首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
CD47/SIRPα Immune Checkpoint Modulation: A Synergistic Strategy for Next-Generation CAR Therapies. CD47/SIRPα免疫检查点调节:新一代CAR治疗的协同策略
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-10 DOI: 10.1158/1535-7163.MCT-25-0531
Mohammad Javad Yousefi-Hashemabad, Amirhossein Kamroo, Ali Rezvanimehr, Kiarash Saleki, Erfan Barootchi, Aida Mehrani, Alireza Kordi, Andia Saleki, Pardis Zamani, Amirhossein Bazdar, Abdolrahman S Nateri, Shayan Barootchi, Nima Rezaei

Cancer immunotherapy has been revolutionized through the implementation of the state-of-the-art "chimeric antigen receptor" (CAR)-mediated therapies. CAR-based technologies, which encompass CAR T cells, CAR macrophages, and CAR-NK cells, show great promise in the treatment of various cancers. Despite the success of CAR-based therapies in treating malignancies, they face numerous challenges, including dysfunction of effector innate and adaptive immune cells, immunosuppressive tumor microenvironment (TME), antigen heterogeneity, and on-target/off-tumor bio-toxicity. The CD47/SIRPα axis is recognized as a critical innate immune checkpoint and is important in regulating myeloid-derived clearance of tumor cells and the innate-adaptive cells' cross-talk in cancer immunity. This signaling axis has risen as a promising target to boost the CAR-based immunotherapies by overcoming phagocytic inhibition and modulating immune evasion. This narrative review explores the integration of CD47/SIRPα modulation as an adjunct to CAR therapies. CD47/SIRPα immune-modulation revealed its potential to boost infiltration, persistence, and phagocytic activity of the immune cells. However, its blockade also poses challenges, including hematologic toxicities, CAR T cell clearance, and compensatory escape pathways. Future work will depend on selective targeting, combinatorial checkpoint modulation, and engineered CAR designs that preserve safety while unlocking durable responses. Herein, we discuss pre-clinical and clinical advancements, safety considerations, and cutting-edge advancements.

通过实施最先进的“嵌合抗原受体”(CAR)介导的治疗,癌症免疫治疗已经发生了革命性的变化。基于CAR的技术,包括CAR T细胞、CAR巨噬细胞和CAR nk细胞,在治疗各种癌症方面显示出巨大的希望。尽管基于car的疗法在治疗恶性肿瘤方面取得了成功,但它们面临着许多挑战,包括效应先天和适应性免疫细胞的功能障碍、免疫抑制肿瘤微环境(TME)、抗原异质性以及靶/肿瘤外生物毒性。CD47/SIRPα轴被认为是一个重要的先天免疫检查点,在调节肿瘤细胞的髓源性清除和先天适应性细胞在癌症免疫中的相互作用中起重要作用。这个信号轴已经成为一个有希望的靶点,通过克服吞噬抑制和调节免疫逃避来促进基于car的免疫疗法。这篇叙述性综述探讨了CD47/SIRPα调节作为CAR治疗辅助的整合。CD47/SIRPα免疫调节揭示了其增强免疫细胞浸润、持久性和吞噬活性的潜力。然而,它的阻断也带来了挑战,包括血液学毒性、CAR - T细胞清除和代偿性逃逸途径。未来的工作将取决于选择性靶向、组合检查点调制和工程化CAR设计,以在解锁持久响应的同时保持安全性。在这里,我们讨论临床前和临床进展,安全考虑和前沿进展。
{"title":"CD47/SIRPα Immune Checkpoint Modulation: A Synergistic Strategy for Next-Generation CAR Therapies.","authors":"Mohammad Javad Yousefi-Hashemabad, Amirhossein Kamroo, Ali Rezvanimehr, Kiarash Saleki, Erfan Barootchi, Aida Mehrani, Alireza Kordi, Andia Saleki, Pardis Zamani, Amirhossein Bazdar, Abdolrahman S Nateri, Shayan Barootchi, Nima Rezaei","doi":"10.1158/1535-7163.MCT-25-0531","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0531","url":null,"abstract":"<p><p>Cancer immunotherapy has been revolutionized through the implementation of the state-of-the-art \"chimeric antigen receptor\" (CAR)-mediated therapies. CAR-based technologies, which encompass CAR T cells, CAR macrophages, and CAR-NK cells, show great promise in the treatment of various cancers. Despite the success of CAR-based therapies in treating malignancies, they face numerous challenges, including dysfunction of effector innate and adaptive immune cells, immunosuppressive tumor microenvironment (TME), antigen heterogeneity, and on-target/off-tumor bio-toxicity. The CD47/SIRPα axis is recognized as a critical innate immune checkpoint and is important in regulating myeloid-derived clearance of tumor cells and the innate-adaptive cells' cross-talk in cancer immunity. This signaling axis has risen as a promising target to boost the CAR-based immunotherapies by overcoming phagocytic inhibition and modulating immune evasion. This narrative review explores the integration of CD47/SIRPα modulation as an adjunct to CAR therapies. CD47/SIRPα immune-modulation revealed its potential to boost infiltration, persistence, and phagocytic activity of the immune cells. However, its blockade also poses challenges, including hematologic toxicities, CAR T cell clearance, and compensatory escape pathways. Future work will depend on selective targeting, combinatorial checkpoint modulation, and engineered CAR designs that preserve safety while unlocking durable responses. Herein, we discuss pre-clinical and clinical advancements, safety considerations, and cutting-edge advancements.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145945177","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of CADM1 as an Immunotherapeutic Target and Evaluation of a Novel CADM1-Targeting Antibody-Drug Conjugate in Preclinical Osteosarcoma Models. 临床前骨肉瘤模型中CADM1作为免疫治疗靶点的鉴定和一种新的CADM1靶向抗体-药物偶联物的评估
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-10 DOI: 10.1158/1535-7163.MCT-25-0450
Yifei Wang, Zhongting Zhang, Caterina Longo, Wendong Zhang, Qi Wang, Amer Najjar, Xiangjun Tian, Rossana N Lazcano Segura, Michael E Roth, Jonathan Gill, Douglas J Harrison, Zhaohui Xu, Yanhua Yi, Xin Zhou, Sylvester Jusu, Timothy M Stearns, Steven B Neuhauser, Carol J Bult, Jing Wang, Alexander J Lazar, Richard Gorlick

Due to the paucity of validated cell surface osteosarcoma-specific targets, patients with this condition have long been excluded from the benefits of antibody-drug conjugate (ADC) therapy observed in patients with several solid and hematologic malignancies. Our comprehensive surfaceome profiling approach previously identified osteosarcoma-specific cell-surface antigens that are highly expressed in osteosarcomas but minimally expressed in normal tissues. As a result, one such antigen, CADM1, was selected for the generation of an ADC. We tested a CADM1-targeting ADC with a tesirine payload (SG3249) in vitro in osteosarcoma, rhabdomyosarcoma, and neuroblastoma patient-derived xenograft cell lines. In vivo, we tested six CADM1-expressing osteosarcoma patient-derived xenograft models. The CADM1 ADC demonstrated significant antitumor activity in vitro across the osteosarcoma, rhabdomyosarcoma, and neuroblastoma cell lines. Additionally, it effectively reduced tumor volume and extended event-free survival in all six osteosarcoma PDX models tested. Notably, the CADM1 ADC achieved a major complete response in one model (OS2), complete responses in two models (OS1 and OS33), and partial responses in three models (OS9, OS17, and OS31). Based on these results, clinical development of CADM1-targeted therapies for osteosarcoma and other CADM1-expressing pediatric solid tumors may be warranted.

由于缺乏经过验证的细胞表面骨肉瘤特异性靶点,这种情况的患者长期以来一直被排除在几种实体和血液恶性肿瘤患者中观察到的抗体-药物偶联(ADC)治疗的益处之外。我们的综合表面体分析方法先前鉴定了骨肉瘤特异性细胞表面抗原,这些抗原在骨肉瘤中高度表达,但在正常组织中表达最低。因此,选择了一种这样的抗原CADM1来产生ADC。我们在骨肉瘤、横纹肌肉瘤和神经母细胞瘤患者来源的异种移植细胞系中体外测试了一种带有替西林有效载荷(SG3249)的cadm1靶向ADC。在体内,我们测试了六个表达cadm1的骨肉瘤患者来源的异种移植模型。CADM1 ADC在体外对骨肉瘤、横纹肌肉瘤和神经母细胞瘤细胞系显示出显著的抗肿瘤活性。此外,在所有六种骨肉瘤PDX模型中,它有效地减少了肿瘤体积,延长了无事件生存期。值得注意的是,CADM1 ADC在一个模型(OS2)中实现了主要的完全响应,在两个模型(OS1和OS33)中实现了完全响应,在三个模型(OS9、OS17和OS31)中实现了部分响应。基于这些结果,临床开发针对骨肉瘤和其他表达cadm1的儿童实体瘤的cadm1靶向治疗可能是有必要的。
{"title":"Identification of CADM1 as an Immunotherapeutic Target and Evaluation of a Novel CADM1-Targeting Antibody-Drug Conjugate in Preclinical Osteosarcoma Models.","authors":"Yifei Wang, Zhongting Zhang, Caterina Longo, Wendong Zhang, Qi Wang, Amer Najjar, Xiangjun Tian, Rossana N Lazcano Segura, Michael E Roth, Jonathan Gill, Douglas J Harrison, Zhaohui Xu, Yanhua Yi, Xin Zhou, Sylvester Jusu, Timothy M Stearns, Steven B Neuhauser, Carol J Bult, Jing Wang, Alexander J Lazar, Richard Gorlick","doi":"10.1158/1535-7163.MCT-25-0450","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0450","url":null,"abstract":"<p><p>Due to the paucity of validated cell surface osteosarcoma-specific targets, patients with this condition have long been excluded from the benefits of antibody-drug conjugate (ADC) therapy observed in patients with several solid and hematologic malignancies. Our comprehensive surfaceome profiling approach previously identified osteosarcoma-specific cell-surface antigens that are highly expressed in osteosarcomas but minimally expressed in normal tissues. As a result, one such antigen, CADM1, was selected for the generation of an ADC. We tested a CADM1-targeting ADC with a tesirine payload (SG3249) in vitro in osteosarcoma, rhabdomyosarcoma, and neuroblastoma patient-derived xenograft cell lines. In vivo, we tested six CADM1-expressing osteosarcoma patient-derived xenograft models. The CADM1 ADC demonstrated significant antitumor activity in vitro across the osteosarcoma, rhabdomyosarcoma, and neuroblastoma cell lines. Additionally, it effectively reduced tumor volume and extended event-free survival in all six osteosarcoma PDX models tested. Notably, the CADM1 ADC achieved a major complete response in one model (OS2), complete responses in two models (OS1 and OS33), and partial responses in three models (OS9, OS17, and OS31). Based on these results, clinical development of CADM1-targeted therapies for osteosarcoma and other CADM1-expressing pediatric solid tumors may be warranted.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145945206","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
Resistance to the KRASG12D Inhibitor MRTX1133 is Associated with Increased Sensitivity to BET Inhibition. KRASG12D抑制剂MRTX1133的耐药性与BET抑制敏感性增加相关。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-10 DOI: 10.1158/1535-7163.MCT-25-0483
Daniel R Principe, Jeffrey H Becker, Anastasia E Metropulos, Alejandra M Marinelarena, Thao D Pham, Alexandre F Aissa, Hidayatullah G Munshi

As many as 90% of human pancreatic ductal adenocarcinoma (PDAC) tumors harbor gain-of-function mutations in the KRAS oncogene. Recently, inhibitors of the most common KRAS mutation, KRASG12D, have entered the clinical arena. However, early evidence suggests that as monotherapy, KRASG12D inhibitors such as MRTX1133 at best provide brief periods of disease stabilization. Hence, there is a growing interest in understanding the mechanisms through which tumors acquire resistance to KRAS inhibition. In the present study, we generated in vitro models of MRTX1133 resistance and subjected parental and drug-resistant cell lines to RNA sequencing. This suggested that MRTX1133-resistant tumor cells undergo a global shift toward histone acetylation. Inhibition of the histone acetyltransferase EP300 reversed the drug-resistant phenotype in vitro, which subsequent RNA sequencing experiments determined was associated with the suppression of pro-survival FOSL1 signaling. Accordingly, siFOSL1 reversed the MRTX1133-resistant phenotype with similar effects on pro-survival signaling. Given the lack of clinically useful EP300 or FOSL1 inhibitors, we next explored whether inhibitors of the acetylation scanning BET proteins would be similarly effective. The addition of BET inhibitors re-sensitized several resistant cell lines to MRTX1133 and impaired FOSL1-mediated survival signaling in vitro. In murine models of MRTX1133-resistant PDAC, BET inhibition cooperated with MRTX1133 to markedly extend overall survival. As BET inhibitors are currently under clinical testing, the combination of MRTX1133 and BET inhibitors warrants further investigation, particularly in tumors that have developed resistance to KRAS inhibition.

多达90%的人类胰腺导管腺癌(PDAC)肿瘤含有KRAS癌基因的功能获得性突变。最近,最常见的KRAS突变KRASG12D的抑制剂已进入临床领域。然而,早期证据表明,作为单药治疗,KRASG12D抑制剂如MRTX1133最多只能提供短暂的疾病稳定期。因此,人们对了解肿瘤获得对KRAS抑制的抗性的机制越来越感兴趣。在本研究中,我们建立了MRTX1133体外耐药模型,并对亲代和耐药细胞系进行了RNA测序。这表明mrtx1133耐药的肿瘤细胞经历了向组蛋白乙酰化的整体转变。组蛋白乙酰转移酶EP300的抑制在体外逆转了耐药表型,随后的RNA测序实验确定这与促生存FOSL1信号的抑制有关。因此,siFOSL1逆转了mrtx1133耐药表型,对促生存信号传导具有类似的作用。鉴于缺乏临床上有用的EP300或FOSL1抑制剂,我们下一步探索乙酰化扫描BET蛋白的抑制剂是否同样有效。在体外实验中,添加BET抑制剂可使几种耐药细胞系对MRTX1133重新致敏,并损害fosl1介导的存活信号。在MRTX1133耐药PDAC小鼠模型中,BET抑制与MRTX1133共同作用可显著延长总生存期。由于BET抑制剂目前正处于临床试验阶段,MRTX1133和BET抑制剂的联合应用值得进一步研究,特别是在对KRAS抑制剂产生耐药性的肿瘤中。
{"title":"Resistance to the KRASG12D Inhibitor MRTX1133 is Associated with Increased Sensitivity to BET Inhibition.","authors":"Daniel R Principe, Jeffrey H Becker, Anastasia E Metropulos, Alejandra M Marinelarena, Thao D Pham, Alexandre F Aissa, Hidayatullah G Munshi","doi":"10.1158/1535-7163.MCT-25-0483","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0483","url":null,"abstract":"<p><p>As many as 90% of human pancreatic ductal adenocarcinoma (PDAC) tumors harbor gain-of-function mutations in the KRAS oncogene. Recently, inhibitors of the most common KRAS mutation, KRASG12D, have entered the clinical arena. However, early evidence suggests that as monotherapy, KRASG12D inhibitors such as MRTX1133 at best provide brief periods of disease stabilization. Hence, there is a growing interest in understanding the mechanisms through which tumors acquire resistance to KRAS inhibition. In the present study, we generated in vitro models of MRTX1133 resistance and subjected parental and drug-resistant cell lines to RNA sequencing. This suggested that MRTX1133-resistant tumor cells undergo a global shift toward histone acetylation. Inhibition of the histone acetyltransferase EP300 reversed the drug-resistant phenotype in vitro, which subsequent RNA sequencing experiments determined was associated with the suppression of pro-survival FOSL1 signaling. Accordingly, siFOSL1 reversed the MRTX1133-resistant phenotype with similar effects on pro-survival signaling. Given the lack of clinically useful EP300 or FOSL1 inhibitors, we next explored whether inhibitors of the acetylation scanning BET proteins would be similarly effective. The addition of BET inhibitors re-sensitized several resistant cell lines to MRTX1133 and impaired FOSL1-mediated survival signaling in vitro. In murine models of MRTX1133-resistant PDAC, BET inhibition cooperated with MRTX1133 to markedly extend overall survival. As BET inhibitors are currently under clinical testing, the combination of MRTX1133 and BET inhibitors warrants further investigation, particularly in tumors that have developed resistance to KRAS inhibition.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145959869","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
MDM2 amplification enables selective PROTAC targeting of tumor cells. MDM2扩增可使PROTAC选择性靶向肿瘤细胞。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-10 DOI: 10.1158/1535-7163.MCT-25-0990
Jiandong Chen, Zainab Fatima, Lihong Chen, Mulan Yin, Yunpeng Cui, Jianfeng Cai

PROTACs are bivalent molecules that simultaneously bind to proteins of interest and cellular ubiquitin E3 ligases to promote target degradation. Tumor-specific expression of E3 should increase therapeutic efficacy and reduce toxicity in cancer therapy applications. The E3 ligases currently employed during PROTAC design such as CRBN, VHL, c-IAP and MDM2 are ubiquitously expressed and not considered tumor-specific. However, MDM2 is part of the p53 negative feedback loop and is dynamically regulated at transcriptional and post-translational levels. MDM2 gene amplification occurs at 4-20% frequency in multiple tumor types. To investigate whether MDM2 can serve as tumor-specific PROTAC E3 in certain setting, we analyzed the benchmark compound A1874 (JQ1-Idasanutlin chimera targeting BRD4) under various conditions that affect MDM2 expression and activity. The results showed that A1874 activity is dependent on p53-mediated induction of MDM2 expression and is inactive in cells with mutant p53. Importantly, A1874 showed on average ~12-fold higher potency in tumor cells with MDM2 amplification compared to non-amplified cells, correlating with enhanced cytotoxicity. The results suggest that tumors with MDM2 amplification or overexpression can be selectively targeted using PROTAC approach.

PROTACs是一种二价分子,可同时与目标蛋白和细胞泛素E3连接酶结合以促进靶标降解。肿瘤特异性表达E3在肿瘤治疗应用中应能提高疗效,降低毒性。目前PROTAC设计中使用的E3连接酶如CRBN、VHL、c-IAP和MDM2是普遍表达的,不被认为是肿瘤特异性的。然而,MDM2是p53负反馈回路的一部分,在转录和翻译后水平上受到动态调节。MDM2基因扩增在多种肿瘤类型中发生率为4-20%。为了研究MDM2在特定环境下是否可以作为肿瘤特异性PROTAC E3,我们分析了基准化合物A1874 (JQ1-Idasanutlin嵌合体靶向BRD4)在不同条件下影响MDM2表达和活性的情况。结果表明,A1874的活性依赖于p53介导的MDM2表达的诱导,在p53突变的细胞中不活跃。重要的是,与未扩增的细胞相比,A1874在MDM2扩增的肿瘤细胞中显示出平均约12倍的效力,与增强的细胞毒性相关。结果表明,使用PROTAC方法可以选择性靶向MDM2扩增或过表达的肿瘤。
{"title":"MDM2 amplification enables selective PROTAC targeting of tumor cells.","authors":"Jiandong Chen, Zainab Fatima, Lihong Chen, Mulan Yin, Yunpeng Cui, Jianfeng Cai","doi":"10.1158/1535-7163.MCT-25-0990","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0990","url":null,"abstract":"<p><p>PROTACs are bivalent molecules that simultaneously bind to proteins of interest and cellular ubiquitin E3 ligases to promote target degradation. Tumor-specific expression of E3 should increase therapeutic efficacy and reduce toxicity in cancer therapy applications. The E3 ligases currently employed during PROTAC design such as CRBN, VHL, c-IAP and MDM2 are ubiquitously expressed and not considered tumor-specific. However, MDM2 is part of the p53 negative feedback loop and is dynamically regulated at transcriptional and post-translational levels. MDM2 gene amplification occurs at 4-20% frequency in multiple tumor types. To investigate whether MDM2 can serve as tumor-specific PROTAC E3 in certain setting, we analyzed the benchmark compound A1874 (JQ1-Idasanutlin chimera targeting BRD4) under various conditions that affect MDM2 expression and activity. The results showed that A1874 activity is dependent on p53-mediated induction of MDM2 expression and is inactive in cells with mutant p53. Importantly, A1874 showed on average ~12-fold higher potency in tumor cells with MDM2 amplification compared to non-amplified cells, correlating with enhanced cytotoxicity. The results suggest that tumors with MDM2 amplification or overexpression can be selectively targeted using PROTAC approach.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145945192","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 of antibody-drug conjugates targeting L1CAM to treat metastatic cancer. 靶向L1CAM的抗体-药物偶联物治疗转移性癌症的研究进展。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-09 DOI: 10.1158/1535-7163.MCT-25-1184
Jin Suk Park, Carson Kenum, Lan He, Abdul G Khan, Mary Ann Pohl, Thomas E White, Sreekumar R Kodangattil, Charles M Rudin, Paul J Balderes, Ivo C Lorenz, Joan Massagué, Karuna Ganesh

Effective treatment for metastatic cancer has remained elusive due to the persistence of drug-resistant metastasis stem cells (MetSCs) that drive relapse. MetSCs are tumor cell subpopulations enriched for their ability to reinitiate and sustain metastatic growth, displaying phenotypic plasticity and resistance to chemotherapy. These cells express the L1 cell adhesion molecule (L1CAM), a transmembrane protein detected in numerous human solid tumor types and at multiple disseminated organ sites. As a selective surface marker of MetSCs, L1CAM is a promising candidate for molecularly targeted drugs aimed at eliminating metastases, yet strategies to date have not achieved clinical success. Here, we develop antibody-drug conjugates (ADCs) to deliver highly toxic PNU-159682 payloads to L1CAM-expressing cells. We report the generation of monoclonal antibodies (mAb) with high binding affinity, specificity and selectivity for the human L1CAM extracellular domain. Optimized L1CAM-targeting mAbs were conjugated to PNU-159682 to generate ADC variants with both cleavable and non-cleavable linkers, with an average drug-antibody-ratio (DAR) of four. ADCs derived from three antibodies targeting various epitopes of the L1CAM extracellular portion potently killed cells exhibiting varying levels of surface L1CAM expression. L1CAM ADCs given as monotherapy resulted in robust tumor control and extended survival in mice harboring subcutaneous L1CAM+ xenografts or L1CAM+ lung metastases from triple-negative basal breast cancer and lung adenocarcinoma. Safety analyses with mouse cross-reactive antibodies indicate a feasible therapeutic window. Our findings offer strong proof-of-concept to support the preclinical development of these novel L1CAM ADCs as therapeutic agents for advanced solid tumors.

由于耐药转移干细胞(MetSCs)的持续存在导致复发,转移性癌症的有效治疗仍然难以捉摸。MetSCs是肿瘤细胞亚群,具有重新启动和维持转移性生长的能力,表现出表型可塑性和对化疗的抗性。这些细胞表达L1细胞粘附分子(L1CAM),这是一种在许多人类实体肿瘤类型和多个播散性器官部位检测到的跨膜蛋白。作为MetSCs的选择性表面标记物,L1CAM是一种很有前途的分子靶向药物,旨在消除转移,但迄今为止尚未取得临床成功。在这里,我们开发了抗体-药物偶联物(adc),将高毒性PNU-159682有效载荷传递到表达l1cam的细胞。我们报道了对人L1CAM细胞外结构域具有高结合亲和力、特异性和选择性的单克隆抗体(mAb)的产生。将优化后的l1cam靶向单克隆抗体与PNU-159682偶联,生成具有可切割和不可切割连接体的ADC变体,平均药抗体比(DAR)为4。adc来源于三种靶向L1CAM细胞外部分不同表位的抗体,它们能杀死表现出不同水平表面L1CAM表达的细胞。在皮下移植L1CAM+异种移植或L1CAM+肺转移的三阴性基底乳腺癌和肺腺癌小鼠中,单药给予L1CAM adc可有效控制肿瘤,延长生存期。小鼠交叉反应性抗体的安全性分析显示了一个可行的治疗窗口。我们的研究结果为支持这些新型L1CAM adc作为晚期实体肿瘤治疗剂的临床前开发提供了强有力的概念证明。
{"title":"Development of antibody-drug conjugates targeting L1CAM to treat metastatic cancer.","authors":"Jin Suk Park, Carson Kenum, Lan He, Abdul G Khan, Mary Ann Pohl, Thomas E White, Sreekumar R Kodangattil, Charles M Rudin, Paul J Balderes, Ivo C Lorenz, Joan Massagué, Karuna Ganesh","doi":"10.1158/1535-7163.MCT-25-1184","DOIUrl":"10.1158/1535-7163.MCT-25-1184","url":null,"abstract":"<p><p>Effective treatment for metastatic cancer has remained elusive due to the persistence of drug-resistant metastasis stem cells (MetSCs) that drive relapse. MetSCs are tumor cell subpopulations enriched for their ability to reinitiate and sustain metastatic growth, displaying phenotypic plasticity and resistance to chemotherapy. These cells express the L1 cell adhesion molecule (L1CAM), a transmembrane protein detected in numerous human solid tumor types and at multiple disseminated organ sites. As a selective surface marker of MetSCs, L1CAM is a promising candidate for molecularly targeted drugs aimed at eliminating metastases, yet strategies to date have not achieved clinical success. Here, we develop antibody-drug conjugates (ADCs) to deliver highly toxic PNU-159682 payloads to L1CAM-expressing cells. We report the generation of monoclonal antibodies (mAb) with high binding affinity, specificity and selectivity for the human L1CAM extracellular domain. Optimized L1CAM-targeting mAbs were conjugated to PNU-159682 to generate ADC variants with both cleavable and non-cleavable linkers, with an average drug-antibody-ratio (DAR) of four. ADCs derived from three antibodies targeting various epitopes of the L1CAM extracellular portion potently killed cells exhibiting varying levels of surface L1CAM expression. L1CAM ADCs given as monotherapy resulted in robust tumor control and extended survival in mice harboring subcutaneous L1CAM+ xenografts or L1CAM+ lung metastases from triple-negative basal breast cancer and lung adenocarcinoma. Safety analyses with mouse cross-reactive antibodies indicate a feasible therapeutic window. Our findings offer strong proof-of-concept to support the preclinical development of these novel L1CAM ADCs as therapeutic agents for advanced solid tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145934233","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
The Proteasome Is Revealed as a Therapeutic Target in Recurrent Glioblastoma Xenografts. 蛋白酶体是复发性胶质母细胞瘤异种移植的治疗靶点。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-08 DOI: 10.1158/1535-7163.MCT-25-0770
Charlotte Degorre, Philip J Tofilon

Radiation remains a primary treatment for glioblastoma (GBM), yet tumors frequently recur within 2 years. In this study, orthotopic xenografts initiated from glioma stem-like cells (GSC) implanted into the right striatum of nude mice were used to investigate the biology of recurrent GBM. In this model, untreated tumors showed diffuse growth pattern across the right hemisphere and olfactory bulb (OB), whereas postirradiation tumors (10 Gy) regrew predominantly within the OB, exhibiting increased cell density and a well-demarcated border indicative of an altered growth pattern. Transcriptomes of untreated and recurrent tumors were assessed using spatial profiling. Comparison of gene expression across regions of interest revealed that recurrent tumors are less heterogeneous and exhibit a distinct transcriptional profile compared with untreated tumors. A total of 463 genes were differentially expressed, and gene set enrichment analysis revealed significant enrichment of pathways related to cell-cycle regulation in the recurrent as compared with untreated tumors. Further analysis of those pathways revealed a significant upregulation of 22 proteasome-related genes in recurrent tumors. Moreover, functional assays revealed significantly higher proteasome activity in recurrent compared with untreated tumors, suggesting the proteasome as a potential therapeutic target unique to recurrent GBM. To evaluate the therapeutic relevance, mice were treated with the combination of radiation followed by the proteasome inhibitor ixazomib. Whereas ixazomib had no effect on untreated tumors, its administration after irradiation significantly prolonged survival in two GSC xenograft models. These results illustrate how defining molecular alterations that develop in recurrent GBM xenografts can lead to the identification of a novel therapeutic target.

放疗仍然是胶质母细胞瘤(GBM)的主要治疗方法,但肿瘤经常在2年内复发。本研究将胶质瘤干细胞样细胞(GSC)原位移植至裸鼠右侧纹状体,研究复发性GBM的生物学特性。在该模型中,未经治疗的肿瘤显示右半球和嗅球(OB)的弥漫性生长模式,而放射后肿瘤(10 Gy)主要在嗅球内再生,细胞密度增加,边界清晰,表明生长模式发生了改变。使用空间谱法评估未治疗和复发肿瘤的转录组。跨感兴趣区域的基因表达比较显示,与未治疗的肿瘤相比,复发肿瘤的异质性较小,表现出不同的转录谱。共有463个基因差异表达,基因集富集分析显示,与未治疗的肿瘤相比,复发肿瘤中与细胞周期调控相关的途径显著富集。对这些通路的进一步分析显示,22个蛋白酶体相关基因在复发性肿瘤中显著上调。此外,功能分析显示,与未治疗的肿瘤相比,复发性肿瘤的蛋白酶体活性明显更高,这表明蛋白酶体是复发性GBM特有的潜在治疗靶点。为了评估治疗相关性,小鼠接受了放疗和蛋白酶体抑制剂ixazomib的联合治疗。虽然ixazomib对未治疗的肿瘤没有影响,但在两种GSC异种移植模型中,照射后给予ixazomib可显著延长生存期。这些结果说明了如何定义复发性GBM异种移植物中发生的分子改变可以导致鉴定新的治疗靶点。
{"title":"The Proteasome Is Revealed as a Therapeutic Target in Recurrent Glioblastoma Xenografts.","authors":"Charlotte Degorre, Philip J Tofilon","doi":"10.1158/1535-7163.MCT-25-0770","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0770","url":null,"abstract":"<p><p>Radiation remains a primary treatment for glioblastoma (GBM), yet tumors frequently recur within 2 years. In this study, orthotopic xenografts initiated from glioma stem-like cells (GSC) implanted into the right striatum of nude mice were used to investigate the biology of recurrent GBM. In this model, untreated tumors showed diffuse growth pattern across the right hemisphere and olfactory bulb (OB), whereas postirradiation tumors (10 Gy) regrew predominantly within the OB, exhibiting increased cell density and a well-demarcated border indicative of an altered growth pattern. Transcriptomes of untreated and recurrent tumors were assessed using spatial profiling. Comparison of gene expression across regions of interest revealed that recurrent tumors are less heterogeneous and exhibit a distinct transcriptional profile compared with untreated tumors. A total of 463 genes were differentially expressed, and gene set enrichment analysis revealed significant enrichment of pathways related to cell-cycle regulation in the recurrent as compared with untreated tumors. Further analysis of those pathways revealed a significant upregulation of 22 proteasome-related genes in recurrent tumors. Moreover, functional assays revealed significantly higher proteasome activity in recurrent compared with untreated tumors, suggesting the proteasome as a potential therapeutic target unique to recurrent GBM. To evaluate the therapeutic relevance, mice were treated with the combination of radiation followed by the proteasome inhibitor ixazomib. Whereas ixazomib had no effect on untreated tumors, its administration after irradiation significantly prolonged survival in two GSC xenograft models. These results illustrate how defining molecular alterations that develop in recurrent GBM xenografts can lead to the identification of a novel therapeutic target.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF9"},"PeriodicalIF":5.5,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917944","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
TPP-45142-an Anti-HER2 T cell Engager-Designed for Selective HER2-Low Cancer Immunotherapy. tpp -45142-一种抗her2 T细胞接合物,设计用于选择性低her2癌症免疫治疗
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-08 DOI: 10.1158/1535-7163.MCT-25-0654
Evelyn De Tavernier, Peter S Kim, Eduardo M Bruch, Virna F Cortez-Retamozo, Lien Timmerman, Alyssa L Flynn, Wouter Van Overbeke, Fabrice Tirode, Valeria Cintra Barbosa-Lorenzi, Peter Piepenhagen, Thuvan Dinh-Le, Ernesto Luna, Aiqun Li, Ann Baker, Alexey Rak, Lily I Pao, Ana Paula B Vintém

The standard of care for patients with HER2-positive cancers is well established, but a significant unmet need exists for patients with HER2-low tumors, who do not meet the eligibility criteria for trastuzumab, and for patients with HER2-positive tumors, who are refractory to trastuzumab treatment. Therefore, in this study, we developed a NANOBODY® domain-based HER2-targeting, T cell receptor (TCR)αβ-based T cell engager (TCE) molecule-TPP-45142; it recognizes a HER2 epitope distinct from that recognized by trastuzumab and pertuzumab and redirects T cells to kill HER2-low cancers such as breast, gastric, and gastroesophageal junction adenocarcinoma (GEJ) cancers. TPP-45142 mediated potent T cell-dependent cytotoxicity against HER2-low cancer cell lines in vitro and inhibited in vivo tumor growth of HER2-low breast cancer xenografts. TPP-45142 was highly selective toward tumor cells expressing low HER2 levels than toward normal cardiac cells and exhibited a favourable therapeutic index as per a cytokine release assay. Thus, TPP-45142, with an improved safety profile, is a promising next-generation TCE for treating challenging HER2-low cancers.

her2阳性癌症患者的护理标准已经建立,但对于her2低肿瘤患者(不符合曲妥珠单抗的资格标准)和her2阳性肿瘤患者(曲妥珠单抗治疗难治性)存在显著的未满足需求。因此,在本研究中,我们开发了一种基于NANOBODY®结构域靶向her2、基于T细胞受体(TCR)αβ的T细胞接合物(TCE)分子- tpp -45142;它识别不同于曲妥珠单抗和帕妥珠单抗识别的HER2表位,并重新引导T细胞杀死HER2低水平的癌症,如乳腺癌、胃癌和胃食管交界处腺癌(GEJ)癌症。TPP-45142在体外介导对her2 -低水平癌细胞的强效T细胞依赖性细胞毒性,并抑制her2 -低水平乳腺癌异种移植瘤的体内肿瘤生长。与正常心肌细胞相比,TPP-45142对HER2表达水平较低的肿瘤细胞具有高度选择性,并且根据细胞因子释放试验显示,TPP-45142具有良好的治疗指数。因此,TPP-45142具有更高的安全性,是治疗具有挑战性的her2低癌的有前景的下一代TCE。
{"title":"TPP-45142-an Anti-HER2 T cell Engager-Designed for Selective HER2-Low Cancer Immunotherapy.","authors":"Evelyn De Tavernier, Peter S Kim, Eduardo M Bruch, Virna F Cortez-Retamozo, Lien Timmerman, Alyssa L Flynn, Wouter Van Overbeke, Fabrice Tirode, Valeria Cintra Barbosa-Lorenzi, Peter Piepenhagen, Thuvan Dinh-Le, Ernesto Luna, Aiqun Li, Ann Baker, Alexey Rak, Lily I Pao, Ana Paula B Vintém","doi":"10.1158/1535-7163.MCT-25-0654","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0654","url":null,"abstract":"<p><p>The standard of care for patients with HER2-positive cancers is well established, but a significant unmet need exists for patients with HER2-low tumors, who do not meet the eligibility criteria for trastuzumab, and for patients with HER2-positive tumors, who are refractory to trastuzumab treatment. Therefore, in this study, we developed a NANOBODY® domain-based HER2-targeting, T cell receptor (TCR)αβ-based T cell engager (TCE) molecule-TPP-45142; it recognizes a HER2 epitope distinct from that recognized by trastuzumab and pertuzumab and redirects T cells to kill HER2-low cancers such as breast, gastric, and gastroesophageal junction adenocarcinoma (GEJ) cancers. TPP-45142 mediated potent T cell-dependent cytotoxicity against HER2-low cancer cell lines in vitro and inhibited in vivo tumor growth of HER2-low breast cancer xenografts. TPP-45142 was highly selective toward tumor cells expressing low HER2 levels than toward normal cardiac cells and exhibited a favourable therapeutic index as per a cytokine release assay. Thus, TPP-45142, with an improved safety profile, is a promising next-generation TCE for treating challenging HER2-low cancers.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145934213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of Small-Molecule Inhibitors that Block the GTP-Binding Pocket of K-Ras and Other Members of the Ras Superfamily of Small GTPases. 阻断K-Ras和其他Ras小gtpase超家族成员gtp结合口袋的小分子抑制剂的鉴定。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-24-0618
Luca Carta, Rebecca Hutcheson, Carolina L Bigarella, Sufang Zhang, Simon A Davis, Michael J Rudolph, Charles H Reynolds, Matthias Quick, Theresa M Williams, Michael Schmertzler, Yaron R Hadari

RAS genes encode small GTPases essential for mammalian cell proliferation, differentiation, and survival. RAS gene mutations are associated with 20% to 30% of all human cancers. Based on earlier reports of extremely high Ras binding affinities for GTP, Ras proteins were previously considered undruggable. Using three independent techniques, we report binding affinities of K-Ras and several K-Ras mutants for GTP in the 250 to 400 nmol/L range, orders of magnitude lower than previously reported (∼10 pmol/L). This discovery suggests that K-Ras and other small-GTPase proteins may indeed be druggable targets. We identified more than 400 small molecules that compete non-covalently with GTP binding to K-Ras. Focusing on two inhibitors, we demonstrate the inhibition of K-Ras in downstream signaling and cellular proliferation in human pancreatic and non-small cell lung cancer cells expressing wild-type or mutant K-Ras. These two compounds represent novel pan-Ras superfamily inhibitors as they also inhibited GTP binding to other members such as RAB5A and RAB35.

RAS基因编码对哺乳动物细胞增殖、分化和存活至关重要的小gtpase。RAS基因突变与20%到30%的人类癌症有关。基于先前关于Ras与GTP结合亲和力极高的报道,Ras蛋白以前被认为是不可药物的。使用三种独立的技术,我们报告了K-Ras和几个K-Ras突变体对GTP的结合亲和力在250至400 nmol/L范围内,比以前报道的(~ 10 pmol/L)低几个数量级。这一发现表明,K-Ras和其他小gtpase蛋白可能确实是可药物治疗的靶标。我们发现了400多个与GTP结合K-Ras的非共价竞争的小分子。以两种抑制剂为重点,我们在表达野生型或突变型K-Ras的人胰腺癌和非小细胞肺癌细胞中证明了K-Ras在下游信号传导和细胞增殖中的抑制作用。这两种化合物代表了新的泛ras超家族抑制剂,因为它们也抑制GTP与其他成员(如RAB5A和RAB35)的结合。
{"title":"Identification of Small-Molecule Inhibitors that Block the GTP-Binding Pocket of K-Ras and Other Members of the Ras Superfamily of Small GTPases.","authors":"Luca Carta, Rebecca Hutcheson, Carolina L Bigarella, Sufang Zhang, Simon A Davis, Michael J Rudolph, Charles H Reynolds, Matthias Quick, Theresa M Williams, Michael Schmertzler, Yaron R Hadari","doi":"10.1158/1535-7163.MCT-24-0618","DOIUrl":"10.1158/1535-7163.MCT-24-0618","url":null,"abstract":"<p><p>RAS genes encode small GTPases essential for mammalian cell proliferation, differentiation, and survival. RAS gene mutations are associated with 20% to 30% of all human cancers. Based on earlier reports of extremely high Ras binding affinities for GTP, Ras proteins were previously considered undruggable. Using three independent techniques, we report binding affinities of K-Ras and several K-Ras mutants for GTP in the 250 to 400 nmol/L range, orders of magnitude lower than previously reported (∼10 pmol/L). This discovery suggests that K-Ras and other small-GTPase proteins may indeed be druggable targets. We identified more than 400 small molecules that compete non-covalently with GTP binding to K-Ras. Focusing on two inhibitors, we demonstrate the inhibition of K-Ras in downstream signaling and cellular proliferation in human pancreatic and non-small cell lung cancer cells expressing wild-type or mutant K-Ras. These two compounds represent novel pan-Ras superfamily inhibitors as they also inhibited GTP binding to other members such as RAB5A and RAB35.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"84-93"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12757722/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145040969","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
Proteolysis targeting chimeric-based technology in myeloma and lymphoma. 靶向嵌合蛋白水解技术在骨髓瘤和淋巴瘤中的应用。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-25-1148
Adrian Bogdan Tigu, Andrei Ivancuta, Ciprian Tomuleasa, Madalina Nistor, David Kegyes, Diana Cenariu, Raluca Munteanu, Anca Dana Buzoianu, Hermann Einsele, Massimo Federico, Sebastian Kobold, Diana Gulei, Aaron Ciechanover

Proteolysis-targeting chimeras (PROTACs) leverage the ubiquitin-proteasome system to selectively degrade oncogenic proteins, including such previously seen as undruggable. Recent preclinical studies indicate that PROTACs may come as novel therapeutic strategy in lymphoma and myeloma. Indeed, preclinically, PROTACs have high efficacy and remarkable selectivity, favorable safety profile and lower toxicity compared to conventional therapies. Their catalytic, reusable mechanism enables drug dosing and offers the perspective of a long-term low dose treatment. PROTACs demonstrated their ability to overcome drug resistance by targeting and degrading overexpressed or mutant proteins, which are responsible for refractory disease. This review aims to offer a comprehensive evaluation of the current existing PROTACs that have been tested in Lymphoma and Myeloma, to highlight the need for drug optimization and further translational research that could translate PROTACs to clinical trials.

靶向蛋白水解嵌合体(PROTACs)利用泛素-蛋白酶体系统选择性地降解致癌蛋白,包括以前认为不可药物的蛋白。最近的临床前研究表明,PROTACs可能成为淋巴瘤和骨髓瘤的新治疗策略。事实上,在临床前,与传统疗法相比,PROTACs具有高疗效和显著的选择性,良好的安全性和较低的毒性。他们的催化,可重复使用的机制,使药物剂量,并提供长期低剂量治疗的前景。PROTACs通过靶向和降解导致难治性疾病的过表达或突变蛋白,证明了它们克服耐药性的能力。本综述旨在对目前已在淋巴瘤和骨髓瘤中进行测试的PROTACs进行全面评估,以强调药物优化和进一步的转化研究的必要性,从而将PROTACs转化为临床试验。
{"title":"Proteolysis targeting chimeric-based technology in myeloma and lymphoma.","authors":"Adrian Bogdan Tigu, Andrei Ivancuta, Ciprian Tomuleasa, Madalina Nistor, David Kegyes, Diana Cenariu, Raluca Munteanu, Anca Dana Buzoianu, Hermann Einsele, Massimo Federico, Sebastian Kobold, Diana Gulei, Aaron Ciechanover","doi":"10.1158/1535-7163.MCT-25-1148","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-1148","url":null,"abstract":"<p><p>Proteolysis-targeting chimeras (PROTACs) leverage the ubiquitin-proteasome system to selectively degrade oncogenic proteins, including such previously seen as undruggable. Recent preclinical studies indicate that PROTACs may come as novel therapeutic strategy in lymphoma and myeloma. Indeed, preclinically, PROTACs have high efficacy and remarkable selectivity, favorable safety profile and lower toxicity compared to conventional therapies. Their catalytic, reusable mechanism enables drug dosing and offers the perspective of a long-term low dose treatment. PROTACs demonstrated their ability to overcome drug resistance by targeting and degrading overexpressed or mutant proteins, which are responsible for refractory disease. This review aims to offer a comprehensive evaluation of the current existing PROTACs that have been tested in Lymphoma and Myeloma, to highlight the need for drug optimization and further translational research that could translate PROTACs to clinical trials.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145889557","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
DS-3939a: A TA-MUC1-Directed Antibody-Drug Conjugate with Broad Antitumor Activity. DS-3939a:一种具有广泛抗肿瘤活性的ta - muc1定向抗体-药物偶联物
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-24-0666
Kohei Takano, Mayuko Yukiura, Kazuki Takahashi, Michiko Kitamura, Hiroko Okuno, Yoshinobu Shiose, Kokichi Honda, Kazunori Oyama, Makiko Yamada, Wataru Obuchi, Kazuyoshi Kumagai, Ken Sakurai, Riki Goto, Akiko Zembutsu, Takashi Kagari, Yuki Abe, Toshinori Agatsuma

Tumor-associated mucin-1 (TA-MUC1) is a glycoform of the MUC1 protein that is aberrantly glycosylated and is primarily observed in cancer cells. TA-MUC1 is highly expressed in various human epithelial cancers, making it an attractive target for cancer therapies. In this study, we describe the development of DS-3939a, a novel TA-MUC1-targeting antibody-drug conjugate that utilizes the potent DNA topoisomerase I inhibitor DXd, and evaluation of its pharmacologic activity in preclinical in vitro and in vivo models. IHC of clinical tumor tissue microarrays of various cancer types exhibited positive staining for TA-MUC1 in a number of samples, with a particularly high positive rate in bladder, lung, and breast cancers. In vitro profiling of DS-3939a confirmed that it could specifically bind to TA-MUC1 and inhibit the growth of TA-MUC1-positive cancer cells by inducing DNA damage and apoptosis. DS-3939a also exhibited significant antitumor effects in multiple TA-MUC1-positive cell line-derived and patient-derived xenograft models. Moreover, DS-3939a elicited strong tumor regression in several xenograft models even following treatment with other cytotoxic antibody-drug conjugates, likely through its efficient payload delivery. Overall, these data provide evidence for the potential utility of DS-3939a for the treatment of TA-MUC1-expressing tumors and support the rationale for the ongoing phase I/II clinical study (NCT05875168).

肿瘤相关粘蛋白-1 (TA-MUC1)是MUC1蛋白的一种糖型,其糖基化异常,主要在癌细胞中观察到。TA-MUC1在各种人类上皮癌中高度表达,使其成为癌症治疗的一个有吸引力的靶点。在这里,我们描述了DS-3939a的开发,这是一种新的靶向ta - muc1的抗体-药物偶联物(ADC),利用有效的DNA拓扑异构酶I抑制剂DXd,并在临床前体外和体内模型中评估其药理活性。不同癌症类型的临床肿瘤组织微阵列免疫组化显示,TA-MUC1在许多样本中呈阳性染色,在膀胱癌、肺癌和乳腺癌中阳性率特别高。DS-3939a的体外分析证实,它可以特异性结合TA-MUC1,并通过诱导DNA损伤和凋亡来抑制TA-MUC1阳性癌细胞的生长。DS-3939a在多种ta - muc1阳性细胞系来源和患者来源的异种移植物模型中也表现出显著的抗肿瘤作用。此外,DS-3939a在几种异种移植模型中甚至在使用其他细胞毒性adc治疗后也引起了强烈的肿瘤消退,可能是通过其有效的有效载荷递送。总的来说,这些数据为DS-3939a治疗表达ta - muc1肿瘤的潜在效用提供了证据,并支持了正在进行的I/II期临床研究的基本原理(NCT05875168)。
{"title":"DS-3939a: A TA-MUC1-Directed Antibody-Drug Conjugate with Broad Antitumor Activity.","authors":"Kohei Takano, Mayuko Yukiura, Kazuki Takahashi, Michiko Kitamura, Hiroko Okuno, Yoshinobu Shiose, Kokichi Honda, Kazunori Oyama, Makiko Yamada, Wataru Obuchi, Kazuyoshi Kumagai, Ken Sakurai, Riki Goto, Akiko Zembutsu, Takashi Kagari, Yuki Abe, Toshinori Agatsuma","doi":"10.1158/1535-7163.MCT-24-0666","DOIUrl":"10.1158/1535-7163.MCT-24-0666","url":null,"abstract":"<p><p>Tumor-associated mucin-1 (TA-MUC1) is a glycoform of the MUC1 protein that is aberrantly glycosylated and is primarily observed in cancer cells. TA-MUC1 is highly expressed in various human epithelial cancers, making it an attractive target for cancer therapies. In this study, we describe the development of DS-3939a, a novel TA-MUC1-targeting antibody-drug conjugate that utilizes the potent DNA topoisomerase I inhibitor DXd, and evaluation of its pharmacologic activity in preclinical in vitro and in vivo models. IHC of clinical tumor tissue microarrays of various cancer types exhibited positive staining for TA-MUC1 in a number of samples, with a particularly high positive rate in bladder, lung, and breast cancers. In vitro profiling of DS-3939a confirmed that it could specifically bind to TA-MUC1 and inhibit the growth of TA-MUC1-positive cancer cells by inducing DNA damage and apoptosis. DS-3939a also exhibited significant antitumor effects in multiple TA-MUC1-positive cell line-derived and patient-derived xenograft models. Moreover, DS-3939a elicited strong tumor regression in several xenograft models even following treatment with other cytotoxic antibody-drug conjugates, likely through its efficient payload delivery. Overall, these data provide evidence for the potential utility of DS-3939a for the treatment of TA-MUC1-expressing tumors and support the rationale for the ongoing phase I/II clinical study (NCT05875168).</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"7-20"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12757726/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144601021","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:604180095
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