutic importance. This review systematically summarizes the biogenesis, biological properties, and functions of CEACAMs, with a focus on their relationship with cancer and potential clinical application. As our knowledge of the relationships among CEACAMs and cancer increases, and as our understanding of the involved molecular mechanisms improves, new therapeutic strategies will evolve for cancer prevention and treatment of patients with cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"939-948"},"PeriodicalIF":5.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140137034","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","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
Molecular Cancer Therapeutics
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