Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1232
D. Debnath, S. Nazzal, S. Acharya, H. Souza, P. F. Filho, R. Fudała, J. Mathis
{"title":"Abstract 1232: Using mesoporous silica nanoparticles (MSNs) for delivering the mesoionic compound MIH 2.4Bl in treating breast cancer","authors":"D. Debnath, S. Nazzal, S. Acharya, H. Souza, P. F. Filho, R. Fudała, J. Mathis","doi":"10.1158/1538-7445.AM2021-1232","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1232","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78244717","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1434
Renuka Raman, Jacques A Villefranc, T. Ullmann, Jessica W. Thiesmeyer, V. Anelli, C. Pauli, R. Bareja, K. Eng, P. Dorsaint, D. Wilkes, Shaham Beg, Reid Shaw, Michael Churchill, Naveen Gumpeni, T. Scognamiglio, M. Rubin, C. Grandori, J. Mosquera, Noah E. Dephoure, A. Sboner, O. Elemento, Y. Houvras
{"title":"Abstract 1434: Uncovering the mechanism of adaptive resistance to RET inhibitors in RET rearranged thyroid cancer","authors":"Renuka Raman, Jacques A Villefranc, T. Ullmann, Jessica W. Thiesmeyer, V. Anelli, C. Pauli, R. Bareja, K. Eng, P. Dorsaint, D. Wilkes, Shaham Beg, Reid Shaw, Michael Churchill, Naveen Gumpeni, T. Scognamiglio, M. Rubin, C. Grandori, J. Mosquera, Noah E. Dephoure, A. Sboner, O. Elemento, Y. Houvras","doi":"10.1158/1538-7445.AM2021-1434","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1434","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78245003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-913
R. Austin, W. Aaron, Manasi Barath, Evan C. Callihan, Scott Gatto, Golzar Hemmati, C. Law, D. LemonBryan, J. O'rear, Purbasa Patnaik, Morgan Thompson, Lihn To, H. Wesche, Kevin Wright, Yinghua Xiao, Stephen Yu, Timothy Z. Yu
FLT3 (fms related receptor tyrosine kinase 3) is a cell surface protein expressed in hematopoietic stem and progenitor cell populations1. FLT3 RNA is overexpressed in greater than 95% of acute myeloid leukemia (AML) samples2, and FLT3 mutations are found in 25% to 35% of AML samples1. In 2020 in the United States, an estimated 19,940 new AML cases and 11,180 AML-related deaths are expected3, exemplifying that better therapies are needed to treat this disease. Given the frequency of FLT3 expression in AML and unmet medical need for patients, FLT3 represents an attractive target for T cell-redirecting immunotherapy in AML. FLT3-targeting Tri-specific T cell Activating Constructs (FLT3 TriTACs) are designed to simultaneously engage FLT3 on a target AML cell and CD3 on a T cell, resulting in T cell activation, proliferation, and lysis of the AML cell. FLT3 TriTACs are ~53 kDa molecules that consist of three binding domains: an N-terminal single domain antibody (sdAb) that binds to human FLT3, a middle sdAb that binds to human serum albumin (HSA) to extend the half-life, and a C-terminal single chain Fv (scFv) that binds to CD3e of the T cell receptor (TCR). Biophysical analyses of these molecules indicate they are stable with high monomer content. FLT3 TriTACs bind to human and cynomolgus monkey FLT3, CD3e and albumin with similar affinities. Flow cytometry analysis of T cells from various normal donors and a panel of FLT3-positive and FLT3-negative tumor cell lines confirmed binding of FLT3 TriTACs to their targets expressed on the cell surface. FLT3 TriTACs induce potent killing of FLT3-expressing AML cell lines in vitro. In co-cultures of T cells from normal human donors, target tumor cells, and HSA, these TriTACs mediated dose-dependent and FLT3-dependent cytotoxicity. Cell lines engineered to express cynomolgus FLT3 were also readily killed by T cells with FLT3 TriTACs. Preclinical evaluation of FLT3 TriTACs in vitro, in mouse tumor models, and cynomolgus monkey pharmacokinetic studies will be presented. References 1. Kazi and Ronnstrand. 2019. Physiol Rev. 99:1433-1466. 2. Data generated by the TCGA Research Network: https://www.cancer.gov/tcga. 3. Howlader et al. 2020. SEER Cancer Statistics Review, 1975-2017. Citation Format: Richard J. Austin, Wade H. Aaron, Manasi Barath, Evan Callihan, Scott Gatto, Golzar Hemmati, Che-Leung Law, Bryan D. Lemon, Jessica O9Rear, Purbasa Patnaik, Morgan Thompson, Lihn To, Holger Wesche, Kevin Wright, Yinghua Xiao, Stephen Yu, Timothy Z. Yu. FLT3-targeting TriTACs are T cell engagers for treatment of acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 913.
FLT3 (fms相关受体酪氨酸激酶3)是一种在造血干细胞和祖细胞群中表达的细胞表面蛋白1。FLT3 RNA在超过95%的急性髓性白血病(AML)样本中过表达2,并且在25%至35%的AML样本中发现FLT3突变1。到2020年,美国预计将有19,940例新的AML病例和11180例AML相关死亡,这表明需要更好的治疗方法来治疗这种疾病。考虑到FLT3在AML中的表达频率和患者未满足的医疗需求,FLT3代表了AML中T细胞重定向免疫治疗的一个有吸引力的靶点。FLT3靶向三特异性T细胞激活构建体(FLT3 TriTACs)旨在同时激活靶AML细胞上的FLT3和T细胞上的CD3,导致T细胞活化、增殖和AML细胞裂解。FLT3 TriTACs是一种约53 kDa的分子,由三个结合域组成:与人FLT3结合的n端单链抗体(sdAb),与人血清白蛋白(HSA)结合以延长半衰期的中间sdAb,以及与T细胞受体(TCR) CD3e结合的c端单链Fv (scFv)。这些分子的生物物理分析表明它们是稳定的,单体含量高。TriTACs与人和食蟹猴FLT3、CD3e和白蛋白结合,具有相似的亲和力。流式细胞术分析了来自各种正常供体的T细胞和FLT3阳性和FLT3阴性肿瘤细胞系,证实FLT3 TriTACs与细胞表面表达的靶标结合。FLT3 TriTACs在体外诱导表达FLT3的AML细胞系的有效杀伤。在来自正常人供体的T细胞、靶肿瘤细胞和HSA的共培养中,这些TriTACs介导了剂量依赖性和flt3依赖性的细胞毒性。表达食蟹FLT3的细胞系也很容易被带有FLT3 TriTACs的T细胞杀死。将介绍FLT3 TriTACs在体外、小鼠肿瘤模型和食蟹猴药代动力学研究中的临床前评估。引用1。Kazi和Ronnstrand, 2019。物理启示录99:1433-1466。2. 数据由TCGA研究网络生成:https://www.cancer.gov/tcga。3.Howlader et al. 2020。癌症统计回顾,1975-2017。引文格式:Richard J. Austin, Wade H. Aaron, Manasi Barath, Evan Callihan, Scott Gatto, Golzar Hemmati, Che-Leung Law, Bryan D. Lemon, Jessica O9Rear, Purbasa Patnaik, Morgan Thompson, Lihn To, Holger Wesche, Kevin Wright, Yinghua Xiao, Stephen Yu, Timothy Z. Yu。靶向flt3的TriTACs是治疗急性髓系白血病的T细胞接合物[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):摘要第913期。
{"title":"Abstract 913: FLT3-targeting TriTACs are T cell engagers for treatment of acute myeloid leukemia","authors":"R. Austin, W. Aaron, Manasi Barath, Evan C. Callihan, Scott Gatto, Golzar Hemmati, C. Law, D. LemonBryan, J. O'rear, Purbasa Patnaik, Morgan Thompson, Lihn To, H. Wesche, Kevin Wright, Yinghua Xiao, Stephen Yu, Timothy Z. Yu","doi":"10.1158/1538-7445.AM2021-913","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-913","url":null,"abstract":"FLT3 (fms related receptor tyrosine kinase 3) is a cell surface protein expressed in hematopoietic stem and progenitor cell populations1. FLT3 RNA is overexpressed in greater than 95% of acute myeloid leukemia (AML) samples2, and FLT3 mutations are found in 25% to 35% of AML samples1. In 2020 in the United States, an estimated 19,940 new AML cases and 11,180 AML-related deaths are expected3, exemplifying that better therapies are needed to treat this disease. Given the frequency of FLT3 expression in AML and unmet medical need for patients, FLT3 represents an attractive target for T cell-redirecting immunotherapy in AML. FLT3-targeting Tri-specific T cell Activating Constructs (FLT3 TriTACs) are designed to simultaneously engage FLT3 on a target AML cell and CD3 on a T cell, resulting in T cell activation, proliferation, and lysis of the AML cell. FLT3 TriTACs are ~53 kDa molecules that consist of three binding domains: an N-terminal single domain antibody (sdAb) that binds to human FLT3, a middle sdAb that binds to human serum albumin (HSA) to extend the half-life, and a C-terminal single chain Fv (scFv) that binds to CD3e of the T cell receptor (TCR). Biophysical analyses of these molecules indicate they are stable with high monomer content. FLT3 TriTACs bind to human and cynomolgus monkey FLT3, CD3e and albumin with similar affinities. Flow cytometry analysis of T cells from various normal donors and a panel of FLT3-positive and FLT3-negative tumor cell lines confirmed binding of FLT3 TriTACs to their targets expressed on the cell surface. FLT3 TriTACs induce potent killing of FLT3-expressing AML cell lines in vitro. In co-cultures of T cells from normal human donors, target tumor cells, and HSA, these TriTACs mediated dose-dependent and FLT3-dependent cytotoxicity. Cell lines engineered to express cynomolgus FLT3 were also readily killed by T cells with FLT3 TriTACs. Preclinical evaluation of FLT3 TriTACs in vitro, in mouse tumor models, and cynomolgus monkey pharmacokinetic studies will be presented. References 1. Kazi and Ronnstrand. 2019. Physiol Rev. 99:1433-1466. 2. Data generated by the TCGA Research Network: https://www.cancer.gov/tcga. 3. Howlader et al. 2020. SEER Cancer Statistics Review, 1975-2017. Citation Format: Richard J. Austin, Wade H. Aaron, Manasi Barath, Evan Callihan, Scott Gatto, Golzar Hemmati, Che-Leung Law, Bryan D. Lemon, Jessica O9Rear, Purbasa Patnaik, Morgan Thompson, Lihn To, Holger Wesche, Kevin Wright, Yinghua Xiao, Stephen Yu, Timothy Z. Yu. FLT3-targeting TriTACs are T cell engagers for treatment of acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 913.","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78505586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-988
N. Savaraj, M. You, Ying-Ying Li, Chunjing Wu, M. Wangpaichitr, Saurez Miguel, Alfred Chicco, M. Kuo, L. Feun
{"title":"Abstract 988: Targeting immunological and apoptotic cell death to improve therapeutic efficacy in melanoma","authors":"N. Savaraj, M. You, Ying-Ying Li, Chunjing Wu, M. Wangpaichitr, Saurez Miguel, Alfred Chicco, M. Kuo, L. Feun","doi":"10.1158/1538-7445.AM2021-988","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-988","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72624938","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-52
Beatrice T. Wang, Tasnim Kothambawala, Kevin C. Hart, Xingjie Chen, M. Desbois, Susan E. Calhoun, P. Yakkundi, Rodnie Rosete, Yuan Cao, Katie Cha, T. Matthew, Ling Wang, P. Hinton, M. Leabman, G. Hernandez, M. Kotturi, E. Humke, A. Sinclair, B. Keyt
{"title":"Abstract 52: Mechanistic evaluation of anti-DR5 IgM antibody IGM-8444 with potent tumor cytotoxicity, without in vitro hepatotoxicity","authors":"Beatrice T. Wang, Tasnim Kothambawala, Kevin C. Hart, Xingjie Chen, M. Desbois, Susan E. Calhoun, P. Yakkundi, Rodnie Rosete, Yuan Cao, Katie Cha, T. Matthew, Ling Wang, P. Hinton, M. Leabman, G. Hernandez, M. Kotturi, E. Humke, A. Sinclair, B. Keyt","doi":"10.1158/1538-7445.AM2021-52","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-52","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72649657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1339
Christine C. Hudson, R. Narayanaswamy, Sébastien Ronseaux, C. Bowden, Brandon Nicolay
{"title":"Abstract 1339: Investigations into rational combination approaches with ivosidenib in a mutant IDH1 acute myeloid leukemia patient-derived xenograft model","authors":"Christine C. Hudson, R. Narayanaswamy, Sébastien Ronseaux, C. Bowden, Brandon Nicolay","doi":"10.1158/1538-7445.AM2021-1339","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1339","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77441948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-958
Elizabeth R. Murray, N. Brown, P. Howard, Luke A Masterson, F. Zammarchi, Patrick Van Berkel, J. Marshall
Pancreatic ductal adenocarcinoma (PDAC) exhibits an extremely poor prognosis, with 5 year-survival rates 80% of human PDAC tumors including their paired metastases but is not significantly expressed by healthy pancreas tissue. Thus, αvβ6 represents a promising therapeutic target. We previously developed the αvβ6-specific peptide-drug conjugate (PDC) SG3299, composed of the DNA-binding pyrrolobenzodiazepine (PBD)-based compound tesirine conjugated to the αvβ6-targeting peptide A20FMDV2. Relative to the non-targeting PDC SG3511, SG3299 showed anti-tumor efficacy against αvβ6-positive human PDAC xenografts in immunodeficient mouse models. The present study aimed to evaluate the efficacy of αvβ6-targeted therapy on primary tumors and metastases in an immunocompetent murine model of PDAC. We have previously reported that KPC (Kras-G12D P53-R172H Pdx1-Cre) transgenic mouse PDAC tumors do not express αvβ6, so we ectopically expressed murine integrin-β6 in the TB32043 KPC-derived PDAC cell line, creating the αvβ6-expressing line TB32043mb6S2. When injected orthotopically into immunocompetent C57BL/6 mice, TB32043mb6S2 cells produced primary pancreatic tumors with αvβ6-positive metastases to the liver, peritoneum, and lung. Mice injected orthotopically with TB32043mb6S2 cells exhibited increased primary tumor desmoplasia, increased incidence of peritoneal metastases (100% vs 20%, p Citation Format: Elizabeth R. Murray, Nicholas F. Brown, Philip Howard, Luke Masterson, Francesca Zammarchi, Patrick H. van Berkel, John F. Marshall. Effective targeting of pancreatic ductal adenocarcinoma metastases with an integrin αvβ6-targeting peptide-drug conjugate [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 958.
胰腺导管腺癌(PDAC)预后极差,人类PDAC肿瘤包括其配对转移瘤的5年生存率为80%,但在健康胰腺组织中未显着表达。因此,αvβ6是一个很有前景的治疗靶点。我们之前开发了αvβ6特异性肽-药物偶联物(PDC) SG3299,由结合dna的吡咯苯二氮卓(PBD)为基础的化合物tesirine偶联到αvβ6靶向肽A20FMDV2组成。相对于非靶向PDC SG3511, SG3299在免疫缺陷小鼠模型中对αvβ6阳性的人PDAC异种移植物具有抗肿瘤作用。本研究旨在评价αvβ6靶向治疗小鼠PDAC的原发肿瘤和转移瘤的疗效。我们之前报道过KPC (Kras-G12D P53-R172H Pdx1-Cre)转基因小鼠PDAC肿瘤不表达αvβ6,因此我们在TB32043 KPC衍生的PDAC细胞系中异位表达小鼠整合素-β6,构建了表达αvβ6的细胞系TB32043mb6S2。将TB32043mb6S2细胞原位注射到免疫功能正常的C57BL/6小鼠体内,产生原发性胰腺肿瘤,并伴有αvβ6阳性转移至肝脏、腹膜和肺部。原位注射TB32043mb6S2细胞的小鼠显示原发性肿瘤结缔组织增生增加,腹膜转移发生率增加(100% vs 20%), p引用格式:Elizabeth R. Murray, Nicholas F. Brown, Philip Howard, Luke Masterson, Francesca Zammarchi, Patrick H. van Berkel, John F. Marshall。整合素αvβ6靶向肽-药物偶联物有效靶向胰腺导管腺癌转移瘤[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):摘要第958期。
{"title":"Abstract 958: Effective targeting of pancreatic ductal adenocarcinoma metastases with an integrin αvβ6-targeting peptide-drug conjugate","authors":"Elizabeth R. Murray, N. Brown, P. Howard, Luke A Masterson, F. Zammarchi, Patrick Van Berkel, J. Marshall","doi":"10.1158/1538-7445.AM2021-958","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-958","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) exhibits an extremely poor prognosis, with 5 year-survival rates 80% of human PDAC tumors including their paired metastases but is not significantly expressed by healthy pancreas tissue. Thus, αvβ6 represents a promising therapeutic target. We previously developed the αvβ6-specific peptide-drug conjugate (PDC) SG3299, composed of the DNA-binding pyrrolobenzodiazepine (PBD)-based compound tesirine conjugated to the αvβ6-targeting peptide A20FMDV2. Relative to the non-targeting PDC SG3511, SG3299 showed anti-tumor efficacy against αvβ6-positive human PDAC xenografts in immunodeficient mouse models. The present study aimed to evaluate the efficacy of αvβ6-targeted therapy on primary tumors and metastases in an immunocompetent murine model of PDAC. We have previously reported that KPC (Kras-G12D P53-R172H Pdx1-Cre) transgenic mouse PDAC tumors do not express αvβ6, so we ectopically expressed murine integrin-β6 in the TB32043 KPC-derived PDAC cell line, creating the αvβ6-expressing line TB32043mb6S2. When injected orthotopically into immunocompetent C57BL/6 mice, TB32043mb6S2 cells produced primary pancreatic tumors with αvβ6-positive metastases to the liver, peritoneum, and lung. Mice injected orthotopically with TB32043mb6S2 cells exhibited increased primary tumor desmoplasia, increased incidence of peritoneal metastases (100% vs 20%, p Citation Format: Elizabeth R. Murray, Nicholas F. Brown, Philip Howard, Luke Masterson, Francesca Zammarchi, Patrick H. van Berkel, John F. Marshall. Effective targeting of pancreatic ductal adenocarcinoma metastases with an integrin αvβ6-targeting peptide-drug conjugate [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 958.","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77549055","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-LB118
C. Klaus, S. Rusin, K. Sharma, S. Bhaduri, M. Weiss, A. Mcdonald, M. Mayo, D. Walker, Rahul Karnik
Relapsed/refractory DLBCL remains an incurable disease, and single-agent therapies typically show low response rates and/or transient clinical responses. Oncogenic MYD88 mutations occur in ~25% of DLBCL and drive constitutive NFkB activation, promoting proliferation and survival. Despite its role in tumor biology, targeting MYD88MT by inhibiting or degrading IRAK4 alone, a key component of the MYD88 complex, does not drive significant antitumor activity in preclinical models. One potential reason is the frequent redundant NFkB pathway activation by co-mutations, highlighting the need for combination therapies to effectively target the NFkB pathway in DLBCL. To address this challenge, we have developed IRAKIMiDs, heterobifunctional degraders that simultaneously degrade both IRAK4 and IMiD substrates, as a rational therapeutic combination in a single molecule. IRAKIMiDs show increased antitumor activity in vitro and in vivo in MYD88MT cells as compared to IMiD or IRAK4-targeting alone, highlighting suggesting their potential as single agents in R/R DLBCL.Our lead IRAKIMiD, KT-413, is a potent degrader of IRAK4 (DC50 6nM) and IMiD substrates (Ikaros/Aiolos DC50 2nM), inducing rapid and potent cell killing in vitro and complete and sustained tumor regressions in vivo in MYD88MT models of DLBCL. This activity is superior to the IMiD CC-220, which has similar activity against IMiD substrates (Ikaros/Aiolos DC50 1 nM), supporting the synergistic role of IRAK4 degradation in the context of IMiD biology. We show here that the combined activity of these 2 mechanisms drives a synergistic effect on NFkB and IRF4 signaling with greater downstream effect on NFkB and type 1 interferon (IFN) signaling and cell cycle gene expression than either mechanism alone. In THP1 cells engineered with NFkB and IRF4 reporters, KT-413 but not CC-220 inhibits TLR-stimulated NFkB and IRF4 transcription, supporting a role for IRAK4 but not IMiDs in MYD88-driven survival and proliferation signals. IMiDs have previously been shown to modulate type1 IFN signaling through downregulation of IRF4. We propose that simultaneous targeting of both NFkB and type 1 IFN signaling with KT-413 drives synergistic cell killing in MYD88MT cells. In MYD88MT OCI-Ly10 cells, KT-413 leads to greater IRF4 downregulation, increased type 1 IFN signaling, and preferential downregulation of NFkB pathway and cell cycle transcripts when compared to CC-220. These data support the hypothesis that the synergistic activity of targeting IRAK4 and IMiD substrates by KT-413 in MYD88MT DLBCL is a result of dual targeting of NFkB and IRF4/Type1 IFN through degradation of both IRAK4 and IMiD substrates,driving significantly greater cell killing as compared to either mechanism alone, supporting the potential for the first single agent targeted therapy in MYD88MT DLBCL. KT-413 is on track for initiation of a Phase 1 trial in B cell lymphoma in 2H 2021. Citation Format: Christine R. Klaus, Scott F. Rusin, Kirti Sharma,
复发/难治性DLBCL仍然是一种无法治愈的疾病,单药治疗通常表现出低缓解率和/或短暂的临床反应。致瘤性MYD88突变发生在约25%的DLBCL中,并驱动构成性NFkB激活,促进增殖和生存。尽管MYD88MT在肿瘤生物学中发挥着重要作用,但在临床前模型中,通过抑制或降解MYD88复合物的关键成分IRAK4来靶向MYD88MT并没有显著的抗肿瘤活性。一个潜在的原因是NFkB通路通过共突变频繁激活,因此需要联合治疗来有效靶向DLBCL中的NFkB通路。为了应对这一挑战,我们开发了IRAKIMiDs,这是一种异质双功能降解剂,可以同时降解IRAK4和IMiD底物,作为一种合理的单分子治疗组合。与单独靶向IMiD或irak4相比,IRAKIMiDs在体外和体内对MYD88MT细胞显示出更高的抗肿瘤活性,突出表明它们作为R/R DLBCL的单一药物的潜力。我们的主要IRAKIMiD KT-413是IRAK4 (DC50 6nM)和IMiD底物(Ikaros/Aiolos DC50 2nM)的有效降解剂,在体外诱导快速有效的细胞杀伤,在MYD88MT DLBCL模型中诱导完全和持续的肿瘤消退。该活性优于IMiD CC-220,后者对IMiD底物(Ikaros/Aiolos DC50 1 nM)具有相似的活性,支持IRAK4在IMiD生物学背景下的协同降解作用。我们在这里表明,这两种机制的联合活性对NFkB和IRF4信号传导产生协同作用,对NFkB和1型干扰素(IFN)信号传导和细胞周期基因表达的下游影响比单独的任何一种机制都要大。在用NFkB和IRF4报告基因工程的THP1细胞中,KT-413而不是CC-220抑制tlr刺激的NFkB和IRF4转录,支持IRAK4而不是IMiDs在myd88驱动的存活和增殖信号中的作用。先前已证明IMiDs通过下调IRF4来调节1型IFN信号。我们提出,KT-413同时靶向NFkB和1型IFN信号可驱动MYD88MT细胞的协同细胞杀伤。在MYD88MT OCI-Ly10细胞中,与CC-220相比,KT-413导致更大的IRF4下调,1型IFN信号传导增加,NFkB通路和细胞周期转录物优先下调。这些数据支持了这样的假设,即在MYD88MT DLBCL中,KT-413靶向IRAK4和IMiD底物的协同活性是通过IRAK4和IMiD底物的降解双重靶向NFkB和IRF4/ 1型IFN的结果,与单独的任何一种机制相比,驱动更大的细胞杀伤,支持了MYD88MT DLBCL中首个单药靶向治疗的潜力。KT-413有望在2021年下半年启动B细胞淋巴瘤的1期临床试验。引文格式:Christine R. Klaus, Scott F. Rusin, Kirti Sharma, Samyabrata Bhaduri, Matthew M. Weiss, Alice A. McDonald, Michele F. Mayo, Duncan Walker, Rahul Karnik。KT-413 (IRAK4和IMiD底物的靶向降解物)在MYD88MTDLBCL中协同作用的机制[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):摘要nr LB118。
{"title":"Abstract LB118: Mechanisms underlying synergistic activity in MYD88MTDLBCL of KT-413, a targeted degrader of IRAK4 and IMiD substrate","authors":"C. Klaus, S. Rusin, K. Sharma, S. Bhaduri, M. Weiss, A. Mcdonald, M. Mayo, D. Walker, Rahul Karnik","doi":"10.1158/1538-7445.AM2021-LB118","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-LB118","url":null,"abstract":"Relapsed/refractory DLBCL remains an incurable disease, and single-agent therapies typically show low response rates and/or transient clinical responses. Oncogenic MYD88 mutations occur in ~25% of DLBCL and drive constitutive NFkB activation, promoting proliferation and survival. Despite its role in tumor biology, targeting MYD88MT by inhibiting or degrading IRAK4 alone, a key component of the MYD88 complex, does not drive significant antitumor activity in preclinical models. One potential reason is the frequent redundant NFkB pathway activation by co-mutations, highlighting the need for combination therapies to effectively target the NFkB pathway in DLBCL. To address this challenge, we have developed IRAKIMiDs, heterobifunctional degraders that simultaneously degrade both IRAK4 and IMiD substrates, as a rational therapeutic combination in a single molecule. IRAKIMiDs show increased antitumor activity in vitro and in vivo in MYD88MT cells as compared to IMiD or IRAK4-targeting alone, highlighting suggesting their potential as single agents in R/R DLBCL.Our lead IRAKIMiD, KT-413, is a potent degrader of IRAK4 (DC50 6nM) and IMiD substrates (Ikaros/Aiolos DC50 2nM), inducing rapid and potent cell killing in vitro and complete and sustained tumor regressions in vivo in MYD88MT models of DLBCL. This activity is superior to the IMiD CC-220, which has similar activity against IMiD substrates (Ikaros/Aiolos DC50 1 nM), supporting the synergistic role of IRAK4 degradation in the context of IMiD biology. We show here that the combined activity of these 2 mechanisms drives a synergistic effect on NFkB and IRF4 signaling with greater downstream effect on NFkB and type 1 interferon (IFN) signaling and cell cycle gene expression than either mechanism alone. In THP1 cells engineered with NFkB and IRF4 reporters, KT-413 but not CC-220 inhibits TLR-stimulated NFkB and IRF4 transcription, supporting a role for IRAK4 but not IMiDs in MYD88-driven survival and proliferation signals. IMiDs have previously been shown to modulate type1 IFN signaling through downregulation of IRF4. We propose that simultaneous targeting of both NFkB and type 1 IFN signaling with KT-413 drives synergistic cell killing in MYD88MT cells. In MYD88MT OCI-Ly10 cells, KT-413 leads to greater IRF4 downregulation, increased type 1 IFN signaling, and preferential downregulation of NFkB pathway and cell cycle transcripts when compared to CC-220. These data support the hypothesis that the synergistic activity of targeting IRAK4 and IMiD substrates by KT-413 in MYD88MT DLBCL is a result of dual targeting of NFkB and IRF4/Type1 IFN through degradation of both IRAK4 and IMiD substrates,driving significantly greater cell killing as compared to either mechanism alone, supporting the potential for the first single agent targeted therapy in MYD88MT DLBCL. KT-413 is on track for initiation of a Phase 1 trial in B cell lymphoma in 2H 2021. Citation Format: Christine R. Klaus, Scott F. Rusin, Kirti Sharma, ","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77729654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1293
K. Lemberg, J. M. H. Aguilar, J. Alt, M. Krečmerová, P. Majer, R. Rais, B. Slusher
{"title":"Abstract 1293: A novel protide purine antimetabolite combines with the prodrug glutamine antagonist JHU395 in preclinical models of Ras-driven sarcomas","authors":"K. Lemberg, J. M. H. Aguilar, J. Alt, M. Krečmerová, P. Majer, R. Rais, B. Slusher","doi":"10.1158/1538-7445.AM2021-1293","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1293","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80037758","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1461
J. Han, Kyu‐Tae Kim, Jeejin Im, Sojung Park, M. Choi, Inki Kim, Ky-Youb Nam, Jeong-Hyeok Yoon
Purpose: Development of the potent and selective checkpoint kinase 2 (CHK2) inhibitor to overcome limiting clinical utility of poly(ADP-ribose) polymerase 1 (PARP1) inhibitors. Experimental Procedure: Preclinical evaluation of PHI-101 for cellular and molecular potency in ovarian and breast cancer cell lines and patient derived primary cells; it includes biochemical binding assay, cellular assays, animal efficacy studies, combination study, signaling pathway effect examination, and cell cycle analysis. Summary: CHK2 is a serine/threonine kinase and a cell cycle checkpoint regulator involved in the ATM-mediated DNA repair upon replication blocks and DNA damage. It has been proposed that Chk2 functions as a barrier to tumorigenesis by maintaining genomic stability, and this DNA damage induction is thought to prevent or delay genetic instability and tumorigenesis. The inhibition of CHK2 is a promising approach to achieve synthetic lethality of cancer cells when combined with PARP1 inhibitors. Solid cancer indications of PHI-101 was identified by the Chemiverse Network module which is an AI and Big data-based in-house drug discovery platform. Biochemical kinase assays for PHI-101 showed stronger affinity to CHK2 over CHK1 more than 5-fold. PHI-101 treatment of ovarian and breast cancer cells for 72 hrs elicit a synergistic lethal response in combination with PARP1 inhibitor Olaparib regardless of functional BRCA and P53 in the cells. PHI-101 also potentiates a countermeasure to dose-limiting toxicity triggered by genotoxic agents such as cisplatin and topotecan. The present results from in vivo and in vitro preclinical testing do demonstrate that PHI-101 is a highly potent inhibitor of CHK2 and may exert mono- and combinational therapeutic activity in ovarian and breast cancer model. Conclusion: The preclinical evaluation of PHI-101, a novel CHK2 inhibitor, showed clear evidence of anticancer activity for refractory ovarian and breast cancer cells and improved efficacy in both in vitro and in vivo models. Consequently, PHI-101 is currently under investigation in Phase 1 clinical trials for relapsed or refractory ovarian cancer patients. Citation Format: June H-J Han, Kyu-Tae Kim, Jeejin Im, Sojung Park, Min Kyung Choi, Inki Kim, Ky-Youb Nam, JeongHyeok Yoon. PHI-101, a potent and novel inhibitor of CHK2 in ovarian and breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1461.
目的:开发有效的选择性检查点激酶2 (CHK2)抑制剂,以克服聚(adp -核糖)聚合酶1 (PARP1)抑制剂的局限性临床应用。实验程序:临床前评估PHI-101在卵巢癌和乳腺癌细胞系和患者源性原代细胞中的细胞和分子效力;它包括生化结合试验、细胞试验、动物功效研究、联合研究、信号通路效应检查和细胞周期分析。摘要:CHK2是一种丝氨酸/苏氨酸激酶和细胞周期检查点调节因子,参与atm介导的DNA复制阻滞和DNA损伤修复。已经提出Chk2通过维持基因组稳定性作为肿瘤发生的屏障,并且这种DNA损伤诱导被认为可以防止或延迟遗传不稳定性和肿瘤发生。当与PARP1抑制剂联合使用时,抑制CHK2是一种很有希望实现癌细胞合成致死的方法。phi101的实体癌适应症是由chemverse Network模块确定的,该模块是一个基于人工智能和大数据的内部药物发现平台。生化激酶实验表明,ph -101对CHK2的亲和力比CHK1强5倍以上。无论细胞中BRCA和P53的功能如何,PHI-101与PARP1抑制剂奥拉帕尼联合治疗卵巢癌和乳腺癌细胞72小时可引起协同致死反应。pi -101还增强了对基因毒性药物如顺铂和拓扑替康引发的剂量限制性毒性的对策。目前的体内和体外临床前试验结果表明,PHI-101是一种高效的CHK2抑制剂,可能在卵巢癌和乳腺癌模型中发挥单一和联合治疗活性。结论:新型CHK2抑制剂PHI-101对难治性卵巢癌和乳腺癌细胞具有明显的抗肿瘤活性,在体外和体内模型中均有提高。因此,PHI-101目前正处于用于复发或难治性卵巢癌患者的i期临床试验研究中。引文格式:June H-J Han, Kyu-Tae Kim, Jeejin Im, Sojung Park, Min Kyung Choi, Inki Kim, Ky-Youb Nam, JeongHyeok YoonPHI-101,卵巢癌和乳腺癌细胞中一种有效的新型CHK2抑制剂[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):摘要nr 1461。
{"title":"Abstract 1461: PHI-101, a potent and novel inhibitor of CHK2 in ovarian and breast cancer cells","authors":"J. Han, Kyu‐Tae Kim, Jeejin Im, Sojung Park, M. Choi, Inki Kim, Ky-Youb Nam, Jeong-Hyeok Yoon","doi":"10.1158/1538-7445.AM2021-1461","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1461","url":null,"abstract":"Purpose: Development of the potent and selective checkpoint kinase 2 (CHK2) inhibitor to overcome limiting clinical utility of poly(ADP-ribose) polymerase 1 (PARP1) inhibitors. Experimental Procedure: Preclinical evaluation of PHI-101 for cellular and molecular potency in ovarian and breast cancer cell lines and patient derived primary cells; it includes biochemical binding assay, cellular assays, animal efficacy studies, combination study, signaling pathway effect examination, and cell cycle analysis. Summary: CHK2 is a serine/threonine kinase and a cell cycle checkpoint regulator involved in the ATM-mediated DNA repair upon replication blocks and DNA damage. It has been proposed that Chk2 functions as a barrier to tumorigenesis by maintaining genomic stability, and this DNA damage induction is thought to prevent or delay genetic instability and tumorigenesis. The inhibition of CHK2 is a promising approach to achieve synthetic lethality of cancer cells when combined with PARP1 inhibitors. Solid cancer indications of PHI-101 was identified by the Chemiverse Network module which is an AI and Big data-based in-house drug discovery platform. Biochemical kinase assays for PHI-101 showed stronger affinity to CHK2 over CHK1 more than 5-fold. PHI-101 treatment of ovarian and breast cancer cells for 72 hrs elicit a synergistic lethal response in combination with PARP1 inhibitor Olaparib regardless of functional BRCA and P53 in the cells. PHI-101 also potentiates a countermeasure to dose-limiting toxicity triggered by genotoxic agents such as cisplatin and topotecan. The present results from in vivo and in vitro preclinical testing do demonstrate that PHI-101 is a highly potent inhibitor of CHK2 and may exert mono- and combinational therapeutic activity in ovarian and breast cancer model. Conclusion: The preclinical evaluation of PHI-101, a novel CHK2 inhibitor, showed clear evidence of anticancer activity for refractory ovarian and breast cancer cells and improved efficacy in both in vitro and in vivo models. Consequently, PHI-101 is currently under investigation in Phase 1 clinical trials for relapsed or refractory ovarian cancer patients. Citation Format: June H-J Han, Kyu-Tae Kim, Jeejin Im, Sojung Park, Min Kyung Choi, Inki Kim, Ky-Youb Nam, JeongHyeok Yoon. PHI-101, a potent and novel inhibitor of CHK2 in ovarian and breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1461.","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80237120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}