首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
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":null,"pages":null},"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
Inhibition of SUMOylation Induces Adaptive Antitumor Immunity against Pancreatic Cancer through Multiple Effects on the Tumor Microenvironment. 抑制 SUMOylation 可通过对肿瘤微环境的多重影响诱导对胰腺癌的适应性抗肿瘤免疫力
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-16 DOI: 10.1158/1535-7163.MCT-23-0572
Suna Erdem, Hyojae James Lee, Jayanth Surya Narayanan Shankara Narayanan, Mohottige Don Neranjan Tharuka, Jorge De la Torre, Tianchen Ren, Yixuan Kuang, Tharindumala Abeywardana, Kevin Li, Allison J Berger, Andrew M Lowy, Rebekah R White, Yuan Chen

Improvement of outcome in patients with pancreatic ductal adenocarcinoma (PDAC) requires exploration of novel therapeutic targets. Thus far, most studies of PDAC therapies, including those inhibiting small ubiquitin-like modifications (SUMOylation), have focused on PDAC epithelial cell biology, yet SUMOylation occurs in a variety of cell types. The mechanisms by which SUMOylation impacts PDAC in the context of its tumor microenvironment are poorly understood. We used clinically relevant orthotopic PDAC mouse models to investigate the effect of SUMOylation inhibition using a specific, clinical-stage compound, TAK-981. In contrast to its inhibition of PDAC cell proliferation in vitro, the survival benefit conferred by TAK-981 in vivo is dependent on the presence of T cells, suggesting that induction of adaptive antitumor immunity is an important antitumor effect of SUMOylation inhibition in vivo. To understand how this adaptive antitumor immunity is promoted, we investigated how SUMOylation inhibition in vivo alters major cell types/subtypes and their communications in the PDAC tumor microenvironment by performing transcriptomic analyses at single-cell resolution, which allowed mapping of cells in our orthotopic mouse model to cells in human PDAC tumors based on gene expression profiles. Findings are further validated by flow cytometry, immunofluorescence, IHC, western blots, and qPCR. The single-cell transcriptome dataset provided here suggests several combination strategies to augment adaptive immune responses that are necessary for durable disease control in patients with PDAC.

改善胰腺导管腺癌(PDAC)患者的预后需要探索新的治疗靶点。迄今为止,大多数有关 PDAC 疗法(包括抑制泛素样小修饰(SUMOylation)的疗法)的研究都集中在 PDAC 上皮细胞生物学方面,但 SUMOylation 存在于多种细胞类型中。人们对 SUMOylation 在肿瘤微环境中影响 PDAC 的机制还知之甚少。我们利用与临床相关的正位 PDAC 小鼠模型,使用一种特定的临床阶段化合物 TAK-981 来研究 SUMOylation 抑制的效果。与体外抑制 PDAC 细胞增殖不同的是,TAK-981 在体内带来的生存益处依赖于 T 细胞的存在,这表明诱导适应性抗肿瘤免疫是体内 SUMO 抑制的重要抗肿瘤效应。为了了解这种适应性抗肿瘤免疫是如何促进的,我们研究了体内 SUMO 抑制是如何改变 PDAC 肿瘤微环境中的主要细胞类型/亚型及其交流的,方法是进行单细胞分辨率的转录组分析,从而根据基因表达谱将我们的小鼠正位模型中的细胞映射到人类 PDAC 肿瘤中的细胞。流式细胞术、免疫荧光、IHC、Western 印迹和 qPCR 进一步验证了研究结果。本文提供的单细胞转录组数据集提示了几种增强适应性免疫反应的组合策略,这些策略是 PDAC 患者持久控制疾病所必需的。
{"title":"Inhibition of SUMOylation Induces Adaptive Antitumor Immunity against Pancreatic Cancer through Multiple Effects on the Tumor Microenvironment.","authors":"Suna Erdem, Hyojae James Lee, Jayanth Surya Narayanan Shankara Narayanan, Mohottige Don Neranjan Tharuka, Jorge De la Torre, Tianchen Ren, Yixuan Kuang, Tharindumala Abeywardana, Kevin Li, Allison J Berger, Andrew M Lowy, Rebekah R White, Yuan Chen","doi":"10.1158/1535-7163.MCT-23-0572","DOIUrl":"10.1158/1535-7163.MCT-23-0572","url":null,"abstract":"<p><p>Improvement of outcome in patients with pancreatic ductal adenocarcinoma (PDAC) requires exploration of novel therapeutic targets. Thus far, most studies of PDAC therapies, including those inhibiting small ubiquitin-like modifications (SUMOylation), have focused on PDAC epithelial cell biology, yet SUMOylation occurs in a variety of cell types. The mechanisms by which SUMOylation impacts PDAC in the context of its tumor microenvironment are poorly understood. We used clinically relevant orthotopic PDAC mouse models to investigate the effect of SUMOylation inhibition using a specific, clinical-stage compound, TAK-981. In contrast to its inhibition of PDAC cell proliferation in vitro, the survival benefit conferred by TAK-981 in vivo is dependent on the presence of T cells, suggesting that induction of adaptive antitumor immunity is an important antitumor effect of SUMOylation inhibition in vivo. To understand how this adaptive antitumor immunity is promoted, we investigated how SUMOylation inhibition in vivo alters major cell types/subtypes and their communications in the PDAC tumor microenvironment by performing transcriptomic analyses at single-cell resolution, which allowed mapping of cells in our orthotopic mouse model to cells in human PDAC tumors based on gene expression profiles. Findings are further validated by flow cytometry, immunofluorescence, IHC, western blots, and qPCR. The single-cell transcriptome dataset provided here suggests several combination strategies to augment adaptive immune responses that are necessary for durable disease control in patients with PDAC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141988385","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":null,"pages":null},"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":null,"pages":null},"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
Novel inhibition of central carbon metabolism pathways by Rac and Cdc42 inhibitor MBQ-167 and paclitaxel. Rac和Cdc42抑制剂MBQ-167与紫杉醇对中枢碳代谢途径的新抑制作用。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/1535-7163.MCT-23-0803
Ailed M Cruz-Collazo, Olga Katsara, Nilmary Grafals-Ruiz, Jessica Colon Gonzalez, Stephanie Dorta-Estremera, Victor P Carlo, Nataliya Chorna, Robert J Schneider, Suranganie Dharmawardhane

Triple negative breast cancer (TNBC) represents a therapeutic challenge where standard chemotherapy is limited to paclitaxel. MBQ-167, a clinical stage small molecule inhibitor that targets Rac and Cdc42, inhibits tumor growth and metastasis in mouse models of TNBC. Herein, we investigated the efficacy of MBQ-167 in combination with paclitaxel in TNBC pre-clinical models, as a prelude to safety trials of this combination in advanced breast cancer patients. Individual MBQ-167 or combination therapy with paclitaxel was more effective at reducing TNBC cell viability and increasing apoptosis compared to paclitaxel alone. In orthotopic mouse models of human TNBC (MDA-MB-231 and MDA-MB-468), individual MBQ-167, paclitaxel, or the combination reduced mammary tumor growth with similar efficacy, with no apparent liver toxicity. However, paclitaxel single agent treatment significantly increased lung metastasis, while MBQ-167, single or combined, reduced lung metastasis. In the syngeneic 4T1/BALB/c model, combined MBQ-167 and paclitaxel decreased established lung metastases by ~80%. To determine the molecular basis for the improved efficacy of the combined treatment on metastasis, 4T1 tumor extracts from BALB/c mice treated with MBQ-167, paclitaxel, or the combination were subjected to transcriptomic analysis. Gene set enrichment identified specific downregulation of central carbon metabolic pathways by the combination of MBQ-167 and Paclitaxel but not individual compounds. Biochemical validation, by immunoblotting and metabolic Seahorse analysis, shows that combined MBQ-167 and paclitaxel reduces glycolysis. This study provides a strong rationale for the clinical testing of MBQ-167 in combination with paclitaxel as a potential therapeutic for TNBC and identifies a unique mechanism of action.

三阴性乳腺癌(TNBC)是一项治疗难题,标准化疗仅限于紫杉醇。MBQ-167是一种靶向Rac和Cdc42的临床阶段小分子抑制剂,可抑制TNBC小鼠模型中的肿瘤生长和转移。在此,我们研究了 MBQ-167 与紫杉醇联用在 TNBC 临床前模型中的疗效,为这种联用在晚期乳腺癌患者中的安全性试验做铺垫。与单独使用紫杉醇相比,单独使用 MBQ-167 或与紫杉醇联合使用能更有效地降低 TNBC 细胞的存活率并增加细胞凋亡。在人类 TNBC(MDA-MB-231 和 MDA-MB-468)的正位小鼠模型中,单独使用 MBQ-167、紫杉醇或联合用药可减少乳腺肿瘤的生长,疗效相似,且无明显的肝毒性。然而,紫杉醇单药治疗会显著增加肺转移,而 MBQ-167(单药或联合用药)则会减少肺转移。在同种异体 4T1/BALB/c 模型中,MBQ-167 和紫杉醇联用可使已形成的肺转移灶减少约 80%。为了确定联合治疗改善转移疗效的分子基础,对接受 MBQ-167、紫杉醇或联合治疗的 BALB/c 小鼠的 4T1 肿瘤提取物进行了转录组分析。基因组富集发现,MBQ-167 和紫杉醇的组合对中心碳代谢途径有特定的下调作用,而对单个化合物则没有。通过免疫印迹和代谢海马分析进行的生化验证表明,MBQ-167 和紫杉醇联用可减少糖酵解。这项研究为将 MBQ-167 与紫杉醇联用作为 TNBC 的潜在疗法进行临床测试提供了强有力的依据,并确定了一种独特的作用机制。
{"title":"Novel inhibition of central carbon metabolism pathways by Rac and Cdc42 inhibitor MBQ-167 and paclitaxel.","authors":"Ailed M Cruz-Collazo, Olga Katsara, Nilmary Grafals-Ruiz, Jessica Colon Gonzalez, Stephanie Dorta-Estremera, Victor P Carlo, Nataliya Chorna, Robert J Schneider, Suranganie Dharmawardhane","doi":"10.1158/1535-7163.MCT-23-0803","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0803","url":null,"abstract":"<p><p>Triple negative breast cancer (TNBC) represents a therapeutic challenge where standard chemotherapy is limited to paclitaxel. MBQ-167, a clinical stage small molecule inhibitor that targets Rac and Cdc42, inhibits tumor growth and metastasis in mouse models of TNBC. Herein, we investigated the efficacy of MBQ-167 in combination with paclitaxel in TNBC pre-clinical models, as a prelude to safety trials of this combination in advanced breast cancer patients. Individual MBQ-167 or combination therapy with paclitaxel was more effective at reducing TNBC cell viability and increasing apoptosis compared to paclitaxel alone. In orthotopic mouse models of human TNBC (MDA-MB-231 and MDA-MB-468), individual MBQ-167, paclitaxel, or the combination reduced mammary tumor growth with similar efficacy, with no apparent liver toxicity. However, paclitaxel single agent treatment significantly increased lung metastasis, while MBQ-167, single or combined, reduced lung metastasis. In the syngeneic 4T1/BALB/c model, combined MBQ-167 and paclitaxel decreased established lung metastases by ~80%. To determine the molecular basis for the improved efficacy of the combined treatment on metastasis, 4T1 tumor extracts from BALB/c mice treated with MBQ-167, paclitaxel, or the combination were subjected to transcriptomic analysis. Gene set enrichment identified specific downregulation of central carbon metabolic pathways by the combination of MBQ-167 and Paclitaxel but not individual compounds. Biochemical validation, by immunoblotting and metabolic Seahorse analysis, shows that combined MBQ-167 and paclitaxel reduces glycolysis. This study provides a strong rationale for the clinical testing of MBQ-167 in combination with paclitaxel as a potential therapeutic for TNBC and identifies a unique mechanism of action.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141860322","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
A DXd/TLR7-agonist dual-conjugate anti-HER2 ADC exerts robust anti-tumor activity through tumor cell killing and immune activation. DXd/TLR7-agonist 双结合抗 HER2 ADC 通过杀伤肿瘤细胞和激活免疫发挥了强大的抗肿瘤活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-31 DOI: 10.1158/1535-7163.MCT-24-0078
Hangtian Yue, Hui Xu, Lanping Ma, Xiyuan Li, Biyu Yang, Xiyuan Wang, Qingzhong Zeng, Han Li, Deqiang Zhang, Meiyu Geng, Tao Meng, Zuoquan Xie

The emergence of trastuzumab deruxtecan (T-DXd), a new-generation antibody-drug conjugate (ADC), has profoundly altered the therapeutic paradigm for HER2-positive solid tumors, demonstrating remarkable clinical benefits. However, the combined outcomes of T-DXd with immunotherapy agents remain ambiguous. In this study, we introduce Tras-DXd-MTL1, an innovative HER2 targeting ADC that integrates the topoisomerase inhibitor DXd and a toll-like receptor 7 (TLR7) agonist MTT-5, linked to trastuzumab via a GGFG tetrapeptide linker. Mechanistically, Tras-DXd-MTL1 retains the DNA-damaging and cell-killing properties of topoisomerase inhibitors while simultaneously enhancing the immune response within the tumor microenvironment (TME). This is achieved by promoting immune cell infiltration and activating dendritic cells and CD8+T cells via MTT-5. In vivo evaluation of Tras-DXd-MTL1's anti-tumor potency revealed a notably superior performance compared to the T-DXd (Tras-DXd) or Tras-MTL1 in immunocompetent mice with trastuzumab-resistant EMT6-HER2 tumor and immunodeficient mice with JIMT-1 tumor. This improved efficacy is primarily attributed to its dual functions of immune stimulation and cytotoxicity. Our findings highlight the potential of incorporating immunostimulatory agents into ADC design to potentiate antitumor effects and establish durable immune memory, thereby reducing tumor recurrence risks. Therefore, our study offers a novel strategy for the design of immune-activating ADCs and provides a potential approach for targeting solid tumors with different levels of HER2 expression.

新一代抗体药物共轭物(ADC)曲妥珠单抗德鲁司坦(T-DXd)的出现,深刻改变了HER2阳性实体瘤的治疗模式,并显示出显著的临床疗效。然而,T-DXd 与免疫疗法药物的联合治疗效果仍不明确。在本研究中,我们介绍了一种创新的 HER2 靶向 ADC--Tras-DXd-MTL1,它整合了拓扑异构酶抑制剂 DXd 和收费样受体 7(TLR7)激动剂 MTT-5,并通过 GGFG 四肽连接体与曲妥珠单抗相连。从机理上讲,Tras-DXd-MTL1 保留了拓扑异构酶抑制剂的 DNA 损伤和细胞杀伤特性,同时增强了肿瘤微环境 (TME) 中的免疫反应。这是通过 MTT-5 促进免疫细胞浸润并激活树突状细胞和 CD8+T 细胞来实现的。对 Tras-DXd-MTL1 抗肿瘤效力的体内评估显示,与 T-DXd (Tras-DXd)或 Tras-MTL1 相比,Tras-DXd-MTL1 在患有曲妥珠单抗耐药 EMT6-HER2 肿瘤的免疫功能健全小鼠和患有 JIMT-1 肿瘤的免疫缺陷小鼠中的表现明显更优。疗效的提高主要归功于其免疫刺激和细胞毒性的双重功能。我们的研究结果凸显了在 ADC 设计中加入免疫刺激剂的潜力,以增强抗肿瘤效果并建立持久的免疫记忆,从而降低肿瘤复发风险。因此,我们的研究为设计免疫激活型 ADC 提供了一种新策略,并为靶向不同 HER2 表达水平的实体瘤提供了一种潜在的方法。
{"title":"A DXd/TLR7-agonist dual-conjugate anti-HER2 ADC exerts robust anti-tumor activity through tumor cell killing and immune activation.","authors":"Hangtian Yue, Hui Xu, Lanping Ma, Xiyuan Li, Biyu Yang, Xiyuan Wang, Qingzhong Zeng, Han Li, Deqiang Zhang, Meiyu Geng, Tao Meng, Zuoquan Xie","doi":"10.1158/1535-7163.MCT-24-0078","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0078","url":null,"abstract":"<p><p>The emergence of trastuzumab deruxtecan (T-DXd), a new-generation antibody-drug conjugate (ADC), has profoundly altered the therapeutic paradigm for HER2-positive solid tumors, demonstrating remarkable clinical benefits. However, the combined outcomes of T-DXd with immunotherapy agents remain ambiguous. In this study, we introduce Tras-DXd-MTL1, an innovative HER2 targeting ADC that integrates the topoisomerase inhibitor DXd and a toll-like receptor 7 (TLR7) agonist MTT-5, linked to trastuzumab via a GGFG tetrapeptide linker. Mechanistically, Tras-DXd-MTL1 retains the DNA-damaging and cell-killing properties of topoisomerase inhibitors while simultaneously enhancing the immune response within the tumor microenvironment (TME). This is achieved by promoting immune cell infiltration and activating dendritic cells and CD8+T cells via MTT-5. In vivo evaluation of Tras-DXd-MTL1's anti-tumor potency revealed a notably superior performance compared to the T-DXd (Tras-DXd) or Tras-MTL1 in immunocompetent mice with trastuzumab-resistant EMT6-HER2 tumor and immunodeficient mice with JIMT-1 tumor. This improved efficacy is primarily attributed to its dual functions of immune stimulation and cytotoxicity. Our findings highlight the potential of incorporating immunostimulatory agents into ADC design to potentiate antitumor effects and establish durable immune memory, thereby reducing tumor recurrence risks. Therefore, our study offers a novel strategy for the design of immune-activating ADCs and provides a potential approach for targeting solid tumors with different levels of HER2 expression.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141856024","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
NAD+ metabolic enzyme inhibitor as radiosensitizer for malignant meningioma and its modulation of P53 expression. NAD+ 代谢酶抑制剂作为恶性脑膜瘤的放射增敏剂及其对 P53 表达的调节。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-23 DOI: 10.1158/1535-7163.MCT-23-0632
Yifan Lv, Yuxuan Deng, Jie Feng, Jinqiu Liu, Mingxu Yang, Zhuonan Pu, Shaodong Zhang, Zhen Wu, Nan Ji, Deric M Park, Shuyu Hao

Surgical resection followed by radiotherapy (RT) is recommended for malignant meningioma but poor outcome is unavoidable. To improve the efficacy of RT in malignant meningioma, a targeted radiosensitizer could be added. Nicotinamide phosphoribosyltransferase (NAMPT), highly expressed in high-grade meningiomas may have a role in determining the radioresponse. Here, we evaluated the impact of NAMPT inhibition on radiosensitivity in malignant meningioma in vivo and in vitro. IOMM-Lee and TTMM705 cells were treated with NAMPT inhibition (FK866 or shRNA NAMPT) before irradiation. The subsequent clonogenic assay demonstrated significantly increased radiosensitivity. Combination treatment with FK866 and irradiation significantly increased the number of G2/M-phase cells, the percentage of apoptotic cells and the γ-H2A.X level compared to FK866 or RT alone. We examined the effect of NAMPT inhibition on NMI and p53 expression in IOMM-Lee and TTMM705 cells. NAMPT inhibition by FK866 and shRNA treatment increased NMI, p53, CDKN1A and BAX expression. Additionally, we assessed the efficacy of FK866/RT combination treatment in vivo. The combination treatment exhibited increased antitumor efficacy compared to either treatment alone. The Ki-67 level was significantly lower and the p53 and γ-H2A.X level was significantly higher in the combination treatment group than in any of the other three groups. In conclusion, these results indicate that FK866 improves radiosensitivity in malignant meningioma, an effect that may be attributed to the increase in p53 expression.

恶性脑膜瘤建议先手术切除,然后进行放射治疗(RT),但疗效不佳是不可避免的。为了提高 RT 对恶性脑膜瘤的疗效,可以添加靶向放射增敏剂。在高级别脑膜瘤中高表达的烟酰胺磷酸核糖转移酶(NAMPT)可能在决定放射反应方面发挥作用。在此,我们评估了体内和体外抑制 NAMPT 对恶性脑膜瘤放射敏感性的影响。IOMM-Lee 和 TTMM705 细胞在照射前接受 NAMPT 抑制剂(FK866 或 shRNA NAMPT)处理。随后进行的克隆生成试验表明,放射敏感性明显提高。与单独使用 FK866 或 RT 相比,联合使用 FK866 和照射可显著增加 G2/M 期细胞数量、凋亡细胞百分比和 γ-H2A.X 水平。我们研究了抑制 NAMPT 对 IOMM-Lee 和 TTMM705 细胞中 NMI 和 p53 表达的影响。通过 FK866 和 shRNA 处理抑制 NAMPT 增加了 NMI、p53、CDKN1A 和 BAX 的表达。此外,我们还评估了 FK866/RT 联合疗法在体内的疗效。与单独治疗相比,联合治疗的抗肿瘤效果更佳。与其他三组相比,联合治疗组的 Ki-67 水平明显降低,p53 和 γ-H2A.X 水平明显升高。总之,这些结果表明,FK866能提高恶性脑膜瘤的放射敏感性,这种效应可能归因于p53表达的增加。
{"title":"NAD+ metabolic enzyme inhibitor as radiosensitizer for malignant meningioma and its modulation of P53 expression.","authors":"Yifan Lv, Yuxuan Deng, Jie Feng, Jinqiu Liu, Mingxu Yang, Zhuonan Pu, Shaodong Zhang, Zhen Wu, Nan Ji, Deric M Park, Shuyu Hao","doi":"10.1158/1535-7163.MCT-23-0632","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0632","url":null,"abstract":"<p><p>Surgical resection followed by radiotherapy (RT) is recommended for malignant meningioma but poor outcome is unavoidable. To improve the efficacy of RT in malignant meningioma, a targeted radiosensitizer could be added. Nicotinamide phosphoribosyltransferase (NAMPT), highly expressed in high-grade meningiomas may have a role in determining the radioresponse. Here, we evaluated the impact of NAMPT inhibition on radiosensitivity in malignant meningioma in vivo and in vitro. IOMM-Lee and TTMM705 cells were treated with NAMPT inhibition (FK866 or shRNA NAMPT) before irradiation. The subsequent clonogenic assay demonstrated significantly increased radiosensitivity. Combination treatment with FK866 and irradiation significantly increased the number of G2/M-phase cells, the percentage of apoptotic cells and the γ-H2A.X level compared to FK866 or RT alone. We examined the effect of NAMPT inhibition on NMI and p53 expression in IOMM-Lee and TTMM705 cells. NAMPT inhibition by FK866 and shRNA treatment increased NMI, p53, CDKN1A and BAX expression. Additionally, we assessed the efficacy of FK866/RT combination treatment in vivo. The combination treatment exhibited increased antitumor efficacy compared to either treatment alone. The Ki-67 level was significantly lower and the p53 and γ-H2A.X level was significantly higher in the combination treatment group than in any of the other three groups. In conclusion, these results indicate that FK866 improves radiosensitivity in malignant meningioma, an effect that may be attributed to the increase in p53 expression.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141748632","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 Ovarian Cancer Stem Cells by Dual Inhibition of the Long Noncoding RNA HOTAIR and Lysine Methyltransferase EZH2. 通过双重抑制长非编码 RNA HOTAIR 和赖氨酸甲基转移酶 EZH2 靶向卵巢癌干细胞
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-23 DOI: 10.1158/1535-7163.MCT-23-0314
Weini Wang, Yanchi Zhou, Ji Wang, Shu Zhang, Ali Ozes, Hongyu Gao, Fang Fang, Yue Wang, Xiaona Chu, Yunlong Liu, Jun Wan, Anirban K Mitra, Heather M O'Hagan, Kenneth P Nephew

Persistence of cancer stem cells (CSCs) is believed to contribute to resistance to platinum-based chemotherapy and disease relapse in ovarian cancer, the fifth leading cause of cancer-related death among US women. HOXC transcript antisense RNA (HOTAIR) is a long noncoding RNA (lncRNA) overexpressed in high-grade serous ovarian cancer and linked to chemoresistance. However, HOTAIR impacts chromatin dynamics in ovarian CSCs and how this oncogenic lncRNA contributes to drug resistant disease are incompletely understood. Here we generated HOTAIR knock-out (KO) high-grade serous ovarian cancer cell lines using paired CRISPR guide RNA design to investigate the function of HOTAIR. We show loss of HOTAIR function re-sensitized ovarian cancer cells to platinum treatment and decreased the population of ovarian CSCs. Furthermore, HOTAIR KO inhibited development of stemness-related phenotypes, including spheroid formation ability, as well as expression of key stemness-associated genes ALDH1A1, NOTCH3, SOX9, and PROM1. HOTAIR KO altered both the cellular transcriptome and chromatin accessibility landscape of multiple oncogenic-associated genes and pathways, including the NF-kB pathway. HOTAIR functions as an oncogene by recruiting enhancer of zeste 2 (EZH2) to catalyze H3K27 tri-methylation to suppress downstream tumor suppressor genes, and it was of interest to inhibit both HOTAIR and EZH2. In vivo, combining a HOTAIR inhibitor with an EZH2 inhibitor and platinum chemotherapy decreased tumor formation and increased survival. These results suggest a key role for HOTAIR in ovarian CSCs and malignant potential. Targeting HOTAIR in combination with epigenetic therapies may represents therapeutic strategy to ameliorate ovarian cancer progression and resistance to platinum-based chemotherapy.

癌症干细胞(CSCs)的持续存在被认为是导致卵巢癌对铂类化疗产生耐药性和疾病复发的原因,而卵巢癌是美国妇女癌症相关死亡的第五大原因。HOXC转录反义RNA(HOTAIR)是一种长非编码RNA(lncRNA),在高级别浆液性卵巢癌中过度表达,与化疗耐药性有关。然而,HOTAIR对卵巢癌细胞间充质干细胞染色质动力学的影响以及这种致癌lncRNA如何导致耐药性疾病还不完全清楚。在这里,我们利用成对的CRISPR引导RNA设计生成了HOTAIR敲除(KO)高级别浆液性卵巢癌细胞系,以研究HOTAIR的功能。我们发现,HOTAIR功能缺失可使卵巢癌细胞对铂治疗重新敏感,并减少卵巢CSCs的数量。此外,HOTAIR KO抑制了干细胞相关表型的发展,包括球形体形成能力,以及干细胞相关关键基因ALDH1A1、NOTCH3、SOX9和PROM1的表达。HOTAIR KO改变了多个致癌相关基因和通路(包括NF-kB通路)的细胞转录组和染色质可及性。HOTAIR通过招募泽斯特增强子2(EZH2)催化H3K27三甲基化来抑制下游肿瘤抑制基因,从而发挥致癌基因的作用,因此同时抑制HOTAIR和EZH2很有意义。在体内,将 HOTAIR 抑制剂与 EZH2 抑制剂和铂类化疗结合使用,可减少肿瘤的形成并提高存活率。这些结果表明,HOTAIR在卵巢造血干细胞和恶性潜能中起着关键作用。靶向 HOTAIR 并结合表观遗传疗法可能是改善卵巢癌进展和铂类化疗耐药性的治疗策略。
{"title":"Targeting Ovarian Cancer Stem Cells by Dual Inhibition of the Long Noncoding RNA HOTAIR and Lysine Methyltransferase EZH2.","authors":"Weini Wang, Yanchi Zhou, Ji Wang, Shu Zhang, Ali Ozes, Hongyu Gao, Fang Fang, Yue Wang, Xiaona Chu, Yunlong Liu, Jun Wan, Anirban K Mitra, Heather M O'Hagan, Kenneth P Nephew","doi":"10.1158/1535-7163.MCT-23-0314","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0314","url":null,"abstract":"<p><p>Persistence of cancer stem cells (CSCs) is believed to contribute to resistance to platinum-based chemotherapy and disease relapse in ovarian cancer, the fifth leading cause of cancer-related death among US women. HOXC transcript antisense RNA (HOTAIR) is a long noncoding RNA (lncRNA) overexpressed in high-grade serous ovarian cancer and linked to chemoresistance. However, HOTAIR impacts chromatin dynamics in ovarian CSCs and how this oncogenic lncRNA contributes to drug resistant disease are incompletely understood. Here we generated HOTAIR knock-out (KO) high-grade serous ovarian cancer cell lines using paired CRISPR guide RNA design to investigate the function of HOTAIR. We show loss of HOTAIR function re-sensitized ovarian cancer cells to platinum treatment and decreased the population of ovarian CSCs. Furthermore, HOTAIR KO inhibited development of stemness-related phenotypes, including spheroid formation ability, as well as expression of key stemness-associated genes ALDH1A1, NOTCH3, SOX9, and PROM1. HOTAIR KO altered both the cellular transcriptome and chromatin accessibility landscape of multiple oncogenic-associated genes and pathways, including the NF-kB pathway. HOTAIR functions as an oncogene by recruiting enhancer of zeste 2 (EZH2) to catalyze H3K27 tri-methylation to suppress downstream tumor suppressor genes, and it was of interest to inhibit both HOTAIR and EZH2. In vivo, combining a HOTAIR inhibitor with an EZH2 inhibitor and platinum chemotherapy decreased tumor formation and increased survival. These results suggest a key role for HOTAIR in ovarian CSCs and malignant potential. Targeting HOTAIR in combination with epigenetic therapies may represents therapeutic strategy to ameliorate ovarian cancer progression and resistance to platinum-based chemotherapy.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141748633","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
Identification of ATP-Competitive Human CMG Helicase Inhibitors for Cancer Intervention that Disrupt CMG-Replisome Function. 鉴定 ATP 竞争性人类 CMG 螺旋酶抑制剂,用于干扰 CMG 重组体功能的癌症干预。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-10 DOI: 10.1158/1535-7163.MCT-23-0904
Shengyan Xiang, Kendall C Craig, Xingju Luo, Darcy L Welch, Renan B Ferreira, Harshani R Lawrence, Nicholas J Lawrence, Damon R Reed, Mark G Alexandrow

The human CMG helicase (Cdc45-MCM-GINS) is a novel target for anti-cancer therapy. Tumor-specific weaknesses in the CMG are caused by oncogene-driven changes that adversely affect CMG function, and a requirement for CMG activity during recovery from replicative stresses such as chemotherapy. Here, we developed an orthogonal biochemical screening approach and identified CMG inhibitors (CMGi) that inhibit ATPase and helicase activities in an ATP-competitive manner at low micromolar concentrations. Structure-activity information, in silico docking, and testing with synthetic chemical compounds indicate that CMGi require specific chemical elements and occupy ATP binding sites and channels within MCM subunits leading to the ATP clefts, which are likely used for ATP/ADP ingress or egress. CMGi are therefore also MCM complex inhibitors (MCMi). Biological testing shows that CMGi/MCMi inhibit cell growth and DNA replication using multiple molecular mechanisms distinct from other chemotherapy agents. CMGi/MCMi block helicase assembly steps that require ATP binding/hydrolysis by the MCM complex, specifically MCM ring assembly on DNA and GINS recruitment to DNA-loaded MCM hexamers. During S-phase, inhibition of MCM ATP binding/hydrolysis by CMGi/MCMi causes a 'reverse allosteric' dissociation of Cdc45/GINS from the CMG that destabilizes replisome components Ctf4, Mcm10, and DNA polymerase-a, -d, -e, resulting in DNA damage. CMGi/MCMi display selective toxicity toward multiple solid tumor cell types with K-Ras mutations, targeting the CMG and inducing DNA damage, Parp cleavage, and loss of viability. This new class of CMGi/MCMi provides a basis for small chemical development of CMG helicase-targeted anti-cancer compounds with distinct mechanisms of action.

人类 CMG 螺旋酶(Cdc45-MCM-GINS)是抗癌疗法的一个新靶点。肿瘤特异性的CMG弱点是由癌基因驱动的变化造成的,这些变化对CMG的功能产生了不利影响,而且在从化疗等复制压力中恢复时需要CMG的活性。在这里,我们开发了一种正交生化筛选方法,并确定了 CMG 抑制剂(CMGi),这些抑制剂能在低微摩浓度下以 ATP 竞争方式抑制 ATP 酶和螺旋酶的活性。结构-活性信息、硅学对接和合成化合物测试表明,CMGi 需要特定的化学元素,并占据 ATP 结合位点和 MCM 亚基内通向 ATP 裂隙的通道,而 ATP/ADP 裂隙可能用于 ATP/ADP 的进入或排出。因此,CMGi 也是 MCM 复合物抑制剂(MCMi)。生物测试表明,CMGi/MCMi 利用不同于其他化疗药物的多种分子机制抑制细胞生长和 DNA 复制。CMGi/MCMi 可阻断需要 MCM 复合物与 ATP 结合/水解的螺旋酶组装步骤,特别是 MCM 环在 DNA 上的组装和 GINS 招募到 DNA 加载的 MCM 六聚体上。在 S 期,CMGi/MCMi 对 MCM ATP 结合/水解的抑制会导致 Cdc45/GINS 与 CMG 的 "反向异构 "解离,从而破坏复制体成分 Ctf4、Mcm10 和 DNA 聚合酶-a、-d、-e 的稳定性,造成 DNA 损伤。CMGi/MCMi 对带有 K-Ras 突变的多种实体瘤细胞具有选择性毒性,以 CMG 为靶点,诱导 DNA 损伤、Parp 断裂和活力丧失。这一类新型 CMGi/MCMi 为开发具有独特作用机制的 CMG 螺旋酶靶向抗癌化合物的小型化学研究奠定了基础。
{"title":"Identification of ATP-Competitive Human CMG Helicase Inhibitors for Cancer Intervention that Disrupt CMG-Replisome Function.","authors":"Shengyan Xiang, Kendall C Craig, Xingju Luo, Darcy L Welch, Renan B Ferreira, Harshani R Lawrence, Nicholas J Lawrence, Damon R Reed, Mark G Alexandrow","doi":"10.1158/1535-7163.MCT-23-0904","DOIUrl":"10.1158/1535-7163.MCT-23-0904","url":null,"abstract":"<p><p>The human CMG helicase (Cdc45-MCM-GINS) is a novel target for anti-cancer therapy. Tumor-specific weaknesses in the CMG are caused by oncogene-driven changes that adversely affect CMG function, and a requirement for CMG activity during recovery from replicative stresses such as chemotherapy. Here, we developed an orthogonal biochemical screening approach and identified CMG inhibitors (CMGi) that inhibit ATPase and helicase activities in an ATP-competitive manner at low micromolar concentrations. Structure-activity information, in silico docking, and testing with synthetic chemical compounds indicate that CMGi require specific chemical elements and occupy ATP binding sites and channels within MCM subunits leading to the ATP clefts, which are likely used for ATP/ADP ingress or egress. CMGi are therefore also MCM complex inhibitors (MCMi). Biological testing shows that CMGi/MCMi inhibit cell growth and DNA replication using multiple molecular mechanisms distinct from other chemotherapy agents. CMGi/MCMi block helicase assembly steps that require ATP binding/hydrolysis by the MCM complex, specifically MCM ring assembly on DNA and GINS recruitment to DNA-loaded MCM hexamers. During S-phase, inhibition of MCM ATP binding/hydrolysis by CMGi/MCMi causes a 'reverse allosteric' dissociation of Cdc45/GINS from the CMG that destabilizes replisome components Ctf4, Mcm10, and DNA polymerase-a, -d, -e, resulting in DNA damage. CMGi/MCMi display selective toxicity toward multiple solid tumor cell types with K-Ras mutations, targeting the CMG and inducing DNA damage, Parp cleavage, and loss of viability. This new class of CMGi/MCMi provides a basis for small chemical development of CMG helicase-targeted anti-cancer compounds with distinct mechanisms of action.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141563768","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":null,"pages":null},"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
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