Molecular Cancer Therapeutics最新文献

筛选
英文 中文
ASCL1 restrains ERK1/2 to promote survival of a subset of neuroendocrine lung cancers. ASCL1抑制ERK1/2,促进神经内分泌肺癌亚群的存活。
IF 5.7 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-19 DOI: 10.1158/1535-7163.mct-24-0355
Ana Martin-Vega,Svetlana A Earnest,Alexander Augustyn,Chonlarat Wichaidit,Luc Girard,Michael Peyton,John D Minna,Jane E Johnson,Melanie H Cobb
{"title":"ASCL1 restrains ERK1/2 to promote survival of a subset of neuroendocrine lung cancers.","authors":"Ana Martin-Vega,Svetlana A Earnest,Alexander Augustyn,Chonlarat Wichaidit,Luc Girard,Michael Peyton,John D Minna,Jane E Johnson,Melanie H Cobb","doi":"10.1158/1535-7163.mct-24-0355","DOIUrl":"https://doi.org/10.1158/1535-7163.mct-24-0355","url":null,"abstract":"The transcription factor achaete-scute complex homolog 1 (ASCL1) is a lineage oncogene that is central in growth and survival of the majority of small cell lung cancers (SCLC) and neuroendocrine non-small cell lung cancers (NSCLC-NE) that express it. Targeting ASCL1, or its downstream pathways, remains a challenge. SCLCs and NSCLC-NE that express ASCL1 exhibit relatively low ERK1/2 activity, in dramatic contrast to NSCLCs in which the ERK pathway has a major role in pathogenesis. ERK1/2 inhibition in ASCL1-expressing lung tumor cells revealed down-regulation of ERK1/2 pathway suppressors SPRY4, SPRED1, DUSP6, and the transcription factor ETV5, which regulates DUSP6. CHIP-seq demonstrated that these genes are bound by ASCL1. Availability of a pharmacological inhibitor directed mechanistic studies towards DUSP6, an ERK1/2-selective phosphatase, in a subset of ASCL1-high NE lung tumors. Inhibition of DUSP6 increased active ERK1/2, which accumulated in the nucleus. Pharmacologic and genetic inhibition of DUSP6 reduced proliferation and survival of these cancers. Resistance developed in DUSP6 KO cells, indicating a bypass mechanism. Although targeting ASCL1 remains a challenge, our findings suggest that expression of ASCL1, DUSP6 and low phospho-ERK1/2 identify neuroendocrine lung cancers for which DUSP6 may be a therapeutic target.","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268794","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
Emerging Tumor-Agnostic Molecular Targets. 新出现的肿瘤诊断分子靶标。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-16 DOI: 10.1158/1535-7163.MCT-23-0725
Dedipya Bhamidipati, Alison M Schram
{"title":"Emerging Tumor-Agnostic Molecular Targets.","authors":"Dedipya Bhamidipati, Alison M Schram","doi":"10.1158/1535-7163.MCT-23-0725","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0725","url":null,"abstract":"<p><p>Advances in tumor molecular profiling have uncovered shared genomic and proteomic alterations across tumor types that can be exploited therapeutically. A biomarker-driven, disease-agnostic approach to oncology drug development can maximize the reach of novel therapeutics. To date, eight drug-biomarker pairs have been approved for the treatment of patients with advanced solid tumors with specific molecular profiles. Emerging biomarkers with the potential for clinical actionability across tumor types include gene fusions involving NRG1, FGFR1/2/3, BRAF, and ALK, mutations in TP53 Y220C, KRAS G12C, FGFR2/3, and BRAF non-V600 (Class II). We explore the growing evidence for clinical actionability of these biomarkers in patients with advanced solid tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291433","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery and Characterization of Brigimadlin, a Novel and Highly Potent MDM2–p53 Antagonist Suitable for Intermittent Dose Schedules 发现并鉴定适合间歇性给药的新型强效 MDM2-p53 拮抗剂 Brigimadlin
IF 5.7 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-11 DOI: 10.1158/1535-7163.mct-23-0783
Andreas Gollner, Dorothea Rudolph, Ulrike Weyer-Czernilofsky, Rosa Baumgartinger, Peter Jung, Harald Weinstabl, Jürgen Ramharter, Rolf Grempler, Jens Quant, Jörg Rinnenthal, Alejandro Pérez Pitarch, Bojana Golubovic, Daniel Gerlach, Gerd Bader, Kristiane Wetzel, Sebastian Otto, Christian Mandl, Guido Boehmelt, Darryl B. McConnell, Norbert Kraut, Patrizia Sini
{"title":"Discovery and Characterization of Brigimadlin, a Novel and Highly Potent MDM2–p53 Antagonist Suitable for Intermittent Dose Schedules","authors":"Andreas Gollner, Dorothea Rudolph, Ulrike Weyer-Czernilofsky, Rosa Baumgartinger, Peter Jung, Harald Weinstabl, Jürgen Ramharter, Rolf Grempler, Jens Quant, Jörg Rinnenthal, Alejandro Pérez Pitarch, Bojana Golubovic, Daniel Gerlach, Gerd Bader, Kristiane Wetzel, Sebastian Otto, Christian Mandl, Guido Boehmelt, Darryl B. McConnell, Norbert Kraut, Patrizia Sini","doi":"10.1158/1535-7163.mct-23-0783","DOIUrl":"https://doi.org/10.1158/1535-7163.mct-23-0783","url":null,"abstract":"p53 is known as the guardian of the genome and is one of the most important tumor-suppressors. It is inactivated in most tumors, either via tumor protein p53 (TP53) gene mutation or copy number amplification of key negative regulators, e.g., mouse double minute 2 (MDM2). Compounds that bind to the MDM2 protein and disrupt its interaction with p53 restore p53 tumor suppressor activity, thereby promoting cell cycle arrest and apoptosis. Previous clinical experience with MDM2–p53 protein–protein interaction antagonists (MDM2–p53 antagonists) have demonstrated that thrombocytopenia and neutropenia represent on-target dose-limiting toxicities that might restrict their therapeutic utility. Dosing less frequently, while maintaining efficacious exposure, represents an approach to mitigate toxicity and improve the therapeutic window of MDM2–p53 antagonists. However, to achieve this, a molecule possessing excellent potency and ideal pharmacokinetic properties is required. Here, we present the discovery and characterization of brigimadlin (BI 907828), a novel, investigational spiro-oxindole MDM2–p53 antagonist. Brigimadlin exhibited high bioavailability and exposure, as well as dose-linear pharmacokinetics in preclinical models. Brigimadlin treatment restored p53 activity and led to apoptosis induction in preclinical models of TP53 wild-type, MDM2-amplified cancer. Oral administration of brigimadlin in an intermittent dosing schedule induced potent tumor growth inhibition in several TP53 wild-type, MDM2-amplified xenograft models. Exploratory clinical pharmacokinetic studies (NCT03449381) showed high systemic exposure and a long plasma elimination half-life in cancer patients who received oral brigimadlin. These findings support the continued clinical evaluation of brigimadlin in patients with MDM2-amplified cancers, such as dedifferentiated liposarcoma.","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142194516","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting the synthetic lethal relationship between FOCAD and TUT7 represents a potential therapeutic opportunity for TUT4/7 small molecule inhibitors in cancer. 针对 FOCAD 和 TUT7 之间的合成致死关系,是 TUT4/7 小分子抑制剂治疗癌症的潜在机会。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-05 DOI: 10.1158/1535-7163.MCT-24-0176
Robinson Triboulet, Khikmet Sadykov, Darren M Harvey, David M Wilson, Michael J Steinbaugh, Christopher B Mayo, Dillon Hawley, Andrew Madanjian, Corey Fyfe, Christina Bracken, Izarys Rivera-Rivera, Anna Ericsson, Andrew R Snyder, Sarah K Knutson, Ross L Stein, Veronica Gibaja, Shomir Ghosh, Robert M Campbell
{"title":"Targeting the synthetic lethal relationship between FOCAD and TUT7 represents a potential therapeutic opportunity for TUT4/7 small molecule inhibitors in cancer.","authors":"Robinson Triboulet, Khikmet Sadykov, Darren M Harvey, David M Wilson, Michael J Steinbaugh, Christopher B Mayo, Dillon Hawley, Andrew Madanjian, Corey Fyfe, Christina Bracken, Izarys Rivera-Rivera, Anna Ericsson, Andrew R Snyder, Sarah K Knutson, Ross L Stein, Veronica Gibaja, Shomir Ghosh, Robert M Campbell","doi":"10.1158/1535-7163.MCT-24-0176","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0176","url":null,"abstract":"<p><p>Targeting synthetic lethal interactions between genes has emerged as a promising strategy for cancer therapy. This study explores the intricate interplay between terminal uridyltransferase 4 (TUT4) and terminal uridyltransferase 7 (TUT7), the 3'-5' exoribonuclease DIS3L2, and the SKI complex-interacting factor Focadhesin (FOCAD) in the context of cancer vulnerability. Using CRISPR and public functional genomics data, we show impairment of cell proliferation upon knockout of TUT7 or DIS3L2, but not TUT4, on cancer cells with FOCAD loss. Moreover, we report the characterization of the first potent and selective TUT4/7 inhibitors that substantially reduce uridylation and demonstrate in vitro and in vivo antiproliferative activity specifically in FOCAD-deleted cancer. FOCAD deficiency post-transcriptionally disrupts the stability of the SKI complex, whose role is to safeguard cells against aberrant RNA. Re-introduction of FOCAD restores the SKI complex and makes these cells less sensitive to TUT4/7 inhibitors, indicating that TUT7 dependency is FOCAD loss-driven. We propose a model where, in absence of FOCAD, TUT7 and DIS3L2 function as a salvage mechanism that degrades aberrant RNA, and genetic or pharmacological inhibition of this pathway leads to cell death. Our findings underscore the significance of FOCAD loss as a genetic driver of TUT7 vulnerability and provide insights into the potential utility of TUT4/7 inhibitors for cancer treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133245","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TR-107, An Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells. TR-107是酪酸肽酶蛋白水解亚基的一种激动剂,它能破坏线粒体代谢并抑制人类结直肠癌细胞的生长。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-05 DOI: 10.1158/1535-7163.MCT-24-0170
Michael Giarrizzo, Joseph F LaComb, Hetvi R Patel, Rohan G Reddy, John D Haley, Lee M Graves, Edwin J Iwanowicz, Agnieszka B Bialkowska
{"title":"TR-107, An Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells.","authors":"Michael Giarrizzo, Joseph F LaComb, Hetvi R Patel, Rohan G Reddy, John D Haley, Lee M Graves, Edwin J Iwanowicz, Agnieszka B Bialkowska","doi":"10.1158/1535-7163.MCT-24-0170","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0170","url":null,"abstract":"<p><p>Oxidative phosphorylation (OXPHOS) is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates non-specific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound, TR-107, has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multi-targeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer (CRC) cells. TR-107 inhibited CRC cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response (UPRmt) and mtDNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of OXPHOS and a reduction in total cellular respiration. Multi-omics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by CRC cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133246","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IL-15/IL-15Rα-Fc-Fusion Protein XmAb24306 Potentiates Activity of CD3 Bispecific Antibodies through Enhancing T-Cell Expansion. IL-15/IL-15Rα-Fc融合蛋白XmAb24306通过增强T细胞扩增来提高CD3双特异性抗体的活性。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-23-0910
Ji Li, Robyn Clark, Dionysos Slaga, Kendra Avery, Ke Liu, Suzanne Schubbert, Rajat Varma, Eugene Chiang, Klara Totpal, Matthew J Bernett, Patrick G Holder, Teemu T Junttila
{"title":"IL-15/IL-15Rα-Fc-Fusion Protein XmAb24306 Potentiates Activity of CD3 Bispecific Antibodies through Enhancing T-Cell Expansion.","authors":"Ji Li, Robyn Clark, Dionysos Slaga, Kendra Avery, Ke Liu, Suzanne Schubbert, Rajat Varma, Eugene Chiang, Klara Totpal, Matthew J Bernett, Patrick G Holder, Teemu T Junttila","doi":"10.1158/1535-7163.MCT-23-0910","DOIUrl":"10.1158/1535-7163.MCT-23-0910","url":null,"abstract":"<p><p>An insufficient quantity of functional T cells is a likely factor limiting the clinical activity of T-cell bispecific antibodies, especially in solid tumor indications. We hypothesized that XmAb24306 (efbalropendekin alfa), a lymphoproliferative interleukin (IL)-15/IL-15 receptor α (IL-15Rα) Fc-fusion protein, may potentiate the activity of T-cell dependent (TDB) antibodies. The activation of human peripheral T cells by cevostamab, an anti-FcRH5/CD3 TDB, or anti-HER2/CD3 TDB resulted in the upregulation of the IL-2/15Rβ (CD122) receptor subunit in nearly all CD8+ and majority of CD4+ T cells, suggesting that TDB treatment may sensitize T cells to IL-15. XmAb24306 enhanced T-cell bispecific antibody-induced CD8+ and CD4+ T-cell proliferation and expansion. In vitro combination of XmAb24306 with cevostamab or anti-HER2/CD3 TDB resulted in significant enhancement of tumor cell killing, which was reversed when T-cell numbers were normalized, suggesting that T-cell expansion is the main mechanism of the observed benefit. Pretreatment of immunocompetent mice with a mouse-reactive surrogate of XmAb24306 (mIL-15-Fc) resulted in a significant increase of T cells in the blood, spleen, and tumors and converted transient anti-HER2/CD3 TDB responses to complete durable responses. In summary, our results support the hypothesis that the number of tumor-infiltrating T cells is rate limiting for the activity of solid tumor-targeting TDBs. Upregulation of CD122 by TDB treatment and the observed synergy with XmAb24306 and T-cell bispecific antibodies support clinical evaluation of this novel immunotherapy combination.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140912524","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
Correction: Oleanolic Acid Inhibits Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma by Promoting iNOS Dimerization. 更正:齐墩果酸通过促进 iNOS 二聚化抑制肝细胞癌的上皮-间质转化
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-24-0565
Hongzhi Wang, Weilong Zhong, Jianmin Zhao, Heng Zhang, Qiang Zhang, Yuan Liang, Shuang Chen, Huijuan Liu, Shumin Zong, Yixuan Tian, Honggang Zhou, Tao Sun, Yanrong Liu, Cheng Yang
{"title":"Correction: Oleanolic Acid Inhibits Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma by Promoting iNOS Dimerization.","authors":"Hongzhi Wang, Weilong Zhong, Jianmin Zhao, Heng Zhang, Qiang Zhang, Yuan Liang, Shuang Chen, Huijuan Liu, Shumin Zong, Yixuan Tian, Honggang Zhou, Tao Sun, Yanrong Liu, Cheng Yang","doi":"10.1158/1535-7163.MCT-24-0565","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0565","url":null,"abstract":"","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142126165","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
Enhancing Neutrophil Cytotoxicity of a Panel of Clinical EGFR Antibodies by Fc Engineering to IgA3.0. 通过 IgA3.0 的 Fc 工程增强一组临床表皮生长因子受体抗体的中性粒细胞毒性。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-24-0217
Chilam Chan, J H Marco Jansen, Ilona S T Hendriks, Ida C van der Peet, Meggy E L Verdonschot, Elsemieke M Passchier, Maria Tsioumpekou, Maaike Nederend, Sharon A Klomp, Thomas Valerius, Matthias Peipp, Jeanette H W Leusen, Patricia A Olofsen
{"title":"Enhancing Neutrophil Cytotoxicity of a Panel of Clinical EGFR Antibodies by Fc Engineering to IgA3.0.","authors":"Chilam Chan, J H Marco Jansen, Ilona S T Hendriks, Ida C van der Peet, Meggy E L Verdonschot, Elsemieke M Passchier, Maria Tsioumpekou, Maaike Nederend, Sharon A Klomp, Thomas Valerius, Matthias Peipp, Jeanette H W Leusen, Patricia A Olofsen","doi":"10.1158/1535-7163.MCT-24-0217","DOIUrl":"10.1158/1535-7163.MCT-24-0217","url":null,"abstract":"<p><p>EGFR plays an essential role in cellular signaling pathways that regulate cell growth, proliferation, and survival and is often dysregulated in cancer. Several monoclonal IgG antibodies have been clinically tested over the years, which exert their function via blocking the ligand binding domain (thereby inhibiting downstream signaling) and inducing Fc-related effector functions, such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). However, these IgG antibodies do not optimally recruit neutrophils, which are the most abundant white blood cell population in humans. Therefore, we reformatted six therapeutic EGFR antibodies (cetuximab, panitumumab, nimotuzumab, necitumumab, zalutumumab, and matuzumab) into the IgA3.0 format, which is an IgA2 isotype adapted for clinical application. Reformatting these antibodies preserved Fab-mediated functions such as EGFR binding, growth inhibition, and ligand blockade. In addition, whole leukocyte ADCC was significantly increased when using this panel of IgA3.0 antibodies compared with their respective IgG counterparts, with no major differences between IgA3.0 antibodies. In vivo, IgA3.0 matuzumab outperformed the other antibodies, resulting in the strongest suppression of tumor outgrowth in a long intraperitoneal model. We showed that neutrophils are important for the suppression of tumor outgrowth. IgA3.0 matuzumab exhibited reduced receptor internalization compared with the other antibodies, possibly accounting for its superior in vivo Fc-mediated tumor cell killing efficacy. In conclusion, reformatting EGFR antibodies into an IgA3.0 format increased Fc-mediated killing while retaining Fab-mediated functions and could therefore be a good alternative for the currently available antibody therapies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492665","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
MEK Inhibition Enhances the Antitumor Effect of Radiotherapy in NF1-Deficient Glioblastoma. 抑制MEK可增强放疗对NF1缺陷胶质母细胞瘤的抗肿瘤效果。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-23-0510
Maria Ioannou, Kriti Lalwani, Abiola A Ayanlaja, Viveka Chinnasamy, Christine A Pratilas, Karisa C Schreck
{"title":"MEK Inhibition Enhances the Antitumor Effect of Radiotherapy in NF1-Deficient Glioblastoma.","authors":"Maria Ioannou, Kriti Lalwani, Abiola A Ayanlaja, Viveka Chinnasamy, Christine A Pratilas, Karisa C Schreck","doi":"10.1158/1535-7163.MCT-23-0510","DOIUrl":"10.1158/1535-7163.MCT-23-0510","url":null,"abstract":"<p><p>Individuals with neurofibromatosis type 1, an autosomal dominant neurogenetic and tumor predisposition syndrome, are susceptible to developing low-grade glioma and less commonly high-grade glioma. These gliomas exhibit loss of the neurofibromin gene [neurofibromin type 1 (NF1)], and 10% to 15% of sporadic high-grade gliomas have somatic NF1 alterations. Loss of NF1 leads to hyperactive RAS signaling, creating opportunity given the established efficacy of MEK inhibitors in plexiform neurofibromas and some individuals with low-grade glioma. We observed that NF1-deficient glioblastoma neurospheres were sensitive to the combination of an MEK inhibitor (mirdametinib) with irradiation, as evidenced by synergistic inhibition of cell growth, colony formation, and increased cell death. In contrast, NF1-intact neurospheres were not sensitive to the combination, despite complete ERK pathway inhibition. No neurosphere lines exhibited enhanced sensitivity to temozolomide combined with mirdametinib. Mirdametinib decreased transcription of homologous recombination genes and RAD51 foci, associated with DNA damage repair, in sensitive models. Heterotopic xenograft models displayed synergistic growth inhibition to mirdametinib combined with irradiation in NF1-deficient glioma xenografts but not in those with intact NF1. In sensitive models, benefits were observed at least 3 weeks beyond the completion of treatment, including sustained phosphor-ERK inhibition on immunoblot and decreased Ki-67 expression. These observations demonstrate synergistic activity between mirdametinib and irradiation in NF1-deficient glioma models and may have clinical implications for patients with gliomas that harbor germline or somatic NF1 alterations.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11374499/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140876835","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
Combination Immunotherapy with Vaccine and Oncolytic HSV Virotherapy Is Time Dependent. 疫苗与溶解性 HSV 病毒疗法的联合免疫疗法取决于时间。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-23-0873
Stacie K Totsch, Andrew S Ishizuka, Kyung-Don Kang, Sam E Gary, Abbey Rocco, Aaron E Fan, Li Zhou, Pablo A Valdes, SeungHo Lee, Jason Li, Luca Peruzzotti-Jametti, Sarah Blitz, Christopher M Garliss, James M Johnston, James M Markert, Geoffrey M Lynn, Joshua D Bernstock, Gregory K Friedman
{"title":"Combination Immunotherapy with Vaccine and Oncolytic HSV Virotherapy Is Time Dependent.","authors":"Stacie K Totsch, Andrew S Ishizuka, Kyung-Don Kang, Sam E Gary, Abbey Rocco, Aaron E Fan, Li Zhou, Pablo A Valdes, SeungHo Lee, Jason Li, Luca Peruzzotti-Jametti, Sarah Blitz, Christopher M Garliss, James M Johnston, James M Markert, Geoffrey M Lynn, Joshua D Bernstock, Gregory K Friedman","doi":"10.1158/1535-7163.MCT-23-0873","DOIUrl":"10.1158/1535-7163.MCT-23-0873","url":null,"abstract":"<p><p>Oncolytic virotherapy or immunovirotherapy is a strategy that utilizes viruses to selectively infect and kill tumor cells while also stimulating an immune response against the tumor. Early clinical trials in both pediatric and adult patients using oncolytic herpes simplex viruses (oHSV) have demonstrated safety and promising efficacy; however, combinatorial strategies designed to enhance oncolysis while also promoting durable T-cell responses for sustaining disease remission are likely required. We hypothesized that combining the direct tumor cell killing and innate immune stimulation by oHSV with a vaccine that promotes T cell-mediated immunity may lead to more durable tumor regression. To this end, we investigated the preclinical efficacy and potential synergy of combining oHSV with a self-assembling nanoparticle vaccine codelivering peptide antigens and Toll-like receptor 7 and 8 agonists (referred to as SNAPvax),which induces robust tumor-specific T-cell immunity. We then assessed how timing of the treatments (i.e., vaccine before or after oHSV) impacts T-cell responses, viral replication, and preclinical efficacy. The sequence of treatments was critical, as survival was significantly enhanced when the SNAPvax vaccine was given prior to oHSV. Increased clinical efficacy was associated with reduced tumor volume and increases in virus replication and tumor antigen-specific CD8+ T cells. These findings substantiate the criticality of combination immunotherapy timing and provide preclinical support for combining SNAPvax with oHSV as a promising treatment approach for both pediatric and adult tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11374504/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140867304","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信