首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
CXCL12-targeted immunomodulatory gene therapy reduces radiation-induced fibrosis in healthy tissues. cxcl12靶向免疫调节基因治疗可减少健康组织中辐射诱导的纤维化。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-12 DOI: 10.1158/1535-7163.MCT-23-0872
James T Paget, Joseph A Ward, Andrew R McKean, David C Mansfield, Martin McLaughlin, Joan N Kyula-Currie, Henry G Smith, Victoria Roulstone, Chunhei Li, You Zhou, Thomas Hardiman, Anita Grigoriadis, Devin O'Brien Coon, Sheeba Irshad, Alan A Melcher, Kevin J Harrington, Aadil Khan

Radiation-induced fibrosis (RIF) is a progressive pathology deleteriously impacting cancer survivorship. CXCL12 is an immune-stromal signal implicated in fibrosis and innate response. We hypothesised that modulation of CXCL12 would phenotypically mitigate RIF. CXCL12 expression was characterised in a rodent model of RIF and its expression modulated by the intravascular delivery of lentiviral vectors encoding small hairpin RNA to silence (LVShCXCL12) or overexpress (LVOeCXCL12) CXCL12. Multi-modal fibrotic outcomes were quantified, flow cytometry and Y-chromosome lineage-tracking studies performed to examine cellular recruitment and activation post-radiotherapy (post-RT). Whole-tissue RNA-seq was used to examine matrisomal response. MATBIII tumours were engrafted within tissues with differing levels of CXCL12 expression and tumoral response to RT evaluated. CXCL12 was upregulated in irradiated fibroblasts demonstrating DNA-damage post-RT and led to the recruitment of CD68+ macrophages. Silencing Cxcl12 with LVShCXCL12 demonstrated reduced RIF phenotype as a result of decreased macrophage recruitment. Transcriptomic profiling identified osteopontin (SPP1) as being highly differentially expressed in LVShCXCL12-treated tissues. Tumours growing in tissues devoid of CXCL12 expression responded better following RT due to reductions in peri-tumoural fibrosis as a result of decreased CXCL12 and OPN expression at the tumour/normal tissue interface. This was also associated with greater CD8+ T cell infiltration in tumours with less fibrosis. Antibody-mediated OPN blockade slowed tumour growth by increased intra-tumoral CD8+ T cell activation. The CXCL12/OPN axis is an important node of immune/matrisomal cross-talk in the development of fibrosis. Therapeutic manipulation of this axis may offer greater anti-tumour efficacy whilst also reducing adverse effects.

辐射诱导纤维化(RIF)是一种进行性病理,有害地影响癌症的生存。CXCL12是一种涉及纤维化和先天反应的免疫基质信号。我们假设CXCL12的调节会在表型上减轻RIF。在RIF啮齿动物模型中表征了CXCL12的表达,并通过血管内递送编码小发夹RNA的慢病毒载体来调节CXCL12的表达,以沉默(LVShCXCL12)或过表达(LVOeCXCL12) CXCL12。通过流式细胞术和y染色体谱系跟踪研究,对多模式纤维化结果进行了量化,以检查放疗后(后rt)的细胞募集和激活。采用全组织RNA-seq检测基质反应。将MATBIII肿瘤植入具有不同CXCL12表达水平的组织中,并评估肿瘤对RT的反应。CXCL12在放射后显示dna损伤的成纤维细胞中上调,导致CD68+巨噬细胞的募集。用LVShCXCL12沉默Cxcl12表明,由于巨噬细胞募集减少,RIF表型减少。转录组学分析发现骨桥蛋白(SPP1)在lvshcxcl12处理的组织中高度差异表达。在缺乏CXCL12表达的组织中生长的肿瘤在RT后反应更好,这是由于肿瘤/正常组织界面上CXCL12和OPN表达减少导致肿瘤周围纤维化减少。这也与较少纤维化的肿瘤中更多的CD8+ T细胞浸润有关。抗体介导的OPN阻断通过增加肿瘤内CD8+ T细胞激活来减缓肿瘤生长。CXCL12/OPN轴是纤维化发生过程中免疫/基质串扰的重要节点。治疗性操作这条轴可以提供更大的抗肿瘤功效,同时也减少了不良反应。
{"title":"CXCL12-targeted immunomodulatory gene therapy reduces radiation-induced fibrosis in healthy tissues.","authors":"James T Paget, Joseph A Ward, Andrew R McKean, David C Mansfield, Martin McLaughlin, Joan N Kyula-Currie, Henry G Smith, Victoria Roulstone, Chunhei Li, You Zhou, Thomas Hardiman, Anita Grigoriadis, Devin O'Brien Coon, Sheeba Irshad, Alan A Melcher, Kevin J Harrington, Aadil Khan","doi":"10.1158/1535-7163.MCT-23-0872","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0872","url":null,"abstract":"<p><p>Radiation-induced fibrosis (RIF) is a progressive pathology deleteriously impacting cancer survivorship. CXCL12 is an immune-stromal signal implicated in fibrosis and innate response. We hypothesised that modulation of CXCL12 would phenotypically mitigate RIF. CXCL12 expression was characterised in a rodent model of RIF and its expression modulated by the intravascular delivery of lentiviral vectors encoding small hairpin RNA to silence (LVShCXCL12) or overexpress (LVOeCXCL12) CXCL12. Multi-modal fibrotic outcomes were quantified, flow cytometry and Y-chromosome lineage-tracking studies performed to examine cellular recruitment and activation post-radiotherapy (post-RT). Whole-tissue RNA-seq was used to examine matrisomal response. MATBIII tumours were engrafted within tissues with differing levels of CXCL12 expression and tumoral response to RT evaluated. CXCL12 was upregulated in irradiated fibroblasts demonstrating DNA-damage post-RT and led to the recruitment of CD68+ macrophages. Silencing Cxcl12 with LVShCXCL12 demonstrated reduced RIF phenotype as a result of decreased macrophage recruitment. Transcriptomic profiling identified osteopontin (SPP1) as being highly differentially expressed in LVShCXCL12-treated tissues. Tumours growing in tissues devoid of CXCL12 expression responded better following RT due to reductions in peri-tumoural fibrosis as a result of decreased CXCL12 and OPN expression at the tumour/normal tissue interface. This was also associated with greater CD8+ T cell infiltration in tumours with less fibrosis. Antibody-mediated OPN blockade slowed tumour growth by increased intra-tumoral CD8+ T cell activation. The CXCL12/OPN axis is an important node of immune/matrisomal cross-talk in the development of fibrosis. Therapeutic manipulation of this axis may offer greater anti-tumour efficacy whilst also reducing adverse effects.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142813767","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
KT-253, A Novel MDM2 Degrader and p53 Stabilizer, Has Superior Potency and Efficacy Than MDM2 Small Molecule Inhibitors. 一种新型的MDM2降解剂和p53稳定剂KT-253,具有比MDM2小分子抑制剂更好的效力和疗效。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-09 DOI: 10.1158/1535-7163.MCT-24-0306
Yogesh K Chutake, Michele F Mayo, Nancy Dumont, Jessica Filiatrault, Susanne B Breitkopf, Patricia Cho, Dapeng Chen, Vaishali S Dixit, William R Proctor, Eric W Kuhn, Sarah Bollinger Martinez, Alice A McDonald, Jianfeng Qi, Kan-Nian Hu, Rahul Karnik, Joseph D Growney, Kirti Sharma, Stefanie S Schalm, Ashwin M Gollerkeri, Nello Mainolfi, Juliet A Williams, Matthew M Weiss

Murine double minute 2 (MDM2) is an E3 ligase that inhibits the tumor suppressor protein p53. Clinical trials employing small-molecule MDM2/p53 interaction inhibitors (SMIs) have demonstrated limited activity, underscoring an unmet need for a better approach to target MDM2. KT 253 is a highly potent and selective heterobifunctional degrader that overcomes the MDM2 feedback loop seen with SMIs and induces apoptosis in a range of hematologic and solid tumor lines. A single intravenous dose of KT 253 triggered rapid apoptosis and sustained tumor regression in p53 wild-type acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) xenograft models. Additionally, a single intravenous dose of KT 253 in combination with standard-of-care (SoC) venetoclax, overcame venetoclax resistance in an AML xenograft model. The data herein define the therapeutic potential of KT-253 and support its clinical development in a range of hematologic and solid p53 wild-type (WT) malignancies, as a monotherapy and in combination with SoC agents.

小鼠双分钟2 (MDM2)是一种E3连接酶,可抑制肿瘤抑制蛋白p53。临床试验表明,使用小分子MDM2/p53相互作用抑制剂(SMIs)的活性有限,这表明对靶向MDM2的更好方法的需求尚未得到满足。KT 253是一种高效的、选择性的异功能降降剂,它克服了SMIs中出现的MDM2反馈回路,并在一系列血液学和实体肿瘤系中诱导细胞凋亡。单次静脉注射KT 253可在p53野生型急性髓性白血病(AML)和急性淋巴细胞白血病(ALL)异种移植模型中引发快速细胞凋亡和持续肿瘤消退。此外,单次静脉注射KT 253联合标准护理(SoC) venetoclax,在AML异种移植模型中克服了venetoclax耐药性。本文的数据确定了KT-253的治疗潜力,并支持其在一系列血液病和实体p53野生型(WT)恶性肿瘤中的临床开发,可作为单一疗法或与SoC药物联合使用。
{"title":"KT-253, A Novel MDM2 Degrader and p53 Stabilizer, Has Superior Potency and Efficacy Than MDM2 Small Molecule Inhibitors.","authors":"Yogesh K Chutake, Michele F Mayo, Nancy Dumont, Jessica Filiatrault, Susanne B Breitkopf, Patricia Cho, Dapeng Chen, Vaishali S Dixit, William R Proctor, Eric W Kuhn, Sarah Bollinger Martinez, Alice A McDonald, Jianfeng Qi, Kan-Nian Hu, Rahul Karnik, Joseph D Growney, Kirti Sharma, Stefanie S Schalm, Ashwin M Gollerkeri, Nello Mainolfi, Juliet A Williams, Matthew M Weiss","doi":"10.1158/1535-7163.MCT-24-0306","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0306","url":null,"abstract":"<p><p>Murine double minute 2 (MDM2) is an E3 ligase that inhibits the tumor suppressor protein p53. Clinical trials employing small-molecule MDM2/p53 interaction inhibitors (SMIs) have demonstrated limited activity, underscoring an unmet need for a better approach to target MDM2. KT 253 is a highly potent and selective heterobifunctional degrader that overcomes the MDM2 feedback loop seen with SMIs and induces apoptosis in a range of hematologic and solid tumor lines. A single intravenous dose of KT 253 triggered rapid apoptosis and sustained tumor regression in p53 wild-type acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) xenograft models. Additionally, a single intravenous dose of KT 253 in combination with standard-of-care (SoC) venetoclax, overcame venetoclax resistance in an AML xenograft model. The data herein define the therapeutic potential of KT-253 and support its clinical development in a range of hematologic and solid p53 wild-type (WT) malignancies, as a monotherapy and in combination with SoC agents.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142794982","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
First results of migoprotafib, a potent and highly selective Src homology-2 domain-containing phosphatase 2 (SHP2) inhibitor in patients with advanced solid tumors. migoprotafib是一种有效的、高选择性的Src同源-2结构域磷酸酶2 (SHP2)抑制剂,用于晚期实体瘤患者。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-05 DOI: 10.1158/1535-7163.MCT-24-0466
Melissa L Johnson, Beni B Wolf, Judy S Wang, Alexander Philipovskiy, Geoffrey I Shapiro, Bruno Bockorny, Wei Guo, Jinshan Shen, Kai Yu Jen, MaryBeth LeRose, Tamieka Lauz Hunter, Mahesh Padval, Oleg Schmidt-Kittler, Namrata Bhatia, Sarita Dubey, Julia Suchomel, Johanna C Bendell, Shekeab Jauhari, Jennifer Eng-Wong, Jessica J Lin

Src homology-2 domain-containing phosphatase 2 (SHP2) promotes RAS-MAPK signaling and tumorigenesis and is a promising therapeutic target for multiple solid tumors. Migoprotafib is a potent and highly selective SHP2 inhibitor designed for the treatment of RAS-MAPK driven cancers, particularly in combination with other targeted agents. Here we report first-in-human study results of single agent migoprotafib in advanced solid tumor patients. We conducted a phase 1a, open-label, multi-center, dose-escalation and expansion study in adult patients with locally advanced or metastatic solid tumors. The key objectives were to evaluate safety, pharmacokinetics, pharmacodynamics (peripheral blood pERK) and preliminary anti-tumor activity. Fifty-six heavily pre-treated patients were treated with migoprotafib (10-150 mg QD). Migoprotafib had a rapid absorption rate (~0.5-2 hours) with dose-dependent increases in exposure and pathway modulation (pERK changes). The maximum tolerated dose was 100 mg and the recommended phase 2 dose (RP2D) was 60 mg daily (QD) based on safety, pharmacokinetics (PK), pharmacodynamics, and anti-tumor activity. Migoprotafib was generally well tolerated with the most frequent adverse events of diarrhea, peripheral edema, dyspnea, anemia, constipation, fatigue, AST increase and platelet count decrease. Stable disease was observed in 10 patients (18%). Migoprotafib had predictable, dose-dependent PK with an effective half-life that supports QD dosing and demonstrated promising safety, tolerability, and clinical activity at the RP2D. Further clinical testing of migoprotafib in combination with other targeted agents is warranted.

Src同源-2结构域磷酸酶2 (SHP2)促进RAS-MAPK信号传导和肿瘤发生,是多发性实体瘤的一个有希望的治疗靶点。Migoprotafib是一种有效的高选择性SHP2抑制剂,用于治疗RAS-MAPK驱动的癌症,特别是与其他靶向药物联合使用。在这里,我们报告了单药migoprotafib治疗晚期实体瘤患者的首次人体研究结果。我们在患有局部晚期或转移性实体瘤的成人患者中进行了一项1a期、开放标签、多中心、剂量递增和扩展研究。主要目的是评价安全性、药代动力学、药效学(外周血pERK)和初步抗肿瘤活性。56例重度预处理患者接受米格普罗他非治疗(10- 150mg QD)。Migoprotafib具有快速的吸收率(约0.5-2小时),暴露和途径调节(pERK变化)的剂量依赖性增加。根据安全性、药代动力学(PK)、药效学和抗肿瘤活性,最大耐受剂量为100mg,推荐的2期剂量(RP2D)为60mg /天(QD)。Migoprotafib总体耐受良好,最常见的不良事件为腹泻、外周水肿、呼吸困难、贫血、便秘、疲劳、AST升高和血小板计数减少。10例(18%)患者病情稳定。Migoprotafib具有可预测的剂量依赖性PK,其有效半衰期支持QD剂量,并在RP2D中显示出良好的安全性、耐受性和临床活性。migoprotafib联合其他靶向药物的进一步临床试验是有必要的。
{"title":"First results of migoprotafib, a potent and highly selective Src homology-2 domain-containing phosphatase 2 (SHP2) inhibitor in patients with advanced solid tumors.","authors":"Melissa L Johnson, Beni B Wolf, Judy S Wang, Alexander Philipovskiy, Geoffrey I Shapiro, Bruno Bockorny, Wei Guo, Jinshan Shen, Kai Yu Jen, MaryBeth LeRose, Tamieka Lauz Hunter, Mahesh Padval, Oleg Schmidt-Kittler, Namrata Bhatia, Sarita Dubey, Julia Suchomel, Johanna C Bendell, Shekeab Jauhari, Jennifer Eng-Wong, Jessica J Lin","doi":"10.1158/1535-7163.MCT-24-0466","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0466","url":null,"abstract":"<p><p>Src homology-2 domain-containing phosphatase 2 (SHP2) promotes RAS-MAPK signaling and tumorigenesis and is a promising therapeutic target for multiple solid tumors. Migoprotafib is a potent and highly selective SHP2 inhibitor designed for the treatment of RAS-MAPK driven cancers, particularly in combination with other targeted agents. Here we report first-in-human study results of single agent migoprotafib in advanced solid tumor patients. We conducted a phase 1a, open-label, multi-center, dose-escalation and expansion study in adult patients with locally advanced or metastatic solid tumors. The key objectives were to evaluate safety, pharmacokinetics, pharmacodynamics (peripheral blood pERK) and preliminary anti-tumor activity. Fifty-six heavily pre-treated patients were treated with migoprotafib (10-150 mg QD). Migoprotafib had a rapid absorption rate (~0.5-2 hours) with dose-dependent increases in exposure and pathway modulation (pERK changes). The maximum tolerated dose was 100 mg and the recommended phase 2 dose (RP2D) was 60 mg daily (QD) based on safety, pharmacokinetics (PK), pharmacodynamics, and anti-tumor activity. Migoprotafib was generally well tolerated with the most frequent adverse events of diarrhea, peripheral edema, dyspnea, anemia, constipation, fatigue, AST increase and platelet count decrease. Stable disease was observed in 10 patients (18%). Migoprotafib had predictable, dose-dependent PK with an effective half-life that supports QD dosing and demonstrated promising safety, tolerability, and clinical activity at the RP2D. Further clinical testing of migoprotafib in combination with other targeted agents is warranted.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142780608","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
Unveiling the Mechanism of Lurbinectedin's Action and Its Potential in Combination Therapies in Small Cell Lung Cancer. 揭示Lurbinectedin在小细胞肺癌中的作用机制及其联合治疗的潜力。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-05 DOI: 10.1158/1535-7163.MCT-24-0050
Antonio Calles, Emiliano Calvo, Gema Santamaría Nuñez, Federico Costanzo, María José Guillén, Marta Martinez Diez, Aparna Gupta, Carmen Cuevas, Jean-Marc Egly, Pablo Aviles

Lurbinectedin is a selective inhibitor of oncogenic transcription approved for the treatment of adult patients with metastatic small cell lung cancer (SCLC) with disease progression on or after platinum-based chemotherapy. Preclinical data provide evidence for lurbinectedin exerting its actions in a unique manner that involves oncogenic transcription inhibition, DNA damage, reshaping of the tumor microenvironment, and inducing anticancer immunity. Understanding the mechanism of action (MoA) has facilitated the rational combination of lurbinectedin and anticancer therapies with complementary modes of action, in order to obtain synergistic effects that could potentially lead to improved efficacy. This review evaluates the MoA for lurbinectedin and provides an overview of the therapeutic landscape with regards to lurbinectedin combination therapies for the treatment of SCLC based on data from preclinical and clinical studies.

Lurbinectedin是一种选择性的致癌转录抑制剂,被批准用于治疗在铂基化疗期间或之后疾病进展的转移性小细胞肺癌(SCLC)的成人患者。临床前数据证明,lurbinectedin以一种独特的方式发挥其作用,包括致癌转录抑制、DNA损伤、肿瘤微环境重塑和诱导抗癌免疫。了解作用机制(MoA)有助于合理地将lurbinectedin与具有互补作用模式的抗癌药物联合使用,以获得可能导致疗效提高的协同效应。本综述基于临床前和临床研究的数据,评估了lurbinectedin的MoA,并概述了lurbinectedin联合疗法治疗SCLC的治疗前景。
{"title":"Unveiling the Mechanism of Lurbinectedin's Action and Its Potential in Combination Therapies in Small Cell Lung Cancer.","authors":"Antonio Calles, Emiliano Calvo, Gema Santamaría Nuñez, Federico Costanzo, María José Guillén, Marta Martinez Diez, Aparna Gupta, Carmen Cuevas, Jean-Marc Egly, Pablo Aviles","doi":"10.1158/1535-7163.MCT-24-0050","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0050","url":null,"abstract":"<p><p>Lurbinectedin is a selective inhibitor of oncogenic transcription approved for the treatment of adult patients with metastatic small cell lung cancer (SCLC) with disease progression on or after platinum-based chemotherapy. Preclinical data provide evidence for lurbinectedin exerting its actions in a unique manner that involves oncogenic transcription inhibition, DNA damage, reshaping of the tumor microenvironment, and inducing anticancer immunity. Understanding the mechanism of action (MoA) has facilitated the rational combination of lurbinectedin and anticancer therapies with complementary modes of action, in order to obtain synergistic effects that could potentially lead to improved efficacy. This review evaluates the MoA for lurbinectedin and provides an overview of the therapeutic landscape with regards to lurbinectedin combination therapies for the treatment of SCLC based on data from preclinical and clinical studies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142786149","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 evaluation of an anchored immunotherapy strategy with aluminum hydroxide-tethered interleukin-12 in dogs with advanced malignant melanoma. 氢氧化铝拴系白介素-12锚定免疫治疗晚期恶性黑色素瘤的临床前评价
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-05 DOI: 10.1158/1535-7163.MCT-24-0317
Matheus Moreno Passos Barbosa, Rebecca L Kamerer, Joanna Schmit, Angel J Lopez, Rachel Uyehara, Robert Tighe, Sailaja Battula, Howard L Kaufman, Timothy M Fan

Melanoma is an aggressive cancer in dogs involving skin and mucosa similar to people. Anchored immunotherapeutics offer a novel approach to increase intratumoral retention of therapeutic payloads while decreasing systemic exposure, and this strategy can be critically evaluated through a comparative oncology approach. JEN-101 is an anchored canine interleukin-12 (IL-12) tethered to aluminum hydroxide administered by local injection. A Phase I study was conducted to determine the tolerability, activity, and immune responses of JEN-101 in dogs with advanced melanoma. A 3+3 dose escalation design was used to evaluate intratumoral injection of JEN-101 at 1, 3, 10, or 20 μg/kg every three weeks for four cycles. A second course was allowable in the absence of disease progression or toxicity. Peripheral blood, serum, and tumor biopsies were collected at baseline and at pre-specified timepoints for pharmacokinetic and immune analyses, which included serum cytokines, immunohistochemistry, and gene expression assessment. JEN-101 was well tolerated with adverse events being fever, lethargy, and isolated elevated liver enzymes. Five dogs experienced grade 3 events and no grade 4 events were observed. Pharmacokinetic analysis showed a trend towards dose-related Cmax within 8 hours of injection. Responding dogs demonstrated increased systemic interferon-γ and IL-10 AUC levels and local recruitment of CD3+ T cells. Increased pro-inflammatory and antigen processing gene expressions were identified in responding lesions. JEN-101 was well tolerated with evidence of biologic and therapeutic activities. Anchored IL-12 immunotherapy merits further investigation in dogs with melanoma and our approach represents an immune competent model to inform human clinical trials.

黑色素瘤是一种侵袭性癌症,狗的皮肤和粘膜与人类相似。锚定免疫疗法提供了一种新的方法来增加肿瘤内治疗有效载荷的保留,同时减少全身暴露,这种策略可以通过比较肿瘤学方法进行批判性评估。JEN-101是一种锚定犬白介素-12 (IL-12),拴在氢氧化铝上,通过局部注射给药。进行了一期研究,以确定jen101在晚期黑色素瘤犬中的耐受性、活性和免疫反应。采用3+3剂量递增设计,每3周以1、3、10或20 μg/kg剂量注射jen101,共4个周期。在没有疾病进展或毒性的情况下,第二疗程是允许的。在基线和预先指定的时间点收集外周血、血清和肿瘤活检,进行药代动力学和免疫分析,包括血清细胞因子、免疫组织化学和基因表达评估。jen101耐受性良好,不良反应为发热、嗜睡和分离性肝酶升高。5只狗经历了3级事件,没有观察到4级事件。药代动力学分析显示,注射后8小时内Cmax呈剂量相关趋势。响应的狗表现出全身干扰素-γ和IL-10 AUC水平的增加和CD3+ T细胞的局部募集。在反应的病变中发现了促炎和抗原处理基因表达的增加。JEN-101耐受性良好,具有生物和治疗活性。锚定IL-12免疫疗法值得在黑色素瘤犬中进一步研究,我们的方法代表了一种免疫胜任模型,为人类临床试验提供信息。
{"title":"Preclinical evaluation of an anchored immunotherapy strategy with aluminum hydroxide-tethered interleukin-12 in dogs with advanced malignant melanoma.","authors":"Matheus Moreno Passos Barbosa, Rebecca L Kamerer, Joanna Schmit, Angel J Lopez, Rachel Uyehara, Robert Tighe, Sailaja Battula, Howard L Kaufman, Timothy M Fan","doi":"10.1158/1535-7163.MCT-24-0317","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0317","url":null,"abstract":"<p><p>Melanoma is an aggressive cancer in dogs involving skin and mucosa similar to people. Anchored immunotherapeutics offer a novel approach to increase intratumoral retention of therapeutic payloads while decreasing systemic exposure, and this strategy can be critically evaluated through a comparative oncology approach. JEN-101 is an anchored canine interleukin-12 (IL-12) tethered to aluminum hydroxide administered by local injection. A Phase I study was conducted to determine the tolerability, activity, and immune responses of JEN-101 in dogs with advanced melanoma. A 3+3 dose escalation design was used to evaluate intratumoral injection of JEN-101 at 1, 3, 10, or 20 μg/kg every three weeks for four cycles. A second course was allowable in the absence of disease progression or toxicity. Peripheral blood, serum, and tumor biopsies were collected at baseline and at pre-specified timepoints for pharmacokinetic and immune analyses, which included serum cytokines, immunohistochemistry, and gene expression assessment. JEN-101 was well tolerated with adverse events being fever, lethargy, and isolated elevated liver enzymes. Five dogs experienced grade 3 events and no grade 4 events were observed. Pharmacokinetic analysis showed a trend towards dose-related Cmax within 8 hours of injection. Responding dogs demonstrated increased systemic interferon-γ and IL-10 AUC levels and local recruitment of CD3+ T cells. Increased pro-inflammatory and antigen processing gene expressions were identified in responding lesions. JEN-101 was well tolerated with evidence of biologic and therapeutic activities. Anchored IL-12 immunotherapy merits further investigation in dogs with melanoma and our approach represents an immune competent model to inform human clinical trials.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142780612","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
Denfivontinib activates effector T-cells through NLRP3-inflammasome, yielding potent anticancer effects by combination with pembrolizumab. Denfivontinib通过nlrp3炎性体激活效应t细胞,与pembrolizumab联合产生有效的抗癌效果。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-05 DOI: 10.1158/1535-7163.MCT-24-0501
Dong Kwon Kim, Chun-Bong Synn, Wongeun Lee, Ha-Ni Jo, Chai Young Lee, Seul Lee, Joon Yeon Hwang, Youngtaek Kim, Seong-San Kang, Sujeong Baek, Kwangmin Na, Seung Min Yang, Mi Hyun Kim, Heekyung Han, Yu Jin Han, Jae Hwan Kim, So Young Park, Young Joon Park, Gang-Taik Lee, Su-Jin Choi, Jie-Ohn Sohn, Sang-Kyu Ye, Jii Bum Lee, Sun Min Lim, Min Hee Hong, Kyoung-Ho Pyo, Byoung Chul Cho

Various combination therapies have been investigated to overcome the limitations of using immune checkpoint inhibitors. However, determining the optimal combination therapy remains challenging. To overcome the therapeutical limitation, we conducted a translational research to elucidate the mechanisms by which AXL inhibition enhances the anti-tumor effects when combined with anti-PD-1 antibody therapy. Herein, we demonstrated improved antitumor effects through combination treatment with denfivontinib and pembrolizumab which resulted in enhanced differentiation into effector CD4+ and CD8+ memory T cells, accompanied by an increase in IFN-γ expression in the YHIM-2004 xenograft model derived from patients with NSCLC. Concurrently, a reduction in the number of immunosuppressive M2 macrophages and myeloid-derived suppressor cells was observed. Mechanistically, denfivontinib potentiated the NOD-like receptor pathway, thereby facilitating the NLRP3 inflammasome formation. This leads to macrophage activation via the NF-kB signaling pathway activation. We have confirmed that the positive interaction between macrophages and T cells arises from the enhanced antigen-presenting machinery of activated macrophages. Furthermore, the observed tumor effects in AXL knock-out mice confirmed that AXL inhibition by denfivontinib enhances the anti-tumor effects, thus opening new avenues for therapeutic interventions aimed at overcoming limitations in immunotherapy. To demonstrate the extent to which our findings reflect clinical results, we analyzed bulk-RNA sequencing data from 21 NSCLC patients undergoing anti-PD-1 immunotherapy. The NLRP3 inflammasome score influenced enhanced immune responses in patient data undergoing anti-PD-1 immunotherapy, suggesting a role for NLRP3 inflammasome in activating immune responses during treatment.

已经研究了各种联合疗法来克服使用免疫检查点抑制剂的局限性。然而,确定最佳的联合治疗仍然具有挑战性。为了克服治疗局限性,我们进行了一项转化研究,以阐明AXL抑制与抗pd -1抗体治疗联合增强抗肿瘤作用的机制。在本研究中,我们证明了通过联合使用denfivontinib和pembrolizumab可以提高抗肿瘤效果,从而增强向效应CD4+和CD8+记忆T细胞的分化,同时在来自NSCLC患者的yim -2004异种移植模型中,IFN-γ表达增加。同时,观察到免疫抑制M2巨噬细胞和髓源性抑制细胞的数量减少。从机制上讲,非非替尼增强了nod样受体途径,从而促进了NLRP3炎性体的形成。这导致巨噬细胞通过NF-kB信号通路激活。我们已经证实,巨噬细胞和T细胞之间的积极相互作用源于活化的巨噬细胞增强的抗原呈递机制。此外,在AXL敲除小鼠中观察到的肿瘤效应证实,denfivontinib抑制AXL增强了抗肿瘤作用,从而为克服免疫治疗的局限性开辟了新的治疗干预途径。为了证明我们的发现在一定程度上反映了临床结果,我们分析了21名接受抗pd -1免疫治疗的非小细胞肺癌患者的大体积rna测序数据。在接受抗pd -1免疫治疗的患者数据中,NLRP3炎症小体评分影响增强的免疫反应,提示NLRP3炎症小体在治疗期间激活免疫反应中的作用。
{"title":"Denfivontinib activates effector T-cells through NLRP3-inflammasome, yielding potent anticancer effects by combination with pembrolizumab.","authors":"Dong Kwon Kim, Chun-Bong Synn, Wongeun Lee, Ha-Ni Jo, Chai Young Lee, Seul Lee, Joon Yeon Hwang, Youngtaek Kim, Seong-San Kang, Sujeong Baek, Kwangmin Na, Seung Min Yang, Mi Hyun Kim, Heekyung Han, Yu Jin Han, Jae Hwan Kim, So Young Park, Young Joon Park, Gang-Taik Lee, Su-Jin Choi, Jie-Ohn Sohn, Sang-Kyu Ye, Jii Bum Lee, Sun Min Lim, Min Hee Hong, Kyoung-Ho Pyo, Byoung Chul Cho","doi":"10.1158/1535-7163.MCT-24-0501","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0501","url":null,"abstract":"<p><p>Various combination therapies have been investigated to overcome the limitations of using immune checkpoint inhibitors. However, determining the optimal combination therapy remains challenging. To overcome the therapeutical limitation, we conducted a translational research to elucidate the mechanisms by which AXL inhibition enhances the anti-tumor effects when combined with anti-PD-1 antibody therapy. Herein, we demonstrated improved antitumor effects through combination treatment with denfivontinib and pembrolizumab which resulted in enhanced differentiation into effector CD4+ and CD8+ memory T cells, accompanied by an increase in IFN-γ expression in the YHIM-2004 xenograft model derived from patients with NSCLC. Concurrently, a reduction in the number of immunosuppressive M2 macrophages and myeloid-derived suppressor cells was observed. Mechanistically, denfivontinib potentiated the NOD-like receptor pathway, thereby facilitating the NLRP3 inflammasome formation. This leads to macrophage activation via the NF-kB signaling pathway activation. We have confirmed that the positive interaction between macrophages and T cells arises from the enhanced antigen-presenting machinery of activated macrophages. Furthermore, the observed tumor effects in AXL knock-out mice confirmed that AXL inhibition by denfivontinib enhances the anti-tumor effects, thus opening new avenues for therapeutic interventions aimed at overcoming limitations in immunotherapy. To demonstrate the extent to which our findings reflect clinical results, we analyzed bulk-RNA sequencing data from 21 NSCLC patients undergoing anti-PD-1 immunotherapy. The NLRP3 inflammasome score influenced enhanced immune responses in patient data undergoing anti-PD-1 immunotherapy, suggesting a role for NLRP3 inflammasome in activating immune responses during treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142780606","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
Immunoproteasome Activation Expands the MHC Class I Immunopeptidome, Unmasks Neoantigens, and Enhances T-cell Anti-Myeloma Activity. 免疫蛋白酶体激活可扩展 MHC I 类免疫肽组,揭示新抗原并增强 T 细胞的抗骨髓瘤活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-23-0931
Priyanka S Rana, James J Ignatz-Hoover, Chunna Guo, Amber L Mosley, Ehsan Malek, Yuriy Federov, Drew J Adams, James J Driscoll

Proteasomes generate antigenic peptides that are presented on the tumor surface to cytotoxic T-lymphocytes. Immunoproteasomes are highly specialized proteasome variants that are expressed at higher levels in antigen-presenting cells and contain replacements of the three constitutive proteasome catalytic subunits to generate peptides with a hydrophobic C-terminus that fit within the groove of MHC class I (MHC-I) molecules. A hallmark of cancer is the ability to evade immunosurveillance by disrupting the antigen presentation machinery and downregulating MHC-I antigen presentation. High-throughput screening was performed to identify compound A, a novel molecule that selectively increased immunoproteasome activity and expanded the number and diversity of MHC-I-bound peptides presented on multiple myeloma cells. Compound A increased the presentation of individual MHC-I-bound peptides by >100-fold and unmasked tumor-specific neoantigens on myeloma cells. Global proteomic integral stability assays determined that compound A binds to the proteasome structural subunit PSMA1 and promotes association of the proteasome activator PA28α/β (PSME1/PSME2) with immunoproteasomes. CRISPR/Cas9 silencing of PSMA1, PSME1, or PSME2 as well as treatment with immunoproteasome-specific suicide inhibitors abolished the effects of compound A on antigen presentation. Treatment of multiple myeloma cell lines and patient bone marrow-derived CD138+ cells with compound A increased the anti-myeloma activity of allogenic and autologous T cells. Compound A was well-tolerated in vivo and co-treatment with allogeneic T cells reduced the growth of myeloma xenotransplants in NOD/SCID gamma mice. Taken together, our results demonstrate the paradigm shifting impact of immunoproteasome activators to diversify the antigenic landscape, expand the immunopeptidome, potentiate T-cell-directed therapy, and reveal actionable neoantigens for personalized T-cell immunotherapy.

蛋白酶体产生抗原肽,并在肿瘤表面呈现给细胞毒性 T 淋巴细胞(CTL)。免疫蛋白酶体是高度特化的蛋白酶体变体,在抗原递呈细胞中的表达量较高,包含三个组成型蛋白酶体催化亚基的替代物,生成的肽具有疏水性 C 端,适合 MHC I 类(MHC-I)分子的沟槽。癌症的一个特征是能够通过破坏抗原递呈机制和下调 MHC-I 抗原递呈来逃避免疫监视。高通量筛选确定了化合物 A,这是一种新型分子,可选择性地提高免疫蛋白酶体的活性,并增加多发性骨髓瘤(MM)细胞上呈递的 MHC-I 结合肽的数量和多样性。化合物 A 能使单个 MHC-I 结合肽的呈现率提高 100 倍以上,并能揭示骨髓瘤细胞上的肿瘤特异性新抗原。全局蛋白质组整体稳定性测定确定,化合物 A 能与蛋白酶体结构亚基 PSMA1 结合,并促进蛋白酶体激活剂 PA28α/β (PSME1/PSME2)与免疫蛋白酶体的结合。CRISPR/Cas9沉默PSMA1、PSME1或PSME2以及用免疫蛋白酶体特异性自杀抑制剂处理可消除化合物A对抗原呈递的影响。用化合物 A 处理 MM 细胞系和患者骨髓来源的 CD138+ 细胞可提高异体和自体 T 细胞的抗骨髓瘤活性。化合物 A 在体内耐受性良好,与异体 T 细胞联合处理可减少骨髓瘤异种移植在 NSG 小鼠体内的生长。综上所述,我们的研究结果表明了免疫蛋白酶体激活剂在使抗原景观多样化、扩大免疫肽体、增强T细胞导向疗法以及揭示可用于个性化T细胞免疫疗法的新抗原等方面的范式转换影响。
{"title":"Immunoproteasome Activation Expands the MHC Class I Immunopeptidome, Unmasks Neoantigens, and Enhances T-cell Anti-Myeloma Activity.","authors":"Priyanka S Rana, James J Ignatz-Hoover, Chunna Guo, Amber L Mosley, Ehsan Malek, Yuriy Federov, Drew J Adams, James J Driscoll","doi":"10.1158/1535-7163.MCT-23-0931","DOIUrl":"10.1158/1535-7163.MCT-23-0931","url":null,"abstract":"<p><p>Proteasomes generate antigenic peptides that are presented on the tumor surface to cytotoxic T-lymphocytes. Immunoproteasomes are highly specialized proteasome variants that are expressed at higher levels in antigen-presenting cells and contain replacements of the three constitutive proteasome catalytic subunits to generate peptides with a hydrophobic C-terminus that fit within the groove of MHC class I (MHC-I) molecules. A hallmark of cancer is the ability to evade immunosurveillance by disrupting the antigen presentation machinery and downregulating MHC-I antigen presentation. High-throughput screening was performed to identify compound A, a novel molecule that selectively increased immunoproteasome activity and expanded the number and diversity of MHC-I-bound peptides presented on multiple myeloma cells. Compound A increased the presentation of individual MHC-I-bound peptides by >100-fold and unmasked tumor-specific neoantigens on myeloma cells. Global proteomic integral stability assays determined that compound A binds to the proteasome structural subunit PSMA1 and promotes association of the proteasome activator PA28α/β (PSME1/PSME2) with immunoproteasomes. CRISPR/Cas9 silencing of PSMA1, PSME1, or PSME2 as well as treatment with immunoproteasome-specific suicide inhibitors abolished the effects of compound A on antigen presentation. Treatment of multiple myeloma cell lines and patient bone marrow-derived CD138+ cells with compound A increased the anti-myeloma activity of allogenic and autologous T cells. Compound A was well-tolerated in vivo and co-treatment with allogeneic T cells reduced the growth of myeloma xenotransplants in NOD/SCID gamma mice. Taken together, our results demonstrate the paradigm shifting impact of immunoproteasome activators to diversify the antigenic landscape, expand the immunopeptidome, potentiate T-cell-directed therapy, and reveal actionable neoantigens for personalized T-cell immunotherapy.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1743-1760"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11612626/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142109603","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
Preclinical Characterization of ARX517, a Site-Specific Stable PSMA-Targeted Antibody-Drug Conjugate for the Treatment of Metastatic Castration-Resistant Prostate Cancer. 用于治疗转移性钙化抗性前列腺癌的特异性稳定 PSMA 靶向抗体药物共轭物 ARX517 的临床前特征。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-23-0927
Lillian K Skidmore, David Mills, Ji Young Kim, Nick A Knudsen, Jay D Nelson, Manoj Pal, Jianing Wang, Kedar Gc, Michael J Gray, Wisam Barkho, Prathap Nagaraja Shastri, Mysore P Ramprasad, Feng Tian, Daniel O'Connor, Ying J Buechler, Shawn Shao-Hui Zhang

Metastatic castration-resistant prostate cancer (mCRPC) is an advanced disease in which patients ultimately fail standard-of-care androgen deprivation therapies and exhibit poor survival rates. The prostate-specific membrane antigen (PSMA) has been validated as an mCRPC tumor antigen with overexpression in tumors and low expression in healthy tissues. Using our proprietary technology for incorporating synthetic amino acids into proteins at selected sites, we have developed ARX517, an antibody-drug conjugate composed of a humanized anti-PSMA antibody site-specifically conjugated to a tubulin inhibitor at a drug-to-antibody ratio of 2. After binding PSMA, ARX517 is internalized and catabolized, leading to cytotoxic payload delivery and apoptosis. To minimize premature payload release and maximize delivery to tumor cells, ARX517 employs a noncleavable polyethylene glycol linker and stable oxime conjugation enabled via synthetic amino acid protein incorporation to ensure its overall stability. In vitro studies demonstrate that ARX517 selectively induces cytotoxicity of PSMA-expressing tumor cell lines. ARX517 exhibited a long terminal half-life and high serum exposure in mice and dose-dependent antitumor activity in both enzalutamide-sensitive and -resistant cell line-derived xenograft and patient-derived xenograft models of prostate cancer. Repeat-dose toxicokinetic studies in nonhuman primates demonstrated that ARX517 was tolerated at exposures well above therapeutic exposures in mouse pharmacology studies, indicating a wide therapeutic index. In summary, ARX517 inhibited tumor growth in diverse mCRPC models, demonstrated a tolerable safety profile in monkeys, and had a wide therapeutic index based on preclinical exposure data. Based on the encouraging preclinical data, ARX517 is currently being evaluated in a phase I clinical trial (NCT04662580).

转移性抗性前列腺癌(mCRPC)是一种晚期疾病,患者最终无法接受标准的雄激素剥夺疗法,生存率很低。前列腺特异性膜抗原(PSMA)已被证实是一种在肿瘤中过度表达而在健康组织中低表达的 mCRPC 肿瘤抗原。利用我们在选定位点将合成氨基酸 (SAAs) 加入蛋白质的专有技术,我们开发出了抗体药物共轭物 (ADC)--ARX517,它由人源化的抗 PSMA 抗体位点特异性地与微管蛋白抑制剂结合而成,药物与抗体的比例为 2。为了最大限度地减少有效载荷的过早释放并最大限度地向肿瘤细胞递送,ARX517 采用了不易破碎的 PEG 连接体,并通过 SAA 蛋白结合实现稳定的肟连接,以确保其整体稳定性。体外研究表明,ARX517 可选择性地诱导表达 PSMA 的肿瘤细胞株产生细胞毒性。ARX517 在小鼠体内具有较长的终末半衰期和较高的血清暴露量,在对恩扎鲁胺敏感和耐药的 CDX 和 PDX 前列腺癌模型中均表现出剂量依赖性抗肿瘤活性。在非人灵长类动物中进行的重复剂量毒物动力学研究表明,ARX517 的耐受暴露量远高于小鼠药理学研究中的治疗暴露量,这表明它具有广泛的治疗指数。总之,ARX517 可抑制多种 mCRPC 模型中的肿瘤生长,在猴子体内表现出可耐受的安全性,并且根据临床前暴露数据,具有广泛的治疗指数。基于令人鼓舞的临床前数据,ARX517 目前正接受 1 期临床试验([NCT04662580])的评估。
{"title":"Preclinical Characterization of ARX517, a Site-Specific Stable PSMA-Targeted Antibody-Drug Conjugate for the Treatment of Metastatic Castration-Resistant Prostate Cancer.","authors":"Lillian K Skidmore, David Mills, Ji Young Kim, Nick A Knudsen, Jay D Nelson, Manoj Pal, Jianing Wang, Kedar Gc, Michael J Gray, Wisam Barkho, Prathap Nagaraja Shastri, Mysore P Ramprasad, Feng Tian, Daniel O'Connor, Ying J Buechler, Shawn Shao-Hui Zhang","doi":"10.1158/1535-7163.MCT-23-0927","DOIUrl":"10.1158/1535-7163.MCT-23-0927","url":null,"abstract":"<p><p>Metastatic castration-resistant prostate cancer (mCRPC) is an advanced disease in which patients ultimately fail standard-of-care androgen deprivation therapies and exhibit poor survival rates. The prostate-specific membrane antigen (PSMA) has been validated as an mCRPC tumor antigen with overexpression in tumors and low expression in healthy tissues. Using our proprietary technology for incorporating synthetic amino acids into proteins at selected sites, we have developed ARX517, an antibody-drug conjugate composed of a humanized anti-PSMA antibody site-specifically conjugated to a tubulin inhibitor at a drug-to-antibody ratio of 2. After binding PSMA, ARX517 is internalized and catabolized, leading to cytotoxic payload delivery and apoptosis. To minimize premature payload release and maximize delivery to tumor cells, ARX517 employs a noncleavable polyethylene glycol linker and stable oxime conjugation enabled via synthetic amino acid protein incorporation to ensure its overall stability. In vitro studies demonstrate that ARX517 selectively induces cytotoxicity of PSMA-expressing tumor cell lines. ARX517 exhibited a long terminal half-life and high serum exposure in mice and dose-dependent antitumor activity in both enzalutamide-sensitive and -resistant cell line-derived xenograft and patient-derived xenograft models of prostate cancer. Repeat-dose toxicokinetic studies in nonhuman primates demonstrated that ARX517 was tolerated at exposures well above therapeutic exposures in mouse pharmacology studies, indicating a wide therapeutic index. In summary, ARX517 inhibited tumor growth in diverse mCRPC models, demonstrated a tolerable safety profile in monkeys, and had a wide therapeutic index based on preclinical exposure data. Based on the encouraging preclinical data, ARX517 is currently being evaluated in a phase I clinical trial (NCT04662580).</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1842-1853"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11612621/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142036417","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
Separable Cell Cycle Arrest and Immune Response Elicited through Pharmacological CDK4/6 and MEK Inhibition in RASmut Disease Models. 在 RASmut 疾病模型中通过药理 CDK4/6 和 MEK 抑制引起可分离的细胞周期停滞和免疫反应。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-24-0369
Jin Wu, Jianxin Wang, Thomas N O'Connor, Stephanie L Tzetzo, Katerina V Gurova, Erik S Knudsen, Agnieszka K Witkiewicz

The combination of CDK4/6 and MEK inhibition as a therapeutic strategy has shown promise in various cancer models, particularly in those harboring RAS mutations. An initial high-throughput drug screen identified high synergy between the CDK4/6 inhibitor palbociclib and the MEK inhibitor trametinib when used in combination in soft tissue sarcomas. In RAS mutant models, combination treatment with palbociclib and trametinib induced significant G1 cell cycle arrest, resulting in a marked reduction in cell proliferation and growth. CRISPR-mediated RB1 depletion resulted in a decreased response to CDK4/6 and MEK inhibition, which was validated in both cell culture and xenograft models. Beyond its cell cycle inhibitory effects, pathway enrichment analysis revealed the robust activation of interferon pathways upon CDK4/6 and MEK inhibition. This induction of gene expression was associated with the upregulation of retroviral elements. The TANK-binding kinase 1 inhibitor GSK8612 selectively blocked the induction of interferon-related genes induced by palbociclib and trametinib treatment and highlighted the separable epigenetic responses elicited by combined CDK4/6 and MEK inhibition. Together, these findings provide key mechanistic insights into the therapeutic potential of CDK4/6 and MEK inhibition in soft tissue sarcomas.

CDK4/6和MEK抑制剂联合作为一种治疗策略已在各种癌症模型中显示出前景,尤其是在携带RAS突变的癌症模型中。最初的高通量药物筛选发现,CDK4/6抑制剂palbociclib和MEK抑制剂曲美替尼联用治疗软组织肉瘤具有高度协同作用。在RAS突变模型中,palbociclib和曲美替尼的联合治疗可诱导G1细胞周期显著停滞,从而明显减少细胞的增殖和生长。CRISPR 介导的 RB1 缺失导致对 CDK4/6 和 MEK 抑制的反应减弱,这在细胞培养和异种移植模型中都得到了验证。除了细胞周期抑制作用外,通路富集分析还显示,CDK4/6和MEK抑制可显著激活干扰素通路。这种基因表达的诱导与逆转录病毒元件的上调有关。TBK1(TANK结合激酶1)抑制剂GSK8612选择性地阻断了palbociclib和曲美替尼治疗诱导的干扰素相关基因的诱导,并强调了CDK4/6和MEK联合抑制引起的可分离的表观遗传学反应。总之,这些发现为CDK4/6和MEK抑制在软组织肉瘤中的治疗潜力提供了重要的机理启示。
{"title":"Separable Cell Cycle Arrest and Immune Response Elicited through Pharmacological CDK4/6 and MEK Inhibition in RASmut Disease Models.","authors":"Jin Wu, Jianxin Wang, Thomas N O'Connor, Stephanie L Tzetzo, Katerina V Gurova, Erik S Knudsen, Agnieszka K Witkiewicz","doi":"10.1158/1535-7163.MCT-24-0369","DOIUrl":"10.1158/1535-7163.MCT-24-0369","url":null,"abstract":"<p><p>The combination of CDK4/6 and MEK inhibition as a therapeutic strategy has shown promise in various cancer models, particularly in those harboring RAS mutations. An initial high-throughput drug screen identified high synergy between the CDK4/6 inhibitor palbociclib and the MEK inhibitor trametinib when used in combination in soft tissue sarcomas. In RAS mutant models, combination treatment with palbociclib and trametinib induced significant G1 cell cycle arrest, resulting in a marked reduction in cell proliferation and growth. CRISPR-mediated RB1 depletion resulted in a decreased response to CDK4/6 and MEK inhibition, which was validated in both cell culture and xenograft models. Beyond its cell cycle inhibitory effects, pathway enrichment analysis revealed the robust activation of interferon pathways upon CDK4/6 and MEK inhibition. This induction of gene expression was associated with the upregulation of retroviral elements. The TANK-binding kinase 1 inhibitor GSK8612 selectively blocked the induction of interferon-related genes induced by palbociclib and trametinib treatment and highlighted the separable epigenetic responses elicited by combined CDK4/6 and MEK inhibition. Together, these findings provide key mechanistic insights into the therapeutic potential of CDK4/6 and MEK inhibition in soft tissue sarcomas.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1801-1814"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11614708/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141988386","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
Integrated Imaging Probe and Bispecific Antibody Development Enables In Vivo Targeting of Glypican-3-Expressing Hepatocellular Carcinoma. 集成成像探针和双特异性抗体开发,实现体内靶向治疗表达 glypican-3 的肝细胞癌。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-12-03 DOI: 10.1158/1535-7163.MCT-23-0470
Peiman Habibollahi, Alexey Gurevich, James Z Hui, Kelly Weinfurtner, George McClung, Justin Adler, Michael C Soulen, David E Kaplan, Gregory J Nadolski, Stephen J Hunt, Andrew Tsourkas, Terence P Gade

Glypican-3 (GPC3) is a proteoglycan with high sensitivity and specificity for hepatocellular carcinoma (HCC). We describe the integrated development and validation of a GPC3-targeting optical imaging probe and T cell-redirecting antibody (TRAB) as a theranostic strategy for the detection and treatment of HCC. A novel TRAB targeting GPC3 on HCC tumor cells and the CD3 T-cell receptor as well as a distinct GPC3-specific optical imaging probe were developed from a short peptide. The efficacy of GPC3/CD3 TRAB was evaluated in vitro using IFNγ release and calcein-AM assays. Patient-derived xenografts were used to assess the in vivo efficacy of GPC3/CD3 TRAB and the GPC3 imaging probe for the detection of GPC3+ HCC. GPC3/CD3 TRAB caused a dose-dependent escalation in IFNγ release from inactive peripheral blood T cells (P = 0.001) and higher tumor-cell lysis (P = 0.01) compared with controls in vitro. Intratumorally injected GPC3/CD3 TRAB resulted in significant prolongation of tumor doubling time in the GPC3+ tumors, with an associated reduction of tumor fluorescent signal from the HiLyte 488-conjugated GPC3-specific peptide on optical imaging. These data demonstrate that HCC cell targeting using a GPC3/CD3 TRAB derived from a small peptide enabled effective T-cell activation and induction of a cytotoxic response toward GPC3+ HCC tumor cells both in vitro and in vivo. GPC3-specific optical imaging enabled the detection of the GPC3+ HCC cells and noninvasive monitoring of tumor response to adoptive immunotherapy. The integrated development of a targeted therapeutic and molecular imaging probe provides a promising paradigm for the development of cancer theranostics.

Glypican-3(GPC3)是一种蛋白聚糖,对肝细胞癌(HCC)具有高灵敏度和特异性。我们介绍了 GPC3 靶向光学成像探针和 T 细胞重定向抗体(TRAB)作为检测和治疗 HCC 的治疗策略的综合开发和验证。研究人员利用一种短肽开发出了一种新型的针对 HCC 肿瘤细胞上的 GPC3 和 CD3 T 细胞受体的 TRAB 以及一种独特的 GPC3 特异性光学成像探针。利用干扰素-γ释放和钙黄绿素-AM测定法在体外评估了GPC3/CD3 TRAB的疗效。患者衍生异种移植(PDX)用于评估 GPC3/CD3 TRAB 和 GPC3 成像探针在检测 GPC3+ HCC 方面的体内疗效。与体外对照组相比,GPC3/CD3 TRAB 可使非活性外周血 T 细胞释放的干扰素-γ 呈剂量依赖性增加(P = 0.001),肿瘤细胞裂解率更高(P = 0.01)。瘤内注射 GPC3/CD3 TRAB 可显著延长 GPC3+ PDX 小鼠的肿瘤倍增时间,同时在光学成像中,HiLyte 488 共轭 GPC3 特异性肽的肿瘤荧光信号也会随之减少。使用源自小肽的 GPC3/CD3 TRAB 靶向 HCC 细胞可有效激活 T 细胞,并在体外和体内诱导针对 GPC3+ HCC 肿瘤细胞的细胞毒反应。GPC3特异性光学成像能够检测GPC3+ HCC细胞,并对肿瘤对采纳性免疫疗法的反应进行无创监测。靶向治疗和分子成像探针的综合开发为开发癌症治疗技术提供了一种新的范例。
{"title":"Integrated Imaging Probe and Bispecific Antibody Development Enables In Vivo Targeting of Glypican-3-Expressing Hepatocellular Carcinoma.","authors":"Peiman Habibollahi, Alexey Gurevich, James Z Hui, Kelly Weinfurtner, George McClung, Justin Adler, Michael C Soulen, David E Kaplan, Gregory J Nadolski, Stephen J Hunt, Andrew Tsourkas, Terence P Gade","doi":"10.1158/1535-7163.MCT-23-0470","DOIUrl":"10.1158/1535-7163.MCT-23-0470","url":null,"abstract":"<p><p>Glypican-3 (GPC3) is a proteoglycan with high sensitivity and specificity for hepatocellular carcinoma (HCC). We describe the integrated development and validation of a GPC3-targeting optical imaging probe and T cell-redirecting antibody (TRAB) as a theranostic strategy for the detection and treatment of HCC. A novel TRAB targeting GPC3 on HCC tumor cells and the CD3 T-cell receptor as well as a distinct GPC3-specific optical imaging probe were developed from a short peptide. The efficacy of GPC3/CD3 TRAB was evaluated in vitro using IFNγ release and calcein-AM assays. Patient-derived xenografts were used to assess the in vivo efficacy of GPC3/CD3 TRAB and the GPC3 imaging probe for the detection of GPC3+ HCC. GPC3/CD3 TRAB caused a dose-dependent escalation in IFNγ release from inactive peripheral blood T cells (P = 0.001) and higher tumor-cell lysis (P = 0.01) compared with controls in vitro. Intratumorally injected GPC3/CD3 TRAB resulted in significant prolongation of tumor doubling time in the GPC3+ tumors, with an associated reduction of tumor fluorescent signal from the HiLyte 488-conjugated GPC3-specific peptide on optical imaging. These data demonstrate that HCC cell targeting using a GPC3/CD3 TRAB derived from a small peptide enabled effective T-cell activation and induction of a cytotoxic response toward GPC3+ HCC tumor cells both in vitro and in vivo. GPC3-specific optical imaging enabled the detection of the GPC3+ HCC cells and noninvasive monitoring of tumor response to adoptive immunotherapy. The integrated development of a targeted therapeutic and molecular imaging probe provides a promising paradigm for the development of cancer theranostics.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1815-1826"},"PeriodicalIF":5.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11726262/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291434","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