首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
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 : 2024-12-03 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 a high prevalence in non-small cell lung cancer, colorectal cancer, and pancreatic cancer. FDA-approved sotorasib and adagrasib provide breakthrough therapies for patients with cancer 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 of Son of Sevenless 1 (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 nanomoles per liter potency and was 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 antiproliferation 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. Combinations 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":"10.1158/1535-7163.MCT-24-0049","url":null,"abstract":"<p><p>KRAS is the most frequently dysregulated oncogene with a high prevalence in non-small cell lung cancer, colorectal cancer, and pancreatic cancer. FDA-approved sotorasib and adagrasib provide breakthrough therapies for patients with cancer 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 of Son of Sevenless 1 (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 nanomoles per liter potency and was 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 antiproliferation 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. Combinations 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":"1703-1716"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","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
Fatty Acid Derivatization and Cyclization of the Immunomodulatory Peptide RP-182 Targeting CD206high Macrophages Improve Antitumor Activity. 针对 CD206 高巨噬细胞的免疫调节肽 RP-182 的脂肪酸衍生化和环化提高了抗肿瘤活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-23-0790
Sitanshu S Singh, Raul Calvo, Anju Kumari, Rushikesh V Sable, Yuhong Fang, Dingyin Tao, Xin Hu, Sarah Gray Castle, Saifun Nahar, Dandan Li, Emily Major, Tino W Sanchez, Rintaro Kato, Xin Xu, Jian Zhou, Liang Liu, Christopher A LeClair, Anton Simeonov, Bolormaa Baljinnyam, Mark J Henderson, Juan Marugan, Udo Rudloff

As tumor-associated macrophages (TAM) exercise a plethora of protumor and immune evasive functions, novel strategies targeting TAMs to inhibit tumor progression have emerged within the current arena of cancer immunotherapy. Activation of the mannose receptor 1 (CD206) is a recent approach that recognizes immunosuppressive CD206high M2-like TAMs as a drug target. Ligation of CD206 both induces reprogramming of CD206high TAMs toward a proinflammatory phenotype and selectively triggers apoptosis in these cells. CD206-activating therapeutics are currently limited to the linear, 10mer peptide RP-182, 1, which is not a drug candidate. In this study, we sought to identify a better suitable candidate for future clinical development by synthesizing and evaluating a series of RP-182 analogs. Surprisingly, fatty acid derivative 1a [RP-182-PEG3-K(palmitic acid)] not only showed improved stability but also increased affinity to the CD206 receptor through enhanced interaction with a hydrophobic binding motif of CD206. Peptide 1a showed superior in vitro activity in cell-based assays of macrophage activation which was restricted to CD206high M2-polarized macrophages. Improvement in responses was disproportionally skewed toward improved induction of phagocytosis including cancer cell phagocytosis. Peptide 1a reprogrammed the immune landscape in genetically engineered murine KPC pancreatic tumors toward increased innate immune surveillance and improved tumor control and effectively suppressed tumor growth of murine B16 melanoma allografts.

由于肿瘤相关巨噬细胞(TAMs)具有大量的促肿瘤和免疫回避功能,因此在当前的癌症免疫疗法领域出现了以 TAMs 为靶点抑制肿瘤进展的新策略。激活甘露糖受体 1(Mrc1;CD206)是最近一种将免疫抑制性 CD206 高 M2 样 TAMs 识别为药物靶点的方法。对 CD206 的连接既能诱导 CD206 高的 TAMs 重编程,使其趋向于促炎表型,又能选择性地触发这些细胞的凋亡。CD206 激活疗法目前仅限于线性 10 聚体肽 RP-182,1,它还不是候选药物。在此,我们试图通过合成和评估一系列 RP-182 类似物,为未来的临床开发找到更合适的候选药物。令人惊讶的是,脂肪酸衍生物 1a(RP-182-PEG3-K(棕榈酸))不仅提高了稳定性,还通过增强与 CD206 的疏水结合基团的相互作用提高了与 CD206 受体的亲和力。在基于细胞的巨噬细胞活化测试中,肽 1a 显示出更高的体外活性,这种活化仅限于 CD206 高的 M2 极化巨噬细胞。反应的改善不成比例地偏向于诱导吞噬功能的改善,包括癌细胞的吞噬功能。1a 重编程了基因工程小鼠 KPC 胰腺肿瘤的免疫格局,使其先天免疫监视增强,肿瘤控制得到改善,并有效抑制了小鼠 B16 黑色素瘤异种移植的肿瘤生长。
{"title":"Fatty Acid Derivatization and Cyclization of the Immunomodulatory Peptide RP-182 Targeting CD206high Macrophages Improve Antitumor Activity.","authors":"Sitanshu S Singh, Raul Calvo, Anju Kumari, Rushikesh V Sable, Yuhong Fang, Dingyin Tao, Xin Hu, Sarah Gray Castle, Saifun Nahar, Dandan Li, Emily Major, Tino W Sanchez, Rintaro Kato, Xin Xu, Jian Zhou, Liang Liu, Christopher A LeClair, Anton Simeonov, Bolormaa Baljinnyam, Mark J Henderson, Juan Marugan, Udo Rudloff","doi":"10.1158/1535-7163.MCT-23-0790","DOIUrl":"10.1158/1535-7163.MCT-23-0790","url":null,"abstract":"<p><p>As tumor-associated macrophages (TAM) exercise a plethora of protumor and immune evasive functions, novel strategies targeting TAMs to inhibit tumor progression have emerged within the current arena of cancer immunotherapy. Activation of the mannose receptor 1 (CD206) is a recent approach that recognizes immunosuppressive CD206high M2-like TAMs as a drug target. Ligation of CD206 both induces reprogramming of CD206high TAMs toward a proinflammatory phenotype and selectively triggers apoptosis in these cells. CD206-activating therapeutics are currently limited to the linear, 10mer peptide RP-182, 1, which is not a drug candidate. In this study, we sought to identify a better suitable candidate for future clinical development by synthesizing and evaluating a series of RP-182 analogs. Surprisingly, fatty acid derivative 1a [RP-182-PEG3-K(palmitic acid)] not only showed improved stability but also increased affinity to the CD206 receptor through enhanced interaction with a hydrophobic binding motif of CD206. Peptide 1a showed superior in vitro activity in cell-based assays of macrophage activation which was restricted to CD206high M2-polarized macrophages. Improvement in responses was disproportionally skewed toward improved induction of phagocytosis including cancer cell phagocytosis. Peptide 1a reprogrammed the immune landscape in genetically engineered murine KPC pancreatic tumors toward increased innate immune surveillance and improved tumor control and effectively suppressed tumor growth of murine B16 melanoma allografts.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1827-1841"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11612619/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142109602","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
Trametinib Potentiates Anti-PD-1 Efficacy in Tumors Established from Chemotherapy-Primed Pancreatic Cancer Cells. 曲美替尼可增强化疗诱导的胰腺癌细胞形成的肿瘤的抗PD-1疗效。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-23-0833
Thao D Pham, Anastasia E Metropulos, Nida Mubin, Jeffrey H Becker, Dhavan Shah, Christina Spaulding, Mario A Shields, David J Bentrem, Hidayatullah G Munshi

Despite advances in immune checkpoint inhibitors, chemotherapy remains the standard therapy for patients with pancreatic ductal adenocarcinoma (PDAC). As the combinations of chemotherapy, including the FOLFIRINOX [5-fluorouracil, F; irinotecan, I; and oxaliplatin, O (FIO)] regimen, and immune checkpoint inhibitors have failed to demonstrate clinical benefit in patients with metastatic PDAC tumors, there is increasing interest in identifying therapeutic approaches to potentiate ICI efficacy in patients with PDAC. In this study, we report that neoadjuvant FOLFIRINOX-treated human PDAC tumors exhibit increased MEK/ERK activation. We also show elevated MEK/ERK signaling in ex vivo PDAC slice cultures and cell lines treated with a combination of FIO. In addition, we find that the KPC-FIO cells, established from repeated treatment of mouse PDAC cell lines with six to eight cycles of FIO, display enhanced ERK phosphorylation and demonstrate increased sensitivity to MEK inhibition in vitro and in vivo. Significantly, the KPC-FIO cells develop tumors with a proinflammatory immune profile similar to human PDAC tumors after neoadjuvant FOLFIRINOX treatment. Furthermore, we found that the MEK inhibitor trametinib enables additional infiltration of highly functional CD8+ T cells into the KPC-FIO tumors and potentiates the efficacy of anti-PD-1 antibody in syngeneic mouse models. Our findings provide a rationale for combining trametinib and anti-PD-1 antibodies in patients with PDAC after neoadjuvant or short-term FOLFIRINOX treatment to achieve effective antitumor responses.

尽管免疫检查点抑制剂(ICIs)取得了进展,但化疗仍是胰腺导管腺癌(PDAC)患者的标准疗法。由于化疗(包括 FOLFIRINOX(5-氟尿嘧啶(5FU)、伊立替康和奥沙利铂)方案)和 ICIs 的联合治疗未能在转移性 PDAC 肿瘤患者中显示出临床疗效,因此人们越来越关注寻找治疗方法来增强 ICI 在 PDAC 患者中的疗效。在本研究中,我们报告了新辅助 FOLFRINOX 治疗的人类 PDAC 肿瘤表现出 MEK/ERK 激活增加。我们还发现,在体内外用 5FU(F)、伊立替康(I)和奥沙利铂(O)(FIO)联合治疗的 PDAC 切片培养物和细胞系中,MEK/ERK 信号增强。此外,我们还发现,小鼠 PDAC 细胞系经 6-8 个周期的 FIO 反复处理后建立的 KPC-FIO 细胞显示出增强的 ERK 磷酸化,并在体外和体内显示出对 MEK 抑制剂更高的敏感性。值得注意的是,KPC-FIO 细胞在新辅助 FOLFIRINOX 治疗后,肿瘤发生了与人类 PDAC 肿瘤相似的促炎免疫特征。此外,我们还发现,MEK 抑制剂曲美替尼可使更多高功能 CD8+ T 细胞渗入 KPC-FIO 肿瘤,并增强抗 PD-1 抗体在合成小鼠模型中的疗效。我们的研究结果为新辅助治疗或短期FOLFIRINOX治疗后的PDAC患者联合使用Trametinib和抗PD-1抗体以实现有效的抗肿瘤反应提供了理论依据。
{"title":"Trametinib Potentiates Anti-PD-1 Efficacy in Tumors Established from Chemotherapy-Primed Pancreatic Cancer Cells.","authors":"Thao D Pham, Anastasia E Metropulos, Nida Mubin, Jeffrey H Becker, Dhavan Shah, Christina Spaulding, Mario A Shields, David J Bentrem, Hidayatullah G Munshi","doi":"10.1158/1535-7163.MCT-23-0833","DOIUrl":"10.1158/1535-7163.MCT-23-0833","url":null,"abstract":"<p><p>Despite advances in immune checkpoint inhibitors, chemotherapy remains the standard therapy for patients with pancreatic ductal adenocarcinoma (PDAC). As the combinations of chemotherapy, including the FOLFIRINOX [5-fluorouracil, F; irinotecan, I; and oxaliplatin, O (FIO)] regimen, and immune checkpoint inhibitors have failed to demonstrate clinical benefit in patients with metastatic PDAC tumors, there is increasing interest in identifying therapeutic approaches to potentiate ICI efficacy in patients with PDAC. In this study, we report that neoadjuvant FOLFIRINOX-treated human PDAC tumors exhibit increased MEK/ERK activation. We also show elevated MEK/ERK signaling in ex vivo PDAC slice cultures and cell lines treated with a combination of FIO. In addition, we find that the KPC-FIO cells, established from repeated treatment of mouse PDAC cell lines with six to eight cycles of FIO, display enhanced ERK phosphorylation and demonstrate increased sensitivity to MEK inhibition in vitro and in vivo. Significantly, the KPC-FIO cells develop tumors with a proinflammatory immune profile similar to human PDAC tumors after neoadjuvant FOLFIRINOX treatment. Furthermore, we found that the MEK inhibitor trametinib enables additional infiltration of highly functional CD8+ T cells into the KPC-FIO tumors and potentiates the efficacy of anti-PD-1 antibody in syngeneic mouse models. Our findings provide a rationale for combining trametinib and anti-PD-1 antibodies in patients with PDAC after neoadjuvant or short-term FOLFIRINOX treatment to achieve effective antitumor responses.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1854-1865"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11614707/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142004821","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
Inhibition of PSF activity overcomes resistance to treatment in cancers harboring mutant p53. 抑制PSF活性可以克服携带p53突变体的癌症对治疗的耐药性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-24-0418
Ken-Ichi Takayama, Tomohiro Sato, Teruki Honma, Minoru Yoshida, Satoshi Inoue

Mutations in the TP53 tumor suppressor genes are prevalent in aggressive cancers. Pharmacological reactivation of dysfunctional p53 due to mutations is a promising strategy for treating such cancers. Recently, a multifunctional proline- and glutamine-rich protein, PTB-associated splicing factor (PSF), was identified as a key driver of aggressive cancers. PSF promotes the expression of numerous oncogenes by modulating epigenetic and splicing mechanisms. We previously screened a small-molecule library and discovered compound No.10-3 as a potent PSF inhibitor. Here, we report the discovery of a No.10-3 analog, C-30, as a potent PSF inhibitor. Compared to No.10-3, C-30 treatment specifically suppressed the growth and induced apoptosis of mutant p53-bearing and therapy-resistant cancer cells. Interestingly, C-30 activated a set of p53-regulated genes in therapy-resistant cancer cells. A comprehensive analysis of PSF and p53 binding regions demonstrated a higher level of PSF-binding potential in mutant p53-expressing cancer cells around genomic regions identified as p53-binding peaks in p53-wild type cancer cells. Treatment of mutant p53-expressing cancer cells with C-30 decreases PSF binding around these sites, leading to activated histone acetylation. We further demonstrated that C-30 impaired tumor growth and increased the expression of p53-target genes in vivo. These results suggested that C-30 produces tumor-suppressive effects similar to the functional reactivation of p53, providing a rationale for the inhibition of PSF activity as a promising therapy against treatment-resistant cancer.

肿瘤抑制基因TP53的突变在侵袭性癌症中很普遍。由于突变导致功能失调的p53的药理学再激活是治疗此类癌症的一种有希望的策略。最近,一种富含脯氨酸和谷氨酰胺的多功能蛋白pdb相关剪接因子(PSF)被确定为侵袭性癌症的关键驱动因素。PSF通过调节表观遗传和剪接机制促进许多癌基因的表达。我们之前筛选了一个小分子文库,发现化合物No.10-3是一个有效的PSF抑制剂。在这里,我们报告了一种No.10-3类似物C-30的发现,它是一种有效的PSF抑制剂。与No.10-3相比,C-30处理特异性抑制p53突变体和耐药癌细胞的生长和诱导凋亡。有趣的是,C-30激活了治疗耐药癌细胞中的一组p53调节基因。对PSF和p53结合区域的综合分析表明,在p53野生型癌细胞中p53结合峰所在的基因组区域附近,表达p53的突变型癌细胞中PSF的结合潜力更高。用C-30处理表达p53突变的癌细胞会减少这些位点周围的PSF结合,导致组蛋白乙酰化活化。我们进一步证明C-30在体内抑制肿瘤生长并增加p53靶基因的表达。这些结果表明,C-30产生类似于p53功能再激活的肿瘤抑制作用,为抑制PSF活性作为治疗耐药癌症的一种有希望的治疗方法提供了理论依据。
{"title":"Inhibition of PSF activity overcomes resistance to treatment in cancers harboring mutant p53.","authors":"Ken-Ichi Takayama, Tomohiro Sato, Teruki Honma, Minoru Yoshida, Satoshi Inoue","doi":"10.1158/1535-7163.MCT-24-0418","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0418","url":null,"abstract":"<p><p>Mutations in the TP53 tumor suppressor genes are prevalent in aggressive cancers. Pharmacological reactivation of dysfunctional p53 due to mutations is a promising strategy for treating such cancers. Recently, a multifunctional proline- and glutamine-rich protein, PTB-associated splicing factor (PSF), was identified as a key driver of aggressive cancers. PSF promotes the expression of numerous oncogenes by modulating epigenetic and splicing mechanisms. We previously screened a small-molecule library and discovered compound No.10-3 as a potent PSF inhibitor. Here, we report the discovery of a No.10-3 analog, C-30, as a potent PSF inhibitor. Compared to No.10-3, C-30 treatment specifically suppressed the growth and induced apoptosis of mutant p53-bearing and therapy-resistant cancer cells. Interestingly, C-30 activated a set of p53-regulated genes in therapy-resistant cancer cells. A comprehensive analysis of PSF and p53 binding regions demonstrated a higher level of PSF-binding potential in mutant p53-expressing cancer cells around genomic regions identified as p53-binding peaks in p53-wild type cancer cells. Treatment of mutant p53-expressing cancer cells with C-30 decreases PSF binding around these sites, leading to activated histone acetylation. We further demonstrated that C-30 impaired tumor growth and increased the expression of p53-target genes in vivo. These results suggested that C-30 produces tumor-suppressive effects similar to the functional reactivation of p53, providing a rationale for the inhibition of PSF activity as a promising therapy against treatment-resistant cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142770321","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 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 : 2024-12-03 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 (PDHK) and pyruvate dehydrogenase phosphatase. Studies have demonstrated that 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 NK cell exhaustion to investigate the impact of dichloroacetate (DCA) on NK cell function. The production of granzyme B, IFNγ, TNFα, 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 and extracellular acidification rate of NK cells. A mouse model was used to investigate the potential of combining DCA with adjuvant NK cell infusion. Our study demonstrated that the hepatocellular carcinoma microenvironment mediated NK cellular exhaustion and 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 s.c. hepatocellular carcinoma 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 antitumor 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":"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 (PDHK) and pyruvate dehydrogenase phosphatase. Studies have demonstrated that 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 NK cell exhaustion to investigate the impact of dichloroacetate (DCA) on NK cell function. The production of granzyme B, IFNγ, TNFα, 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 and extracellular acidification rate of NK cells. A mouse model was used to investigate the potential of combining DCA with adjuvant NK cell infusion. Our study demonstrated that the hepatocellular carcinoma microenvironment mediated NK cellular exhaustion and 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 s.c. hepatocellular carcinoma 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 antitumor immunity.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1731-1742"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","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
Targeting the Synthetic Lethal Relationship between FOCAD and TUT7 Represents a Potential Therapeutic Opportunity for TUT4/7 Small-Molecule Inhibitors in Cancer. 针对 FOCAD 和 TUT7 之间的合成致死关系,是 TUT4/7 小分子抑制剂治疗癌症的潜在机会。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-24-0176
Robinson Triboulet, Khikmet Sadykov, Darren M Harvey, David M Wilson, Michael J Steinbaugh, Christopher B Mayo, Dillon Hawley, Andrew Madanjian, Corey Fyfe, Christina Bracken, Izarys Rivera-Rivera, Anna Ericsson, Andrew R Snyder, Sarah K Knutson, Ross L Stein, Veronica Gibaja, Shomir Ghosh, Robert M Campbell

Targeting synthetic lethal interactions among genes has emerged as a promising strategy for cancer therapy. This study explores the intricate interplay among terminal uridylyltransferase 4 (TUT4) and terminal uridylyltransferase 7 (TUT7), the 3'-5' exoribonuclease DIS3L2, and the superkiller (SKI) complex-interacting factor focadhesin (FOCAD) in the context of cancer vulnerability. Using CRISPR and public functional genomics data, we show impairment of cell proliferation upon knockout of TUT7 or DIS3L2, but not TUT4, on cancer cells with FOCAD loss. Moreover, we report the characterization of the first potent and selective TUT4/7 inhibitors that substantially reduce uridylation and demonstrate in vitro and in vivo antiproliferative activity specifically in FOCAD-deleted cancer. FOCAD deficiency posttranscriptionally disrupts the stability of the SKI complex, whose role is to safeguard cells against aberrant RNA. Reintroduction of FOCAD restores the SKI complex and makes these cells less sensitive to TUT4/7 inhibitors, indicating that TUT7 dependency is driven by FOCAD loss. We propose a model in which, in the absence of FOCAD, TUT7 and DIS3L2 function as a salvage mechanism that degrades aberrant RNA, and genetic or pharmacologic inhibition of this pathway leads to cell death. Our findings underscore the significance of FOCAD loss as a genetic driver of TUT7 vulnerability and provide insights into the potential utility of TUT4/7 inhibitors for cancer treatment.

靶向基因间的合成致死相互作用已成为一种很有前景的癌症治疗策略。本研究探讨了末端尿苷基转移酶4(TUT4)和末端尿苷基转移酶7(TUT7)、3'-5'外切核酸酶DIS3L2和SKI复合物相互作用因子Focadhesin(FOCAD)之间在癌症易感性方面错综复杂的相互作用。利用 CRISPR 和公开的功能基因组学数据,我们发现在 FOCAD 缺失的癌细胞上敲除 TUT7 或 DIS3L2 后,细胞增殖会受到影响,而敲除 TUT4 则不会。此外,我们还报告了第一种强效选择性 TUT4/7 抑制剂的特性,这种抑制剂能显著降低尿苷酸化,并在体外和体内显示出专门针对 FOCAD 缺失癌症的抗增殖活性。FOCAD 的缺乏会在转录后破坏 SKI 复合物的稳定性,而 SKI 复合物的作用是保护细胞免受异常 RNA 的侵害。重新引入FOCAD可恢复SKI复合物,并使这些细胞对TUT4/7抑制剂不那么敏感,这表明TUT7依赖性是由FOCAD缺失驱动的。我们提出了这样一个模型:在缺乏FOCAD的情况下,TUT7和DIS3L2发挥着降解异常RNA的挽救机制的作用,而对这一途径的遗传或药物抑制会导致细胞死亡。我们的发现强调了FOCAD缺失作为TUT7脆弱性遗传驱动因素的重要性,并为TUT4/7抑制剂在癌症治疗中的潜在作用提供了启示。
{"title":"Targeting the Synthetic Lethal Relationship between FOCAD and TUT7 Represents a Potential Therapeutic Opportunity for TUT4/7 Small-Molecule Inhibitors in Cancer.","authors":"Robinson Triboulet, Khikmet Sadykov, Darren M Harvey, David M Wilson, Michael J Steinbaugh, Christopher B Mayo, Dillon Hawley, Andrew Madanjian, Corey Fyfe, Christina Bracken, Izarys Rivera-Rivera, Anna Ericsson, Andrew R Snyder, Sarah K Knutson, Ross L Stein, Veronica Gibaja, Shomir Ghosh, Robert M Campbell","doi":"10.1158/1535-7163.MCT-24-0176","DOIUrl":"10.1158/1535-7163.MCT-24-0176","url":null,"abstract":"<p><p>Targeting synthetic lethal interactions among genes has emerged as a promising strategy for cancer therapy. This study explores the intricate interplay among terminal uridylyltransferase 4 (TUT4) and terminal uridylyltransferase 7 (TUT7), the 3'-5' exoribonuclease DIS3L2, and the superkiller (SKI) complex-interacting factor focadhesin (FOCAD) in the context of cancer vulnerability. Using CRISPR and public functional genomics data, we show impairment of cell proliferation upon knockout of TUT7 or DIS3L2, but not TUT4, on cancer cells with FOCAD loss. Moreover, we report the characterization of the first potent and selective TUT4/7 inhibitors that substantially reduce uridylation and demonstrate in vitro and in vivo antiproliferative activity specifically in FOCAD-deleted cancer. FOCAD deficiency posttranscriptionally disrupts the stability of the SKI complex, whose role is to safeguard cells against aberrant RNA. Reintroduction of FOCAD restores the SKI complex and makes these cells less sensitive to TUT4/7 inhibitors, indicating that TUT7 dependency is driven by FOCAD loss. We propose a model in which, in the absence of FOCAD, TUT7 and DIS3L2 function as a salvage mechanism that degrades aberrant RNA, and genetic or pharmacologic inhibition of this pathway leads to cell death. Our findings underscore the significance of FOCAD loss as a genetic driver of TUT7 vulnerability and provide insights into the potential utility of TUT4/7 inhibitors for cancer treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1779-1788"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133245","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
TR-107, an Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells. TR-107是酪酸肽酶蛋白水解亚基的一种激动剂,它能破坏线粒体代谢并抑制人类结直肠癌细胞的生长。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-24-0170
Michael Giarrizzo, Joseph F LaComb, Hetvi R Patel, Rohan G Reddy, John D Haley, Lee M Graves, Edwin J Iwanowicz, Agnieszka B Bialkowska

Oxidative phosphorylation is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates nonspecific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound TR-107 has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multitargeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer cells. TR-107 inhibited colorectal cancer cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response and mitochondrial DNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of oxidative phosphorylation and a reduction in total cellular respiration. Multiomics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by colorectal cancer cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107.

氧化磷酸化(OXPHOS)是癌症增殖和抗药性的重要代谢过程。ClpXP 复合物通过降解折叠错误的蛋白质来维持线粒体蛋白稳态。Madera Therapeutics 公司已开发出一类 ClpXP 成分酪蛋白溶肽酶蛋白水解亚基(ClpP)的高效、选择性小分子激活剂(TR 化合物)。这种癌症治疗方法消除了底物识别,激活了线粒体内的非特异性蛋白酶功能,在多种恶性肿瘤中显示出令人鼓舞的临床前疗效。与多靶点临床药物ONC201相比,同类领先化合物TR-107在ClpP亲和力和激活方面的效力显著提高,药代动力学特性也有所增强。在本研究中,我们研究了 TR-107 对人类结直肠癌(CRC)细胞的体外疗效。在低纳摩尔浓度下,TR-107以剂量和时间依赖性方式抑制CRC细胞增殖,并诱导细胞周期停滞。从机理上讲,TR-107 下调了参与线粒体未折叠蛋白反应(UPRmt)以及 mtDNA 转录和翻译的蛋白质的表达。TR-107 降低了耗氧率和糖酵解补偿,证实了 OXPHOS 失活和细胞总呼吸量减少。对处理过的细胞进行的多组学分析表明,呼吸链复合物亚基下调,有丝分裂和铁凋亡途径上调。对铁变态反应的进一步评估显示,抗氧化和铁毒性防御功能耗竭,这可能会增强对联合化疗药物的敏感性。总之,这项研究为 CRC 细胞应对强效 ClpP 激动所采用的亚细胞机制提供了证据和见解。我们的研究结果表明了破坏线粒体代谢的有效方法,支持 TR-107 的转化潜力。
{"title":"TR-107, an Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells.","authors":"Michael Giarrizzo, Joseph F LaComb, Hetvi R Patel, Rohan G Reddy, John D Haley, Lee M Graves, Edwin J Iwanowicz, Agnieszka B Bialkowska","doi":"10.1158/1535-7163.MCT-24-0170","DOIUrl":"10.1158/1535-7163.MCT-24-0170","url":null,"abstract":"<p><p>Oxidative phosphorylation is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates nonspecific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound TR-107 has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multitargeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer cells. TR-107 inhibited colorectal cancer cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response and mitochondrial DNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of oxidative phosphorylation and a reduction in total cellular respiration. Multiomics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by colorectal cancer cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1761-1778"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11614700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133246","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
Advanced Human Papillomavirus-Negative Head and Neck Squamous Cell Carcinoma: Unmet Need and Emerging Therapies. 晚期人类乳头瘤病毒阴性头颈部鳞状细胞癌:未满足的需求和新兴疗法。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-24-0281
Robin Park, Christine H Chung

Despite notable progress in the treatment of advanced head and neck squamous cell carcinoma (HNSCC), survival remains poor in patients with recurrent and/or metastatic (R/M) human papillomavirus (HPV)-negative HNSCC. Worse outcomes in patients who are HPV-negative may be partly related to loss of cell-cycle regulators and tumor suppressors as well as a noninflamed and hypoxic tumor microenvironment, both of which contribute to treatment resistance and disease progression. Anti-programmed cell death protein 1-based regimens as current standard-of-care treatment for R/M HNSCC are associated with durable responses in a limited number of patients. The anti-EGFR mAb, cetuximab, has antitumor activity in this treatment setting, but responses are short-lived and inevitably curtailed due to treatment resistance. Crosstalk between the EGFR and hepatocyte growth factor-dependent mesenchymal-epithelial transition (c-MET) receptor tyrosine kinase pathway is a known mechanism of resistance to cetuximab. Dual targeting of EGFR and c-MET pathways may overcome resistance to cetuximab in patients with HPV-negative HNSCC. Here, we review clinical data of treatments evaluated in patients with R/M HPV-negative HNSCC and highlight the potential role of combining hepatocyte growth factor/c-MET and EGFR pathway inhibitors to overcome cetuximab resistance in this population.

尽管晚期头颈部鳞状细胞癌(HNSCC)的治疗取得了显著进展,但复发性和/或转移性(R/M)人乳头瘤病毒(HPV)阴性 HNSCC 患者的生存率仍然很低。HPV阴性患者预后较差的部分原因可能与细胞周期调节因子和肿瘤抑制因子的缺失以及非炎症和缺氧的肿瘤微环境有关,这两种因素都会导致耐药性和疾病进展。以抗程序性细胞死亡蛋白 1 为基础的治疗方案是目前治疗 R/M HNSCC 的标准疗法,但只有少数患者能获得持久的疗效。抗表皮生长因子受体(EGFR)单克隆抗体西妥昔单抗(cetuximab)在这种治疗环境中具有抗肿瘤活性,但疗效短暂,而且不可避免地会因耐药性而减弱。表皮生长因子受体(EGFR)与肝细胞生长因子(HGF)依赖性间充质-上皮转化(c-MET)受体酪氨酸激酶通路之间的串扰是已知的西妥昔单抗耐药机制。对表皮生长因子受体和c-MET通路进行双重靶向治疗可能会克服HPV阴性HNSCC患者对西妥昔单抗的耐药性。在此,我们回顾了对R/M型HPV阴性HNSCC患者进行治疗评估的临床数据,并强调了联合HGF/c-MET和表皮生长因子受体途径抑制剂在克服该人群西妥昔单抗耐药方面的潜在作用。
{"title":"Advanced Human Papillomavirus-Negative Head and Neck Squamous Cell Carcinoma: Unmet Need and Emerging Therapies.","authors":"Robin Park, Christine H Chung","doi":"10.1158/1535-7163.MCT-24-0281","DOIUrl":"10.1158/1535-7163.MCT-24-0281","url":null,"abstract":"<p><p>Despite notable progress in the treatment of advanced head and neck squamous cell carcinoma (HNSCC), survival remains poor in patients with recurrent and/or metastatic (R/M) human papillomavirus (HPV)-negative HNSCC. Worse outcomes in patients who are HPV-negative may be partly related to loss of cell-cycle regulators and tumor suppressors as well as a noninflamed and hypoxic tumor microenvironment, both of which contribute to treatment resistance and disease progression. Anti-programmed cell death protein 1-based regimens as current standard-of-care treatment for R/M HNSCC are associated with durable responses in a limited number of patients. The anti-EGFR mAb, cetuximab, has antitumor activity in this treatment setting, but responses are short-lived and inevitably curtailed due to treatment resistance. Crosstalk between the EGFR and hepatocyte growth factor-dependent mesenchymal-epithelial transition (c-MET) receptor tyrosine kinase pathway is a known mechanism of resistance to cetuximab. Dual targeting of EGFR and c-MET pathways may overcome resistance to cetuximab in patients with HPV-negative HNSCC. Here, we review clinical data of treatments evaluated in patients with R/M HPV-negative HNSCC and highlight the potential role of combining hepatocyte growth factor/c-MET and EGFR pathway inhibitors to overcome cetuximab resistance in this population.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1717-1730"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11612620/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291432","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
Response to systemic therapies in patient-derived cell lines from primary and recurrent adult granulosa cell tumors. 原发性和复发性成人颗粒细胞瘤患者衍生细胞系对全身疗法的反应。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-11-27 DOI: 10.1158/1535-7163.MCT-24-0223
Geertruid J Brink, Nizar Hami, Sander Mertens, Hans W Nijman, Luc Rcw van Lonkhuijzen, Eva Maria Roes, Christine A R Lok, Cornelis D de Kroon, Jurgen Mj Piek, Ward Hofhuis, Hugo J G Snippert, Jolijn Willemijntje Groeneweg, Petronella O Witteveen, Ronald P Zweemer

In patients with the rare adult-type granulosa cell tumors (aGCT), surgery is the primary treatment for both primary and recurrent disease. In cases of inoperable disease, systematic therapy is administered, but variable response rates and drug resistance complicate predicting the most effective therapy. Drug screen testing on patient-derived cell lines may offer a solution. In a national prospective study on aGCT, fresh tissue was cultured into 2D cell lines, testing 27 clinically and experimental drugs. Dose-response curves and synergy were calculated using GraphPad Prism and Compusyn software. We established 34 patient-derived cell lines from tissue of 20 adult granulosa cell tumor patients. Of these, seven patients had a primary diagnosis of adult granulosa cell tumor and 13 patients had recurrent disease. In eight patients multiple tumor locations were cultured. On each cell line 10 monotherapies and 17 combinations of drugs were tested. Carboplatin/gemcitabine showed efficacy and synergy in almost all patient-derived cell lines. Synergy could not be detected in the regular carboplatin/paclitaxel and carboplatin/etoposide combinations. Experimental combinations alpelisib/fulvestrant and alpelisib/gemcitabine showed efficacy of more than 75%. Drug screens on patient-derived tumor cell lines reflects the reality of the variable response of systemic therapy in aGCT patients. In future research, this technique may be used to personalize the systemic treatment of aGCT patients in a clinical study. The good response to carboplatin/gemcitabine in our patient-derived cell lines can then be confirmed in a clinical setting.

对于罕见的成人型颗粒细胞瘤(aGCT)患者,手术是治疗原发性和复发性疾病的主要方法。对于无法手术的病例,则采用系统治疗,但反应率和耐药性的变化使预测最有效的治疗方法变得复杂。对源自患者的细胞系进行药物筛选测试可能是一种解决方案。在一项关于 aGCT 的全国性前瞻性研究中,将新鲜组织培养成二维细胞系,测试了 27 种临床和实验药物。使用 GraphPad Prism 和 Compusyn 软件计算剂量反应曲线和协同作用。我们从 20 名成人颗粒细胞瘤患者的组织中建立了 34 个患者来源细胞系。其中,7 名患者初诊为成人颗粒细胞瘤,13 名患者病情复发。在 8 名患者中,对多个肿瘤位置进行了培养。在每个细胞系上测试了 10 种单一疗法和 17 种药物组合。卡铂/吉西他滨在几乎所有患者衍生细胞系中都显示出疗效和协同作用。常规的卡铂/紫杉醇和卡铂/依托泊苷组合无法产生协同作用。实验性的阿柏西尼/氟维司群和阿柏西尼/吉西他滨组合显示出 75% 以上的疗效。对患者来源的肿瘤细胞系进行药物筛选,反映了 aGCT 患者对全身治疗反应不一的现实。在未来的研究中,这项技术可用于临床研究中对 aGCT 患者进行个性化的全身治疗。我们的病人衍生细胞系对卡铂/吉西他滨的良好反应可以在临床环境中得到证实。
{"title":"Response to systemic therapies in patient-derived cell lines from primary and recurrent adult granulosa cell tumors.","authors":"Geertruid J Brink, Nizar Hami, Sander Mertens, Hans W Nijman, Luc Rcw van Lonkhuijzen, Eva Maria Roes, Christine A R Lok, Cornelis D de Kroon, Jurgen Mj Piek, Ward Hofhuis, Hugo J G Snippert, Jolijn Willemijntje Groeneweg, Petronella O Witteveen, Ronald P Zweemer","doi":"10.1158/1535-7163.MCT-24-0223","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0223","url":null,"abstract":"<p><p>In patients with the rare adult-type granulosa cell tumors (aGCT), surgery is the primary treatment for both primary and recurrent disease. In cases of inoperable disease, systematic therapy is administered, but variable response rates and drug resistance complicate predicting the most effective therapy. Drug screen testing on patient-derived cell lines may offer a solution. In a national prospective study on aGCT, fresh tissue was cultured into 2D cell lines, testing 27 clinically and experimental drugs. Dose-response curves and synergy were calculated using GraphPad Prism and Compusyn software. We established 34 patient-derived cell lines from tissue of 20 adult granulosa cell tumor patients. Of these, seven patients had a primary diagnosis of adult granulosa cell tumor and 13 patients had recurrent disease. In eight patients multiple tumor locations were cultured. On each cell line 10 monotherapies and 17 combinations of drugs were tested. Carboplatin/gemcitabine showed efficacy and synergy in almost all patient-derived cell lines. Synergy could not be detected in the regular carboplatin/paclitaxel and carboplatin/etoposide combinations. Experimental combinations alpelisib/fulvestrant and alpelisib/gemcitabine showed efficacy of more than 75%. Drug screens on patient-derived tumor cell lines reflects the reality of the variable response of systemic therapy in aGCT patients. In future research, this technique may be used to personalize the systemic treatment of aGCT patients in a clinical study. The good response to carboplatin/gemcitabine in our patient-derived cell lines can then be confirmed in a clinical setting.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142730738","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
Development and Characterization of a Lysosome-Targeting SLC3A2/PD-L1 Bispecific Antibody-Drug Conjugate for Enhanced Anti-Tumor Efficacy in Solid Tumors. 开发溶酶体靶向 SLC3A2/PD-L1 双特异性抗体-药物共轭物并确定其特性,增强实体瘤的抗肿瘤疗效
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-11-27 DOI: 10.1158/1535-7163.MCT-24-0319
Zeng Wang, Meijun Zheng, Mengyao Li, Huaqing Lu, Nanxi Liu, Yongdong Chen, Nian Yang, Wanqin Zeng, Yijun Dong, Jia Li, Zhixiong Zhu, Chen Yang, Zongliang Zhang, Qizhong Lu, Hexian Li, Liangxue Zhou, Hui Yang, Aiping Tong

Bispecific antibodies (BsAbs) and antibody-drug conjugates (ADCs) have shown significant promise in cancer treatment, enhancing drug selectivity and therapeutic efficacy as demonstrated in multiple clinical studies. Bispecific antibody-drug conjugates (BsADCs), which combine the targeting capabilities of BsAbs with the cytotoxic potential of ADCs, offer a novel approach to overcoming several challenges associated with ADCs, including limited internalization, off-target toxicity, and drug resistance. In this study, we identified solute carrier family 3 member 2 (SLC3A2) as a highly expressed protein in a variety of solid tumors, making it a promising therapeutic target. We developed a bispecific antibody targeting SLC3A2 and PD-L1, and conjugated it to monomethyl auristatin E (MMAE) to create the SLC3A2/PD-L1 BsADC. The SLC3A2/PD-L1 BsAb effectively blocked PD-1 binding to PD-L1 and activated T cells, while also facilitating lysosomal targeting and degradation of poorly internalized PD-L1 antibodies. The SLC3A2/PD-L1 BsADC demonstrated superior anti-tumor efficacy in PD-L1 low-expressing tumor cells compared to single-target ADCs in both in vitro studies and in multiple xenograft and immunocompetent mouse models. Overall, our engineered SLC3A2/PD-L1 BsADC exhibited enhanced internalization and improved tumor cell targeting, highlighting the potential of lysosome-targeting BsAbs in advancing ADC therapeutic strategies for solid tumors.

多项临床研究表明,双特异性抗体(BsAbs)和抗体药物共轭物(ADCs)在癌症治疗中大有可为,可提高药物选择性和疗效。双特异性抗体-药物共轭物(BsADCs)结合了 BsAbs 的靶向能力和 ADCs 的细胞毒性潜力,为克服与 ADCs 相关的一些挑战(包括有限的内化、脱靶毒性和耐药性)提供了一种新方法。在这项研究中,我们发现溶质运载家族 3 成员 2(SLC3A2)是一种在多种实体瘤中高表达的蛋白质,因此它是一个很有前景的治疗靶点。我们开发了一种靶向 SLC3A2 和 PD-L1 的双特异性抗体,并将其与单甲基金丝桃素 E(MMAE)共轭,制成了 SLC3A2/PD-L1 BsADC。SLC3A2/PD-L1 BsAb 能有效阻断 PD-1 与 PD-L1 和活化 T 细胞的结合,同时还能促进溶酶体靶向和降解内化不良的 PD-L1 抗体。与单靶点 ADC 相比,SLC3A2/PD-L1 BsADC 在体外研究以及多种异种移植和免疫功能健全的小鼠模型中对 PD-L1 低表达肿瘤细胞的抗肿瘤疗效都更胜一筹。总之,我们设计的 SLC3A2/PD-L1 BsADC 表现出更强的内化能力和更好的肿瘤细胞靶向性,突显了溶酶体靶向 BsAbs 在推进实体瘤 ADC 治疗策略方面的潜力。
{"title":"Development and Characterization of a Lysosome-Targeting SLC3A2/PD-L1 Bispecific Antibody-Drug Conjugate for Enhanced Anti-Tumor Efficacy in Solid Tumors.","authors":"Zeng Wang, Meijun Zheng, Mengyao Li, Huaqing Lu, Nanxi Liu, Yongdong Chen, Nian Yang, Wanqin Zeng, Yijun Dong, Jia Li, Zhixiong Zhu, Chen Yang, Zongliang Zhang, Qizhong Lu, Hexian Li, Liangxue Zhou, Hui Yang, Aiping Tong","doi":"10.1158/1535-7163.MCT-24-0319","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0319","url":null,"abstract":"<p><p>Bispecific antibodies (BsAbs) and antibody-drug conjugates (ADCs) have shown significant promise in cancer treatment, enhancing drug selectivity and therapeutic efficacy as demonstrated in multiple clinical studies. Bispecific antibody-drug conjugates (BsADCs), which combine the targeting capabilities of BsAbs with the cytotoxic potential of ADCs, offer a novel approach to overcoming several challenges associated with ADCs, including limited internalization, off-target toxicity, and drug resistance. In this study, we identified solute carrier family 3 member 2 (SLC3A2) as a highly expressed protein in a variety of solid tumors, making it a promising therapeutic target. We developed a bispecific antibody targeting SLC3A2 and PD-L1, and conjugated it to monomethyl auristatin E (MMAE) to create the SLC3A2/PD-L1 BsADC. The SLC3A2/PD-L1 BsAb effectively blocked PD-1 binding to PD-L1 and activated T cells, while also facilitating lysosomal targeting and degradation of poorly internalized PD-L1 antibodies. The SLC3A2/PD-L1 BsADC demonstrated superior anti-tumor efficacy in PD-L1 low-expressing tumor cells compared to single-target ADCs in both in vitro studies and in multiple xenograft and immunocompetent mouse models. Overall, our engineered SLC3A2/PD-L1 BsADC exhibited enhanced internalization and improved tumor cell targeting, highlighting the potential of lysosome-targeting BsAbs in advancing ADC therapeutic strategies for solid tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142730737","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