首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
A PD-L1/CD3 BISPECIFIC ANTIBODY ENHANCES THE ANTI-TUMOR EFFECTS OF REGORAFENIB AGAINST COLON CANCER.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-09 DOI: 10.1158/1535-7163.MCT-24-1015
Izuchukwu F Okpalanwaka, Elizabeth A Daugherity, Amanda L McCormick, Trevor S Anderson, Savanna L Smith, Caryn Lawrence, Duke Appiah, Devin B Lowe

Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. The current standard of care for patients may involve surgery, chemotherapy, and immune checkpoint inhibitors (ICIs), but these approaches typically fail to secure durable responses against late-stage disease. Regorafenib (REG) is an FDA-approved tyrosine kinase inhibitor with immunomodulating properties for CRC patients who progress on standard care, but 5-year relative survival rates for individuals dosed with the drug as a monotherapy are poor. We hypothesize that REG may be more appropriately leveraged alongside immunotherapeutic agents that specifically stimulate T cell infiltration and activation within the tumor microenvironment (TME). We engineered a PD-L1/CD3 bispecific antibody (bsAb) that simultaneously binds PD-L1-expressing CRC cells and stimulates activated T cells in order to investigate combination strategies with REG in pre-clinical models of CRC. Combined REG + bsAb therapy safely initiated and sustained inhibition against MC38 and CT26 progression in vivo, and these effects correlated to improved CD8+ T cell infiltration and activity within a Type-1-prone TME. Additionally, cytotoxic CD8+ T cells from REG + bsAb-sensitized mice exhibited heightened tumor cell reactivity compared to animals treated with either agent alone. Therefore, the immunomodulatory benefits of REG can be effectively paired with a bsAb that anchors to CRC cells, diminishes immunosuppression (through PD-L1 blockade), and activates/sustains antigen-specific CD8+ T cells within the TME. Our newly described REG + bsAb regimen led to improved anti-tumor outcomes pre-clinically and may represent a promising future approach for CRC patients.

{"title":"A PD-L1/CD3 BISPECIFIC ANTIBODY ENHANCES THE ANTI-TUMOR EFFECTS OF REGORAFENIB AGAINST COLON CANCER.","authors":"Izuchukwu F Okpalanwaka, Elizabeth A Daugherity, Amanda L McCormick, Trevor S Anderson, Savanna L Smith, Caryn Lawrence, Duke Appiah, Devin B Lowe","doi":"10.1158/1535-7163.MCT-24-1015","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-1015","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. The current standard of care for patients may involve surgery, chemotherapy, and immune checkpoint inhibitors (ICIs), but these approaches typically fail to secure durable responses against late-stage disease. Regorafenib (REG) is an FDA-approved tyrosine kinase inhibitor with immunomodulating properties for CRC patients who progress on standard care, but 5-year relative survival rates for individuals dosed with the drug as a monotherapy are poor. We hypothesize that REG may be more appropriately leveraged alongside immunotherapeutic agents that specifically stimulate T cell infiltration and activation within the tumor microenvironment (TME). We engineered a PD-L1/CD3 bispecific antibody (bsAb) that simultaneously binds PD-L1-expressing CRC cells and stimulates activated T cells in order to investigate combination strategies with REG in pre-clinical models of CRC. Combined REG + bsAb therapy safely initiated and sustained inhibition against MC38 and CT26 progression in vivo, and these effects correlated to improved CD8+ T cell infiltration and activity within a Type-1-prone TME. Additionally, cytotoxic CD8+ T cells from REG + bsAb-sensitized mice exhibited heightened tumor cell reactivity compared to animals treated with either agent alone. Therefore, the immunomodulatory benefits of REG can be effectively paired with a bsAb that anchors to CRC cells, diminishes immunosuppression (through PD-L1 blockade), and activates/sustains antigen-specific CD8+ T cells within the TME. Our newly described REG + bsAb regimen led to improved anti-tumor outcomes pre-clinically and may represent a promising future approach for CRC patients.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143811909","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
Broadening the Therapeutic Window of ADCs Using Site-Specific Bioconjugation Showcased by an MMAE-Containing Peptide Linker in a CD79b-Targeting ADC.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1158/1535-7163.MCT-24-0983
Philipp Probst, Isabella Attinger-Toller, Romain Bertrand, Ramona Stark, Roger Santimaria, Bernd Schlereth, Dragan Grabulovski, Philipp René Spycher

The limitations of first-generation antibody-drug conjugate (ADC) technologies include suboptimal stability and efficacy, poor safety profiles, and challenging manufacturing processes. In this study, we describe an anti-CD79b-monomethyl auristatin E (MMAE) ADC generated using a novel peptide-based linker technology that allows for site-specific linker-payload conjugation to native antibodies in only one step. The ADC comprises native polatuzumab as the targeting antibody and a linker-payload consisting of a RKAA-peptide linker and MMAE. We compared our anti-CD79b-RKAA-MMAE ADC with polatuzumab vedotin (PV), the FDA-approved ADC for diffuse large B-cell lymphoma. In the clinic, PV shows significant instability in circulation, leading to strong and dose-limiting side effects, including neutropenia and peripheral neuropathy. The anti-CD79b-RKAA-MMAE ADC showed optimal biophysical properties with a well-defined drug-to-antibody ratio of 2. It demonstrated potent cytotoxicity in multiple cancer cell lines and was very stable in mouse, cynomolgus monkey, and human sera. The anti-CD79b-RKAA-MMAE conjugate showed equal antitumor efficacy at half the payload dose compared with PV in different xenograft models. At equal MMAE concentrations, greater tumor growth inhibition and a considerably longer duration of response were observed. Ultimately, the highest nonseverely toxic dose of 30 mg/kg was determined in a 4-week repeat-dose toxicology study in rats, which is a 3-fold higher ADC dose than reported for PV. In summary, the data show that our novel site-specific bioconjugation technology enabled the generation of an anti-CD79b-RKAA-MMAE ADC with highly favorable biophysical properties and a greatly improved therapeutic index by a factor of 4 to 6 compared with PV. The ADC may therefore represent a safe and efficacious alternative for patients with diffuse large B-cell lymphoma.

第一代抗体药物共轭物(ADC)技术的局限性包括稳定性和疗效不理想、安全性差以及生产工艺具有挑战性。在本研究中,我们介绍了一种抗 CD79b-单甲基曙红 E(MMAE)ADC,它采用了一种新型肽基连接体技术,只需一个步骤就能将特定位点的连接体-负载连接到原生抗体上。这种 ADC 包括作为靶向抗体的原生 Polatuzumab 和由 RKAA 肽连接体和 MMAE 组成的连接体-负载。我们将抗 CD79b-RKAA-MMAE ADC 与 FDA 批准用于弥漫大 B 细胞淋巴瘤的 ADC--泊拉珠单抗维多汀(Polatuzumab vedotin,PV)进行了比较。在临床中,PV 在循环中表现出明显的不稳定性,导致强烈的剂量限制性副作用,包括中性粒细胞减少和周围神经病变。抗 CD79b-RKAA-MMAE ADC 表现出最佳的生物物理特性,药物与抗体的比例明确为 2,在多种癌细胞系中表现出强大的细胞毒性,在小鼠、猕猴和人类血清中非常稳定。在不同的异种移植模型中,抗 CD79b-RKAA-MMAE 结合物在有效载荷剂量为 PV 一半的情况下,与 PV 相比具有相同的抗肿瘤疗效。在相同的 MMAE 浓度下,可观察到更大的肿瘤生长抑制作用和更长的反应持续时间。最终,在对大鼠进行的为期 4 周的重复剂量毒理学研究中,确定了 30 毫克/千克的最高无剧毒剂量,这一 ADC 剂量是已报道的 PV 剂量的 3 倍。总之,这些数据表明,我们的新型位点特异性生物共轭技术能够生成一种抗 CD79b-RKAA-MMAE ADC,它具有非常有利的生物物理特性,与 PV 相比,治疗指数大大提高了 4 到 6 倍。因此,这种 ADC 可能是弥漫大 B 细胞淋巴瘤患者的一种安全有效的替代疗法。
{"title":"Broadening the Therapeutic Window of ADCs Using Site-Specific Bioconjugation Showcased by an MMAE-Containing Peptide Linker in a CD79b-Targeting ADC.","authors":"Philipp Probst, Isabella Attinger-Toller, Romain Bertrand, Ramona Stark, Roger Santimaria, Bernd Schlereth, Dragan Grabulovski, Philipp René Spycher","doi":"10.1158/1535-7163.MCT-24-0983","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0983","url":null,"abstract":"<p><p>The limitations of first-generation antibody-drug conjugate (ADC) technologies include suboptimal stability and efficacy, poor safety profiles, and challenging manufacturing processes. In this study, we describe an anti-CD79b-monomethyl auristatin E (MMAE) ADC generated using a novel peptide-based linker technology that allows for site-specific linker-payload conjugation to native antibodies in only one step. The ADC comprises native polatuzumab as the targeting antibody and a linker-payload consisting of a RKAA-peptide linker and MMAE. We compared our anti-CD79b-RKAA-MMAE ADC with polatuzumab vedotin (PV), the FDA-approved ADC for diffuse large B-cell lymphoma. In the clinic, PV shows significant instability in circulation, leading to strong and dose-limiting side effects, including neutropenia and peripheral neuropathy. The anti-CD79b-RKAA-MMAE ADC showed optimal biophysical properties with a well-defined drug-to-antibody ratio of 2. It demonstrated potent cytotoxicity in multiple cancer cell lines and was very stable in mouse, cynomolgus monkey, and human sera. The anti-CD79b-RKAA-MMAE conjugate showed equal antitumor efficacy at half the payload dose compared with PV in different xenograft models. At equal MMAE concentrations, greater tumor growth inhibition and a considerably longer duration of response were observed. Ultimately, the highest nonseverely toxic dose of 30 mg/kg was determined in a 4-week repeat-dose toxicology study in rats, which is a 3-fold higher ADC dose than reported for PV. In summary, the data show that our novel site-specific bioconjugation technology enabled the generation of an anti-CD79b-RKAA-MMAE ADC with highly favorable biophysical properties and a greatly improved therapeutic index by a factor of 4 to 6 compared with PV. The ADC may therefore represent a safe and efficacious alternative for patients with diffuse large B-cell lymphoma.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF13"},"PeriodicalIF":5.3,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143772654","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 Application of Radioactive Ligands Targeting Fibroblasts with Albumin-Binding Sites.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1158/1535-7163.MCT-24-1108
Tongtong Wu, Zhicong Yang, Sufan Tang, Hongmei Yuan, Yang Liu, Haiyang Li, Nan Liu, Zhanwen Huang, Yue Chen, Zhijun Zhou

Fibroblast activation protein (FAP) is overexpressed on cancer-related fibroblasts (CAFs), making it an important target for cancer diagnosis and treatment, but limited tumor retention hinders late-stage diagnosis and radionuclide therapy. In this study, three albumin-bound FAPI radioligands, 68Ga/177Lu-DOTA-ALB-01, 68Ga/177Lu-DOTA-ALB-02, and 68Ga/177Lu-DOTA-ALB-03, were synthesized and evaluated for their in vitro stability, binding affinity, in vivo biodistribution, and tumor uptake using 68Ga and 177Lu labeling. All radioligands are stable in saline and plasma and exhibit high FAP binding affinity. 177Lu-DOTA-ALB-02 has longer retention in circulation than 177Lu-FAPI-46 and other radioligands. Continuous tumor accumulation was observed during imaging with both 177Lu-DOTA-ALB-01 and 177Lu-DOTA-ALB-02. Notably, 177Lu-DOTA-ALB-02 had a significant tumor/ nontarget (T/NT) ratio as indicated by biodistribution data. The outstanding tumor retention properties of 177Lu-DOTA-ALB-02 have been demonstrated in small animal single photon emission computed tomography (micro-SPECT) imaging and biodistribution studies, therefore it is considered the albumin-binding FAPI with the most favorable pharmacokinetic and imaging properties, worthy of further clinical investigation.

{"title":"Development and Application of Radioactive Ligands Targeting Fibroblasts with Albumin-Binding Sites.","authors":"Tongtong Wu, Zhicong Yang, Sufan Tang, Hongmei Yuan, Yang Liu, Haiyang Li, Nan Liu, Zhanwen Huang, Yue Chen, Zhijun Zhou","doi":"10.1158/1535-7163.MCT-24-1108","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-1108","url":null,"abstract":"<p><p>Fibroblast activation protein (FAP) is overexpressed on cancer-related fibroblasts (CAFs), making it an important target for cancer diagnosis and treatment, but limited tumor retention hinders late-stage diagnosis and radionuclide therapy. In this study, three albumin-bound FAPI radioligands, 68Ga/177Lu-DOTA-ALB-01, 68Ga/177Lu-DOTA-ALB-02, and 68Ga/177Lu-DOTA-ALB-03, were synthesized and evaluated for their in vitro stability, binding affinity, in vivo biodistribution, and tumor uptake using 68Ga and 177Lu labeling. All radioligands are stable in saline and plasma and exhibit high FAP binding affinity. 177Lu-DOTA-ALB-02 has longer retention in circulation than 177Lu-FAPI-46 and other radioligands. Continuous tumor accumulation was observed during imaging with both 177Lu-DOTA-ALB-01 and 177Lu-DOTA-ALB-02. Notably, 177Lu-DOTA-ALB-02 had a significant tumor/ nontarget (T/NT) ratio as indicated by biodistribution data. The outstanding tumor retention properties of 177Lu-DOTA-ALB-02 have been demonstrated in small animal single photon emission computed tomography (micro-SPECT) imaging and biodistribution studies, therefore it is considered the albumin-binding FAPI with the most favorable pharmacokinetic and imaging properties, worthy of further clinical investigation.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143772588","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
Target-Specific Locked Nucleic Acid Gapmer Decreases Growth and Metastases of Pancreatic Cancer.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1158/1535-7163.MCT-24-1059
Jill P Smith, Narayan Shivapurkar, Wenqiang Chen, Godhanjali Chekuri, Amani Dabney, Kyle Holmes, Hong Cao, Ruvanthi N Kularatne, Stephan T Stern

Precision medicine and genomic profiling with target-specific therapy directed to cancer cell receptors have improved the outcome of many recalcitrant cancers. Strategies to deliver gene therapy to downregulate cancer driver genes have been challenging in vivo. Pancreatic cancer has the poorest survival of all solid tumors due to the lack of target-specific therapies and its characteristic tumor microenvironment with dense fibrosis and abundant immunosuppressive M2-polarized macrophages. In this study, we designed a panel of locked nucleic acid (LNA) gapmer antisense oligonucleotides (ASO) directed to human gastrin mRNA. We tested their efficacy by downregulation of mRNA and growth inhibition in vitro. The most effective, Gapmer-90, was modified for in vivo therapeutics by thiol-maleimide click chemistry to render it target-specific to the CCK-B receptor. This G-protein coupled receptor is over-expressed in pancreatic cancers. Mice bearing orthotopic human pancreatic tumors were treated with PBS (control), an untargeted gapmer, or receptor-targeted gapmers at low (60nM) and high (120nM) concentrations. Uptake of the gapmer was measured in tissues using a complementary probe. We found that the receptor-targeted gapmer significantly enhanced uptake in vivo and decreased growth and metastases of human pancreatic tumors in a dose-related fashion without off-target toxicity. The targeted-specific gapmer also altered the tumor microenvironment by decreasing fibrosis and reducing M2-polarized macrophages. Collectively, our results provide evidence that LNA gapmers are a unique tool to deliver antisense oligonucleotides for therapy to recalcitrant cancers. Rendering the gapmers target-specific allows for selective uptake by receptor internalization, improving efficacy, and decreasing off-target toxicity.

{"title":"Target-Specific Locked Nucleic Acid Gapmer Decreases Growth and Metastases of Pancreatic Cancer.","authors":"Jill P Smith, Narayan Shivapurkar, Wenqiang Chen, Godhanjali Chekuri, Amani Dabney, Kyle Holmes, Hong Cao, Ruvanthi N Kularatne, Stephan T Stern","doi":"10.1158/1535-7163.MCT-24-1059","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-1059","url":null,"abstract":"<p><p>Precision medicine and genomic profiling with target-specific therapy directed to cancer cell receptors have improved the outcome of many recalcitrant cancers. Strategies to deliver gene therapy to downregulate cancer driver genes have been challenging in vivo. Pancreatic cancer has the poorest survival of all solid tumors due to the lack of target-specific therapies and its characteristic tumor microenvironment with dense fibrosis and abundant immunosuppressive M2-polarized macrophages. In this study, we designed a panel of locked nucleic acid (LNA) gapmer antisense oligonucleotides (ASO) directed to human gastrin mRNA. We tested their efficacy by downregulation of mRNA and growth inhibition in vitro. The most effective, Gapmer-90, was modified for in vivo therapeutics by thiol-maleimide click chemistry to render it target-specific to the CCK-B receptor. This G-protein coupled receptor is over-expressed in pancreatic cancers. Mice bearing orthotopic human pancreatic tumors were treated with PBS (control), an untargeted gapmer, or receptor-targeted gapmers at low (60nM) and high (120nM) concentrations. Uptake of the gapmer was measured in tissues using a complementary probe. We found that the receptor-targeted gapmer significantly enhanced uptake in vivo and decreased growth and metastases of human pancreatic tumors in a dose-related fashion without off-target toxicity. The targeted-specific gapmer also altered the tumor microenvironment by decreasing fibrosis and reducing M2-polarized macrophages. Collectively, our results provide evidence that LNA gapmers are a unique tool to deliver antisense oligonucleotides for therapy to recalcitrant cancers. Rendering the gapmers target-specific allows for selective uptake by receptor internalization, improving efficacy, and decreasing off-target toxicity.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143772589","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
Pan-KRAS Inhibitors BI-2493 and BI-2865 Display Potent Antitumor Activity in Tumors with KRAS Wild-type Allele Amplification. Pan-KRAS抑制剂BI-2493和BI-2865在KRAS野生型等位基因扩增的肿瘤中显示出有效的抗肿瘤活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1158/1535-7163.MCT-24-0386
Antonio Tedeschi, Fiorella Schischlik, Francesca Rocchetti, Johannes Popow, Florian Ebner, Daniel Gerlach, Antonia Geyer, Valeria Santoro, Andrew S Boghossian, Matthew G Rees, Melissa M Ronan, Jennifer A Roth, Jesse Lipp, Matthias Samwer, Michael Gmachl, Norbert Kraut, Mark Pearson, Dorothea Rudolph

KRASG12C selective inhibitors, such as sotorasib and adagrasib, have raised hopes of targeting other KRAS-mutant alleles in patients with cancer. We report that KRAS wild-type (WT)-amplified tumor models are sensitive to treatment with the small-molecule KRAS inhibitors BI-2493 and BI-2865. These pan-KRAS inhibitors directly target the "OFF" state of KRAS and result in potent antitumor activity in preclinical models of cancers driven by KRAS-mutant proteins. In this study, we used the high-throughput cellular viability Profiling Relative Inhibition Simultaneously in Mixtures assay to assess the antiproliferative activity of BI-2493 in a 900+ cancer cell line panel, expanding on our previous work. KRAS WT-amplified cancer cell lines, with a copy number >7, were identified as the most sensitive, across cell lines with any KRAS alterations, to our pan-KRAS inhibitors. Importantly, our data suggest that a KRAS "OFF" inhibitor is better suited to treat KRAS WT-amplified tumors than a KRAS "ON" inhibitor. KRAS WT amplification is common in patients with gastroesophageal cancers in which it has been shown to act as a unique cancer driver with little overlap to other actionable mutations. The pan-KRAS inhibitors BI-2493 and BI-2865 show potent antitumor activity in vitro and in vivo in KRAS WT-amplified cell lines from this and other tumor types. In conclusion, this is the first study to demonstrate that direct pharmacologic inhibition of KRAS shows antitumor activity in preclinical models of cancer with KRAS WT amplification, suggesting a novel therapeutic concept for patients with cancers bearing this KRAS alteration.

KRASG12C选择性抑制剂,如sotorasib和adagrasib,已经为癌症患者靶向其他KRAS突变等位基因带来了希望。我们报道KRAS野生型扩增肿瘤模型对KRAS小分子抑制剂BI-2493和BI-2865治疗敏感。这些泛KRAS抑制剂直接靶向KRAS的“关闭”状态,并在由KRAS突变蛋白驱动的癌症临床前模型中产生有效的抗肿瘤活性。在这里,我们使用高通量细胞活力PRISM实验来评估BI-2493在900多个癌细胞系面板中的抗增殖活性,扩展了我们之前的工作。KRAS野生型扩增的癌细胞系,拷贝数为>7,被鉴定为对我们的泛KRAS抑制剂最敏感,跨越任何KRAS改变的细胞系。重要的是,我们的数据表明,KRAS“OFF”抑制剂比KRAS“ON”抑制剂更适合治疗KRAS野生型扩增肿瘤。KRAS野生型扩增在胃食管癌患者中很常见,它已被证明是一种独特的癌症驱动因素,与其他可操作的突变几乎没有重叠。泛KRAS抑制剂BI-2493和BI-2865在体外和体内对KRAS野生型扩增细胞系和其他肿瘤类型显示出有效的抗肿瘤活性。总之,这是第一个证明直接药理抑制KRAS在KRAS野生型扩增的癌症临床前模型中具有抗肿瘤活性的研究,为携带这种KRAS突变的癌症患者提供了一种新的治疗理念。
{"title":"Pan-KRAS Inhibitors BI-2493 and BI-2865 Display Potent Antitumor Activity in Tumors with KRAS Wild-type Allele Amplification.","authors":"Antonio Tedeschi, Fiorella Schischlik, Francesca Rocchetti, Johannes Popow, Florian Ebner, Daniel Gerlach, Antonia Geyer, Valeria Santoro, Andrew S Boghossian, Matthew G Rees, Melissa M Ronan, Jennifer A Roth, Jesse Lipp, Matthias Samwer, Michael Gmachl, Norbert Kraut, Mark Pearson, Dorothea Rudolph","doi":"10.1158/1535-7163.MCT-24-0386","DOIUrl":"10.1158/1535-7163.MCT-24-0386","url":null,"abstract":"<p><p>KRASG12C selective inhibitors, such as sotorasib and adagrasib, have raised hopes of targeting other KRAS-mutant alleles in patients with cancer. We report that KRAS wild-type (WT)-amplified tumor models are sensitive to treatment with the small-molecule KRAS inhibitors BI-2493 and BI-2865. These pan-KRAS inhibitors directly target the \"OFF\" state of KRAS and result in potent antitumor activity in preclinical models of cancers driven by KRAS-mutant proteins. In this study, we used the high-throughput cellular viability Profiling Relative Inhibition Simultaneously in Mixtures assay to assess the antiproliferative activity of BI-2493 in a 900+ cancer cell line panel, expanding on our previous work. KRAS WT-amplified cancer cell lines, with a copy number >7, were identified as the most sensitive, across cell lines with any KRAS alterations, to our pan-KRAS inhibitors. Importantly, our data suggest that a KRAS \"OFF\" inhibitor is better suited to treat KRAS WT-amplified tumors than a KRAS \"ON\" inhibitor. KRAS WT amplification is common in patients with gastroesophageal cancers in which it has been shown to act as a unique cancer driver with little overlap to other actionable mutations. The pan-KRAS inhibitors BI-2493 and BI-2865 show potent antitumor activity in vitro and in vivo in KRAS WT-amplified cell lines from this and other tumor types. In conclusion, this is the first study to demonstrate that direct pharmacologic inhibition of KRAS shows antitumor activity in preclinical models of cancer with KRAS WT amplification, suggesting a novel therapeutic concept for patients with cancers bearing this KRAS alteration.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"550-562"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11962398/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142877408","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
Identification of a TNIK-CDK9 Axis as a Targetable Strategy for Platinum-Resistant Ovarian Cancer.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1158/1535-7163.MCT-24-0785
Noah Puleo, Harini Ram, Michele L Dziubinski, Dylan Carvette, Jessica Teitel, Sreeja C Sekhar, Karan Bedi, Aaron Robida, Michael M Nakashima, Sadaf Farsinejad, Marcin Iwanicki, Wojciech Senkowski, Arpita Ray, Thomas J Bollerman, James Dunbar, Peter Richardson, Andrea Taddei, Chantelle Hudson, Analisa DiFeo

Up to 90% of patients with high-grade serous ovarian cancer (HGSC) will develop resistance to platinum-based chemotherapy, posing substantial therapeutic challenges due to a lack of universally druggable targets. Leveraging BenevolentAI's artificial intelligence (AI)-driven approach to target discovery, we screened potential AI-predicted therapeutic targets mapped to unapproved tool compounds in patient-derived 3D models. This identified TNIK, which is modulated by NCB-0846, as a novel target for platinum-resistant HGSC. Targeting by this compound demonstrated efficacy across both in vitro and ex vivo organoid platinum-resistant models. Additionally, NCB-0846 treatment effectively decreased Wnt activity, a known driver of platinum resistance; however, we found that these effects were not solely mediated by TNIK inhibition. Comprehensive AI, in silico, and in vitro analyses revealed CDK9 as another key target driving NCB-0846's efficacy. Interestingly, TNIK and CDK9 co-expression positively correlated, and chromosomal gains in both served as prognostic markers for poor patient outcomes. Combined knockdown of TNIK and CDK9 markedly diminished downstream Wnt targets and reduced chemotherapy-resistant cell viability. Furthermore, we identified CDK9 as a novel mediator of canonical Wnt activity, providing mechanistic insights into the combinatorial effects of TNIK and CDK9 inhibition and offering a new understanding of NCB-0846 and CDK9 inhibitor function. Our findings identified the TNIK-CDK9 axis as druggable targets mediating platinum resistance and cell viability in HGSC. With AI at the forefront of drug discovery, this work highlights how to ensure that AI findings are biologically relevant by combining compound screens with physiologically relevant models, thus supporting the identification and validation of potential drug targets.

{"title":"Identification of a TNIK-CDK9 Axis as a Targetable Strategy for Platinum-Resistant Ovarian Cancer.","authors":"Noah Puleo, Harini Ram, Michele L Dziubinski, Dylan Carvette, Jessica Teitel, Sreeja C Sekhar, Karan Bedi, Aaron Robida, Michael M Nakashima, Sadaf Farsinejad, Marcin Iwanicki, Wojciech Senkowski, Arpita Ray, Thomas J Bollerman, James Dunbar, Peter Richardson, Andrea Taddei, Chantelle Hudson, Analisa DiFeo","doi":"10.1158/1535-7163.MCT-24-0785","DOIUrl":"10.1158/1535-7163.MCT-24-0785","url":null,"abstract":"<p><p>Up to 90% of patients with high-grade serous ovarian cancer (HGSC) will develop resistance to platinum-based chemotherapy, posing substantial therapeutic challenges due to a lack of universally druggable targets. Leveraging BenevolentAI's artificial intelligence (AI)-driven approach to target discovery, we screened potential AI-predicted therapeutic targets mapped to unapproved tool compounds in patient-derived 3D models. This identified TNIK, which is modulated by NCB-0846, as a novel target for platinum-resistant HGSC. Targeting by this compound demonstrated efficacy across both in vitro and ex vivo organoid platinum-resistant models. Additionally, NCB-0846 treatment effectively decreased Wnt activity, a known driver of platinum resistance; however, we found that these effects were not solely mediated by TNIK inhibition. Comprehensive AI, in silico, and in vitro analyses revealed CDK9 as another key target driving NCB-0846's efficacy. Interestingly, TNIK and CDK9 co-expression positively correlated, and chromosomal gains in both served as prognostic markers for poor patient outcomes. Combined knockdown of TNIK and CDK9 markedly diminished downstream Wnt targets and reduced chemotherapy-resistant cell viability. Furthermore, we identified CDK9 as a novel mediator of canonical Wnt activity, providing mechanistic insights into the combinatorial effects of TNIK and CDK9 inhibition and offering a new understanding of NCB-0846 and CDK9 inhibitor function. Our findings identified the TNIK-CDK9 axis as druggable targets mediating platinum resistance and cell viability in HGSC. With AI at the forefront of drug discovery, this work highlights how to ensure that AI findings are biologically relevant by combining compound screens with physiologically relevant models, thus supporting the identification and validation of potential drug targets.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"639-656"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11962390/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143053056","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
Novel Amanitin-Based Antibody-Drug Conjugates Targeting TROP2 for the Treatment of Pancreatic Cancer. 用于治疗胰腺癌的靶向 TROP2 的新型鹅膏蕈素类抗体药物结合物 (ATAC®)。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1158/1535-7163.MCT-24-0266
Eleni Papacharisi, Alexandra C Braun, Marija Vranic, Andreas M Pahl, Torsten Hechler

Trophoblast cell surface antigen 2 (TROP2) exhibits aberrant expression in pancreatic cancer, correlating with metastasis, advanced tumor stage, and poor prognosis in patients with pancreatic ductal adenocarcinoma. TROP2 has been recognized as a promising therapeutic target for antibody-drug conjugates (ADC), as evidenced by the approval of the anti-TROP2 ADC Trodelvy for the treatment of triple-negative breast cancer (TNBC). In this study, we report the generation of novel second-generation amanitin-based ADCs (ATAC) targeting TROP2, comprising the humanized RS7 antibody of Trodelvy (hRS7) and the highly potent payload amanitin. The specific in vitro binding, efficient antigen internalization, and high cytotoxicity of hRS7 ATACs with EC50 values in the picomolar range in TROP2-expressing cells constituted the foundation for preclinical in vivo evaluation. The hRS7 ATACs demonstrated a significant reduction in tumor growth in vivo in subcutaneous xenograft mouse models of pancreatic cancer and TNBC at well-tolerated doses. The antitumor efficacy correlated with the level of TROP2 expression on the tumors and the in vivo tumor uptake of the ATACs. The long half-life of 9.7 to 10.7 days of hRS7 ATACs without premature payload release in serum supported a high therapeutic index. Notably, the efficacy of the hRS7 ATACs was superior to that of Trodelvy with complete tumor eradication in both refractory pancreatic cancer and TNBC xenograft models. In summary, hRS7 ATACs represent a highly effective and well-tolerated targeted therapy, and our data support their development for pancreatic cancer and other TROP2-expressing tumors.

滋养层细胞表面抗原 2(TROP2)在胰腺癌中异常表达,与胰腺导管腺癌(PDAC)患者的转移、肿瘤晚期和预后不良有关。抗 TROP2 ADC Trodelvy® 已被批准用于治疗三阴性乳腺癌,由此可见,TROP2 已被认为是抗体药物共轭物(ADC)的一个有前景的治疗靶点。在本研究中,我们报告了以TROP2为靶点的新型第二代基于amanitin的ADC(ATAC®s)的生成情况,该ADC由Trodelvy®的人源化RS7抗体(hRS7)和高效力有效载荷amanitin组成。hRS7 ATAC®s 具有特异性体外结合、高效抗原内化和高细胞毒性,在表达 TROP2 的细胞中的半数最大有效浓度(EC50)值在皮摩尔范围内,这为临床前体内评估奠定了基础。在胰腺癌和三阴性乳腺癌皮下异种移植小鼠模型中,hRS7 ATAC®s 以良好的耐受剂量显著降低了体内肿瘤的生长。抗肿瘤效果与肿瘤上 TROP2 的表达水平和 ATAC®s 在体内的肿瘤吸收率相关。hRS7 ATAC®s 的半衰期长达 9.7-10.7 天,不会在血清中过早释放有效载荷,因此具有很高的治疗指数。值得注意的是,在难治性胰腺癌和三阴性乳腺癌异种移植模型中,hRS7 ATAC®s 的疗效优于 Trodelvy®,能完全根除肿瘤。总之,hRS7 ATAC®s 是一种高效且耐受性良好的靶向疗法,我们的数据支持将其用于胰腺癌和其他表达 TROP2 的肿瘤。
{"title":"Novel Amanitin-Based Antibody-Drug Conjugates Targeting TROP2 for the Treatment of Pancreatic Cancer.","authors":"Eleni Papacharisi, Alexandra C Braun, Marija Vranic, Andreas M Pahl, Torsten Hechler","doi":"10.1158/1535-7163.MCT-24-0266","DOIUrl":"10.1158/1535-7163.MCT-24-0266","url":null,"abstract":"<p><p>Trophoblast cell surface antigen 2 (TROP2) exhibits aberrant expression in pancreatic cancer, correlating with metastasis, advanced tumor stage, and poor prognosis in patients with pancreatic ductal adenocarcinoma. TROP2 has been recognized as a promising therapeutic target for antibody-drug conjugates (ADC), as evidenced by the approval of the anti-TROP2 ADC Trodelvy for the treatment of triple-negative breast cancer (TNBC). In this study, we report the generation of novel second-generation amanitin-based ADCs (ATAC) targeting TROP2, comprising the humanized RS7 antibody of Trodelvy (hRS7) and the highly potent payload amanitin. The specific in vitro binding, efficient antigen internalization, and high cytotoxicity of hRS7 ATACs with EC50 values in the picomolar range in TROP2-expressing cells constituted the foundation for preclinical in vivo evaluation. The hRS7 ATACs demonstrated a significant reduction in tumor growth in vivo in subcutaneous xenograft mouse models of pancreatic cancer and TNBC at well-tolerated doses. The antitumor efficacy correlated with the level of TROP2 expression on the tumors and the in vivo tumor uptake of the ATACs. The long half-life of 9.7 to 10.7 days of hRS7 ATACs without premature payload release in serum supported a high therapeutic index. Notably, the efficacy of the hRS7 ATACs was superior to that of Trodelvy with complete tumor eradication in both refractory pancreatic cancer and TNBC xenograft models. In summary, hRS7 ATACs represent a highly effective and well-tolerated targeted therapy, and our data support their development for pancreatic cancer and other TROP2-expressing tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"485-496"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11962393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676379","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
Novel Combination Therapy Targeting Oncogenic Signaling Kinase P21-Activated Kinase 1 and Chemotherapeutic Drugs Against Triple-Negative Breast Cancer. 针对致癌信号激酶P21活化激酶-1和化疗药物联合治疗三阴性乳腺癌的新疗法。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1158/1535-7163.MCT-24-0404
Inemai Ezhil, Abirami Seetharaman, Rahul Kanumuri, Barathidasan Rajamani, Ranga Rao Gangavarapu, Ganesh Venkatraman, Suresh K Rayala

Most of the triple-negative phenotypes or basal-like molecular subtypes of breast cancers are associated with aggressive clinical behavior and show poor disease prognosis. Current treatment options are constrained, emphasizing the need for novel combinatorial therapies for this particular tumor subtype. Our group has demonstrated that functionally active p21-activated kinase 1 (PAK1) exhibits significantly higher expression levels in clinical triple-negative breast cancer (TNBC) samples compared with other subtypes, as well as adjacent normal tissues. Low PAK1 expression in TNBC was significantly linked to better prognosis, with improved overall survival (P = 0.00236) and relapse-free survival (P = 0.0314), as shown by Gene expression-based Outcome for Breast cancer Online analysis. To confirm the role of PAK1 as a therapeutic target and to discover novel synergistic chemotherapy drug combinations, we conducted a drug combination screen using TNBC cell lines and a mouse metastatic tumor cell line. We identified the combined inhibition of PAK1 inhibitor NVS-PAK1 with doxorubicin/paclitaxel/methotrexate as a synergistic novel therapeutic approach for treating metastatic TNBC to improve overall survival. This study also indicated a reduction in the effective dosage of the chemotherapeutic drug when combined with NVS-PAK1. Our study demonstrates that combining NVS-PAK1 with each individual chemotherapeutic drug such as doxorubicin, paclitaxel, and methotrexate resulted in decreased colony formation, reduced wound-healing capability, and diminished migratory and invasive potential in both TNBC cell lines and 4T1 in vitro. These findings were further validated in orthotopic mouse mammary tumors, confirming that simultaneous PAK1 inhibition alongside chemotherapy significantly enhanced antitumor efficacy and reduced metastasis.

大多数乳腺癌的三阴性表型或基底样分子亚型与侵袭性临床行为相关,疾病预后较差。目前的治疗方案是有限的,强调需要新的组合治疗这种特殊的肿瘤亚型。我们的研究小组已经证明,功能活跃的p21活化激酶1 (PAK1)在临床三阴性乳腺癌(TNBC)样本中的表达水平明显高于其他亚型以及邻近的正常组织。GOBO分析显示,TNBC中PAK1的低表达与更好的预后显著相关,总生存期(OS, p=0.00236)和无复发生存期(RFS, p=0.0314)均有改善。为了确认PAK1作为治疗靶点的作用,并发现新的协同化疗药物组合,我们使用三阴性乳腺癌细胞系和小鼠转移性肿瘤细胞系进行了药物联合筛选。我们发现联合抑制PAK1抑制剂NVS-PAK1与阿霉素/紫杉醇/甲氨蝶呤作为一种新的协同治疗方法来治疗转移性TNBC,以提高总生存率。该研究还表明,当与NVS-PAK1联合使用时,化疗药物的有效剂量降低。我们的研究表明,NVS-PAK1与每种化疗药物的阿霉素、紫杉醇和甲氨蝶呤联合使用,可减少TNBC细胞系和4T1的集落形成,降低伤口愈合能力,降低迁移和侵袭潜力。这些发现在原位小鼠乳腺肿瘤中得到进一步验证,证实在化疗的同时抑制PAK1可显著增强抗肿瘤疗效并减少转移。
{"title":"Novel Combination Therapy Targeting Oncogenic Signaling Kinase P21-Activated Kinase 1 and Chemotherapeutic Drugs Against Triple-Negative Breast Cancer.","authors":"Inemai Ezhil, Abirami Seetharaman, Rahul Kanumuri, Barathidasan Rajamani, Ranga Rao Gangavarapu, Ganesh Venkatraman, Suresh K Rayala","doi":"10.1158/1535-7163.MCT-24-0404","DOIUrl":"10.1158/1535-7163.MCT-24-0404","url":null,"abstract":"<p><p>Most of the triple-negative phenotypes or basal-like molecular subtypes of breast cancers are associated with aggressive clinical behavior and show poor disease prognosis. Current treatment options are constrained, emphasizing the need for novel combinatorial therapies for this particular tumor subtype. Our group has demonstrated that functionally active p21-activated kinase 1 (PAK1) exhibits significantly higher expression levels in clinical triple-negative breast cancer (TNBC) samples compared with other subtypes, as well as adjacent normal tissues. Low PAK1 expression in TNBC was significantly linked to better prognosis, with improved overall survival (P = 0.00236) and relapse-free survival (P = 0.0314), as shown by Gene expression-based Outcome for Breast cancer Online analysis. To confirm the role of PAK1 as a therapeutic target and to discover novel synergistic chemotherapy drug combinations, we conducted a drug combination screen using TNBC cell lines and a mouse metastatic tumor cell line. We identified the combined inhibition of PAK1 inhibitor NVS-PAK1 with doxorubicin/paclitaxel/methotrexate as a synergistic novel therapeutic approach for treating metastatic TNBC to improve overall survival. This study also indicated a reduction in the effective dosage of the chemotherapeutic drug when combined with NVS-PAK1. Our study demonstrates that combining NVS-PAK1 with each individual chemotherapeutic drug such as doxorubicin, paclitaxel, and methotrexate resulted in decreased colony formation, reduced wound-healing capability, and diminished migratory and invasive potential in both TNBC cell lines and 4T1 in vitro. These findings were further validated in orthotopic mouse mammary tumors, confirming that simultaneous PAK1 inhibition alongside chemotherapy significantly enhanced antitumor efficacy and reduced metastasis.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"576-586"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142971555","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
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 : 2025-04-02 DOI: 10.1158/1535-7163.MCT-24-0223
Geertruid J Brink, Nizar Hami, Sander Mertens, Hans W Nijman, Luc R C W van Lonkhuijzen, Eva Maria Roes, Christine A R Lok, Cornelis D de Kroon, Jurgen M J Piek, Ward Hofhuis, Hugo J G Snippert, Jolijn W Groeneweg, Petronella O Witteveen, Ronald P Zweemer

In patients with the rare adult-type granulosa cell tumor (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 clinical 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 patients with aGCT. Of these, seven patients had a primary diagnosis of aGCT 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 reflect the reality of the variable response of systemic therapy in patients with aGCT. In future research, this technique may be used to personalize the systemic treatment of patients with aGCT 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 R C W van Lonkhuijzen, Eva Maria Roes, Christine A R Lok, Cornelis D de Kroon, Jurgen M J Piek, Ward Hofhuis, Hugo J G Snippert, Jolijn W Groeneweg, Petronella O Witteveen, Ronald P Zweemer","doi":"10.1158/1535-7163.MCT-24-0223","DOIUrl":"10.1158/1535-7163.MCT-24-0223","url":null,"abstract":"<p><p>In patients with the rare adult-type granulosa cell tumor (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 clinical 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 patients with aGCT. Of these, seven patients had a primary diagnosis of aGCT 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 reflect the reality of the variable response of systemic therapy in patients with aGCT. In future research, this technique may be used to personalize the systemic treatment of patients with aGCT 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":"628-638"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","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
Preclinical Development of SGN-CD47M: Protease-Activated Antibody Technology Enables Selective Tumor Targeting of the Innate Immune Checkpoint Receptor CD47. SGN-CD47M 的临床前开发:蛋白酶激活抗体技术实现了先天性免疫检查点受体 CD47 的选择性肿瘤靶向。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1158/1535-7163.MCT-24-0371
Matthew R Levengood, Christopher M Carosino, Xinqun Zhang, Sasha Lucas, David J Ortiz, Lori Westendorf, Alice P Chin, Arlan D Martin, Abbie Wong, Shawna M Hengel, Hao Sun, Weiping Zeng, Roma Yumul, Melissa M C Dominguez, Yufei Chen, Janet H Zheng, Courtney A B Karlsson, Vivian H Trang, Peter D Senter, Shyra J Gardai

CD47 is a cell-surface glycoprotein that is expressed on normal human tissues and plays a key role as a marker of self. Tumor cells have co-opted CD47 overexpression to evade immune surveillance, and thus blockade of CD47 is a highly active area of clinical exploration in oncology. However, clinical development of CD47-targeted agents has been complicated by its robust expression in normal tissues and the toxicities that arise from blocking this inhibitory signal. Furthermore, pro-phagocytic signals are not uniformly expressed in tumors, and antibody blockade alone is often not sufficient to drive antitumor activity. The inclusion of an IgG1 antibody backbone into therapeutic design has been shown to not only serve as an additional pro-phagocytic signal but also exacerbate toxicities in normal tissues. Therefore, a need persists for more selective therapeutic modalities targeting CD47. To address these challenges, we developed SGN-CD47M, a humanized anti-CD47 IgG1 mAb linked to novel masking peptides through linkers designed to be cleaved by active proteases enriched in the tumor microenvironment (TME). Masking technology has the potential to increase the amount of drug that reaches the TME while concomitantly reducing systemic toxicities. We demonstrate that SGN-CD47M is well tolerated in cynomolgus monkeys and displays a 20-fold improvement in tolerability to hematologic toxicities when compared with the unmasked antibody. SGN-CD47M also displays preferential activation in the TME that leads to robust single-agent antitumor activity. For these reasons, SGN-CD47M may have enhanced antitumor activity and improved tolerability relative to existing therapies that target the CD47-signal regulatory protein α interaction.

CD47 是一种细胞表面糖蛋白,在正常人体组织中表达,作为自身标志物起着关键作用。肿瘤细胞借助 CD47 的过度表达来逃避免疫监视,因此阻断 CD47 是肿瘤学中一个非常活跃的临床探索领域。然而,由于 CD47 在正常组织中的强表达以及阻断这种抑制信号所产生的毒性,CD47 靶向药物的临床开发变得复杂起来。此外,促吞噬信号在肿瘤中的表达并不一致,仅靠抗体阻断往往不足以激发抗肿瘤活性。在治疗设计中加入 IgG1 抗体骨架已被证明可作为额外的促吞噬信号,但也会加重正常组织的毒性。因此,针对 CD47 的更具选择性的治疗模式仍有存在的必要。为了应对这些挑战,我们开发了 SGN-CD47M,这是一种人源化的抗 CD47 IgG1 单克隆抗体,通过设计成可被肿瘤微环境中富集的活性蛋白酶裂解的连接体与新型掩蔽肽相连。掩蔽技术有可能增加到达肿瘤微环境的药物量,同时降低全身毒性。我们的研究表明,SGN-CD47M 在猴体内耐受性良好,与未掩蔽抗体相比,血液毒性耐受性提高了 20 倍。SGN-CD47M 还能优先激活肿瘤微环境,从而产生强大的单药抗肿瘤活性。由于这些原因,与针对 CD47-SIRPα 相互作用的现有疗法相比,SGN-CD47M 可能具有更强的抗肿瘤活性和更好的耐受性。
{"title":"Preclinical Development of SGN-CD47M: Protease-Activated Antibody Technology Enables Selective Tumor Targeting of the Innate Immune Checkpoint Receptor CD47.","authors":"Matthew R Levengood, Christopher M Carosino, Xinqun Zhang, Sasha Lucas, David J Ortiz, Lori Westendorf, Alice P Chin, Arlan D Martin, Abbie Wong, Shawna M Hengel, Hao Sun, Weiping Zeng, Roma Yumul, Melissa M C Dominguez, Yufei Chen, Janet H Zheng, Courtney A B Karlsson, Vivian H Trang, Peter D Senter, Shyra J Gardai","doi":"10.1158/1535-7163.MCT-24-0371","DOIUrl":"10.1158/1535-7163.MCT-24-0371","url":null,"abstract":"<p><p>CD47 is a cell-surface glycoprotein that is expressed on normal human tissues and plays a key role as a marker of self. Tumor cells have co-opted CD47 overexpression to evade immune surveillance, and thus blockade of CD47 is a highly active area of clinical exploration in oncology. However, clinical development of CD47-targeted agents has been complicated by its robust expression in normal tissues and the toxicities that arise from blocking this inhibitory signal. Furthermore, pro-phagocytic signals are not uniformly expressed in tumors, and antibody blockade alone is often not sufficient to drive antitumor activity. The inclusion of an IgG1 antibody backbone into therapeutic design has been shown to not only serve as an additional pro-phagocytic signal but also exacerbate toxicities in normal tissues. Therefore, a need persists for more selective therapeutic modalities targeting CD47. To address these challenges, we developed SGN-CD47M, a humanized anti-CD47 IgG1 mAb linked to novel masking peptides through linkers designed to be cleaved by active proteases enriched in the tumor microenvironment (TME). Masking technology has the potential to increase the amount of drug that reaches the TME while concomitantly reducing systemic toxicities. We demonstrate that SGN-CD47M is well tolerated in cynomolgus monkeys and displays a 20-fold improvement in tolerability to hematologic toxicities when compared with the unmasked antibody. SGN-CD47M also displays preferential activation in the TME that leads to robust single-agent antitumor activity. For these reasons, SGN-CD47M may have enhanced antitumor activity and improved tolerability relative to existing therapies that target the CD47-signal regulatory protein α interaction.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"471-484"},"PeriodicalIF":5.3,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11962404/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504289","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
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