Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-404
M. Oshi, L. Le, Y. Tokumaru, A. Patel, R. Matsuyama, I. Endo, Li Yan, M. Katz, K. Takabe
BACKGROUND: Pancreatic cancer is one of the most aggressive cancers with a 5-year survival of less than 10%. Surgery is the only cure for pancreatic cancer but the majority of patients present with metastatic disease at the time of diagnosis. Systemic chemotherapy remains the primary treatment option. Given the insidious clinical presentation and poor treatment response, there is an urgent need to discover a predictive biomarker for appropriate patient selection. We hypothesized that the 4-gene score, which reflects tumor cell proliferation, is both a prognostic and predictive biomarker for pancreatic cancer. METHODS: The 4-gene score is derived from tumor expression of DOK4, HCCS, PGF, and SHCBP1 genes, which were identified based on differential mRNA expression analysis of a human breast cell line, its metastatic variant cells, and clinical outcome data of breast cancer patient cohorts, as previously reported. A total of 954 pancreatic cancer patients were analyzed for both discovery and validation of the 4-gene score from publicly available datasets to investigate the relationship between the score with clinical features such as metastasis, cancer aggressiveness, immune cell infiltration, patient survival, and resectability. RESULTS: We found that the 4-gene score correlation in pancreatic cancer was higher than in breast cancer cohorts, specifically in clinically aggressive parameters such as pathological grade and MKI67 expression. Also, the score in metastatic tumor cohorts was higher than in primary cancer cohorts (p CONCLUSION: The 4-gene score identified poor survival in pancreatic cancer and has potential as a predictive biomarker for R0 resection and treatment response in metastatic pancreatic cancer. Evaluating the degree of the 4-gene score could be a valuable potential prognostic tool. Citation Format: Masanori Oshi, Lan Le, Yoshihisa Tokumaru, Ankit Patel, Ryusei Matsuyama, Itaru Endo, Li Yan, Matthew H.G. Katz, Kazuaki Takabe. A novel 4-gene score predict patient survival as well as pathologically complete (R0) resection in pancreatic cancer [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 404.
背景:胰腺癌是最具侵袭性的癌症之一,5年生存率低于10%。手术是治疗胰腺癌的唯一方法,但大多数患者在诊断时已出现转移性疾病。全身化疗仍然是主要的治疗选择。鉴于隐匿的临床表现和不良的治疗反应,迫切需要发现一种预测性的生物标志物,以适当地选择患者。我们假设反映肿瘤细胞增殖的4基因评分是胰腺癌的预后和预测性生物标志物。方法:4基因评分来源于肿瘤中DOK4、HCCS、PGF和SHCBP1基因的表达,这些基因是根据人类乳腺细胞系及其转移变异细胞的差异mRNA表达分析和乳腺癌患者队列的临床结局数据确定的,如先前报道的那样。我们对954例胰腺癌患者进行了分析,从公开数据集中发现并验证了4基因评分,以探讨该评分与转移、肿瘤侵袭性、免疫细胞浸润、患者生存和可切除性等临床特征之间的关系。结果:我们发现胰腺癌的4基因评分相关性高于乳腺癌队列,特别是在临床侵袭性参数如病理分级和MKI67表达方面。此外,转移性肿瘤组的评分高于原发癌组(p结论:4基因评分可识别胰腺癌患者的不良生存率,并有可能作为转移性胰腺癌R0切除术和治疗反应的预测性生物标志物。评估4基因评分的程度可能是一种有价值的潜在预后工具。引文格式:Masanori Oshi, Lan Le, Yoshihisa Tokumaru, Ankit Patel, Ryusei Matsuyama, Itaru Endo, Li Yan, Matthew H.G. Katz, Kazuaki Takabe。一种新的4基因评分预测胰腺癌患者的生存和病理完全切除(R0)[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404页。
{"title":"Abstract 404: A novel 4-gene score predict patient survival as well as pathologically complete (R0) resection in pancreatic cancer","authors":"M. Oshi, L. Le, Y. Tokumaru, A. Patel, R. Matsuyama, I. Endo, Li Yan, M. Katz, K. Takabe","doi":"10.1158/1538-7445.AM2021-404","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-404","url":null,"abstract":"BACKGROUND: Pancreatic cancer is one of the most aggressive cancers with a 5-year survival of less than 10%. Surgery is the only cure for pancreatic cancer but the majority of patients present with metastatic disease at the time of diagnosis. Systemic chemotherapy remains the primary treatment option. Given the insidious clinical presentation and poor treatment response, there is an urgent need to discover a predictive biomarker for appropriate patient selection. We hypothesized that the 4-gene score, which reflects tumor cell proliferation, is both a prognostic and predictive biomarker for pancreatic cancer. METHODS: The 4-gene score is derived from tumor expression of DOK4, HCCS, PGF, and SHCBP1 genes, which were identified based on differential mRNA expression analysis of a human breast cell line, its metastatic variant cells, and clinical outcome data of breast cancer patient cohorts, as previously reported. A total of 954 pancreatic cancer patients were analyzed for both discovery and validation of the 4-gene score from publicly available datasets to investigate the relationship between the score with clinical features such as metastasis, cancer aggressiveness, immune cell infiltration, patient survival, and resectability. RESULTS: We found that the 4-gene score correlation in pancreatic cancer was higher than in breast cancer cohorts, specifically in clinically aggressive parameters such as pathological grade and MKI67 expression. Also, the score in metastatic tumor cohorts was higher than in primary cancer cohorts (p CONCLUSION: The 4-gene score identified poor survival in pancreatic cancer and has potential as a predictive biomarker for R0 resection and treatment response in metastatic pancreatic cancer. Evaluating the degree of the 4-gene score could be a valuable potential prognostic tool. Citation Format: Masanori Oshi, Lan Le, Yoshihisa Tokumaru, Ankit Patel, Ryusei Matsuyama, Itaru Endo, Li Yan, Matthew H.G. Katz, Kazuaki Takabe. A novel 4-gene score predict patient survival as well as pathologically complete (R0) resection in pancreatic cancer [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 404.","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"104 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86966296","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.AM2021-520
M. Nam, L. Kim, William Cheng, W. Bae, J. Hwang, Y. Choi, Yeun Ho Lee, W. K. Hur, C. Jung, Heayoon S. Cho, Y. Chae
{"title":"Abstract 520: Potential role of serum proteome in predicting immune-related adverse events from immunotherapy in non-small cell lung cancer","authors":"M. Nam, L. Kim, William Cheng, W. Bae, J. Hwang, Y. Choi, Yeun Ho Lee, W. K. Hur, C. Jung, Heayoon S. Cho, Y. Chae","doi":"10.1158/1538-7445.AM2021-520","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-520","url":null,"abstract":"","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"31 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87066005","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.AM2021-354
H. Biesma, A.T.T.D. Soeratram, K. Sikorska, I. Caspers, H. F. Essen, J. Egthuijsen, A. Mookhoek, D. Hoek, W. Vos, H. Laarhoven, M. Nordsmark, D. L. Peet, F. Warmerdam, M. Geenen, O. Loosveld, J. Portielje, M. Los, E. Kranenbarg, H. Hartgrink, J. Sandick, C. V. D. Velde, M. Verheij, A. Cats, B. Ylstra, N. Grieken
Background: Epstein-Barr virus positivity (EBV+) and microsatellite instability (MSI-high) have been shown to be positive prognostic factors for long term survival in resectable gastric cancer (GC) in several studies. However, the benefit of perioperative treatment in patients with MSI-high tumors remains topic of discussion. Here, we present the clinicopathological outcome of patients with EBV+ and MSI-high GCs treated with surgery only in the Dutch D1/D2 trial, and treated with chemotherapy or chemoradiotherapy after preoperative chemotherapy and surgery in the CRITICS trial. Patients and methods: EBV was determined in tumor tissue using EBV-encoded RNA in situ hybridization (EBER-ISH). PCR and/or immunohistochemistry were performed to determine MSI status. Results were correlated to histopathological response, morphological tumor characteristics and survival. Results: In the Dutch D1/D2 trial 10.5% (47/447) of tumors were EBV+ and 10.5% (47/447) were MSI-high. In the CRITICS trial 5.5% (25/451) of tumors were EBV+ and 5.5% (25/451) were MSI-high tumors. In the Dutch D1/D2 trial, five-year overall survival probability was 51.1% for EBV+, 46.8% for MSI-high, and 42.5% for EBV-/MSS (P=0.19). In the CRITICS trial, five-year overall survival was 56.0% for EBV+, 47.3% for MSI-high, and 36.5% for EBV-/MSS (P=0.22). In the CRITICS trial, 3 (12.5%) MSI-high tumors showed moderate to complete histopathological response. Interestingly, all three showed a mucinous phenotype. Eight (36.4%) EBV+ and 114 (29.9%) EBV-/MSS tumors showed moderate to complete histopathological response. None of the EBV+ GCs showed mucinous differentiation. Conclusions: The favorable outcome of GC patients with resectable EBV+ or MSI-high tumors compared to EBV-/MSS tumors remains after perioperative chemotherapy. In MSI-high tumors significant histopathological response to neoadjuvant chemotherapy was found only in those with a mucinous phenotype. Citation Format: H.D. Biesma, A.T.T.D. Soeratram, K. Sikorska, I.A. Caspers, H.F. van Essen, J.M.P. Egthuijsen, A. Mookhoek, D.M. Hoek, W. Vos, H.W.M. van Laarhoven, M. Nordsmark, D.L. van der Peet, F.A.R.M. Warmerdam, M.M. Geenen, O.J.L. Loosveld, J.E.A. Portielje, M. Los, E. Meershoek - Klein Kranenbarg, H.H. Hartgrink, J. van Sandick, C.J.H. van de Velde, M. Verheij, A. Cats, B. Ylstra, N.C.T. van Grieken, On behalf of the CRITICS investigators. Mucinous phenotype is associated with response to neoadjuvant chemotherapy in microsatellite instable resectable gastric cancer [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 354.
背景:Epstein-Barr病毒阳性(EBV+)和微卫星不稳定性(MSI-high)在一些研究中已被证明是可切除胃癌(GC)长期生存的积极预后因素。然而,高msi肿瘤患者围手术期治疗的益处仍然是讨论的主题。在此,我们介绍了荷兰D1/D2试验中EBV+和msi高GCs患者仅接受手术治疗,而在CRITICS试验中术前化疗和手术后接受化疗或放化疗的临床病理结果。患者和方法:采用EBV编码RNA原位杂交法(EBER-ISH)检测EBV在肿瘤组织中的表达。采用PCR和/或免疫组织化学检测MSI状态。结果与组织病理反应、肿瘤形态特征及生存率相关。结果:在荷兰D1/D2试验中,10.5%(47/447)的肿瘤为EBV+, 10.5%(47/447)的肿瘤为msi高。在CRITICS试验中,5.5%(25/451)的肿瘤为EBV+, 5.5%(25/451)为msi高肿瘤。在荷兰D1/D2试验中,EBV+组的5年总生存率为51.1%,MSI-high组为46.8%,EBV-/MSS组为42.5% (P=0.19)。在CRITICS试验中,EBV+组的5年总生存率为56.0%,MSI-high组为47.3%,EBV-/MSS组为36.5% (P=0.22)。在CRITICS试验中,3例(12.5%)msi高的肿瘤表现出中度到完全的组织病理反应。有趣的是,这三个人都表现出粘液表型。8例EBV+肿瘤(36.4%)和114例EBV-/MSS肿瘤(29.9%)表现出中度至完全的组织病理反应。EBV+ GCs均未出现粘液分化。结论:与EBV-/MSS肿瘤相比,可切除的EBV+或msi -高肿瘤胃癌患者在围手术期化疗后仍具有良好的预后。在msi高的肿瘤中,只有黏液表型的肿瘤才对新辅助化疗有显著的组织病理学反应。引文格式:H.D. Biesma, A.T.T.D. Soeratram, K. Sikorska, I.A. Caspers, H.F. van Essen, J.M.P. Egthuijsen, A. Mookhoek, D.M. Hoek, W. Vos, H.W.M. van Laarhoven, M. Nordsmark, D.L. van der Peet, F.A.R.M. Warmerdam, M.M. Geenen, O.J.L. Loosveld, J.E.A. Portielje, M. Los, E. Meershoek - Klein Kranenbarg, H.H. Hartgrink, J. van Sandick, C.J.H. van de Velde, M. Verheij, A. Cats, B. Ylstra, N.C.T. van Grieken,代表评论家调查人员。黏液表型与微卫星不稳定可切除胃癌对新辅助化疗的反应有关[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354期。
{"title":"Abstract 354: Mucinous phenotype is associated with response to neoadjuvant chemotherapy in microsatellite instable resectable gastric cancer","authors":"H. Biesma, A.T.T.D. Soeratram, K. Sikorska, I. Caspers, H. F. Essen, J. Egthuijsen, A. Mookhoek, D. Hoek, W. Vos, H. Laarhoven, M. Nordsmark, D. L. Peet, F. Warmerdam, M. Geenen, O. Loosveld, J. Portielje, M. Los, E. Kranenbarg, H. Hartgrink, J. Sandick, C. V. D. Velde, M. Verheij, A. Cats, B. Ylstra, N. Grieken","doi":"10.1158/1538-7445.AM2021-354","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-354","url":null,"abstract":"Background: Epstein-Barr virus positivity (EBV+) and microsatellite instability (MSI-high) have been shown to be positive prognostic factors for long term survival in resectable gastric cancer (GC) in several studies. However, the benefit of perioperative treatment in patients with MSI-high tumors remains topic of discussion. Here, we present the clinicopathological outcome of patients with EBV+ and MSI-high GCs treated with surgery only in the Dutch D1/D2 trial, and treated with chemotherapy or chemoradiotherapy after preoperative chemotherapy and surgery in the CRITICS trial. Patients and methods: EBV was determined in tumor tissue using EBV-encoded RNA in situ hybridization (EBER-ISH). PCR and/or immunohistochemistry were performed to determine MSI status. Results were correlated to histopathological response, morphological tumor characteristics and survival. Results: In the Dutch D1/D2 trial 10.5% (47/447) of tumors were EBV+ and 10.5% (47/447) were MSI-high. In the CRITICS trial 5.5% (25/451) of tumors were EBV+ and 5.5% (25/451) were MSI-high tumors. In the Dutch D1/D2 trial, five-year overall survival probability was 51.1% for EBV+, 46.8% for MSI-high, and 42.5% for EBV-/MSS (P=0.19). In the CRITICS trial, five-year overall survival was 56.0% for EBV+, 47.3% for MSI-high, and 36.5% for EBV-/MSS (P=0.22). In the CRITICS trial, 3 (12.5%) MSI-high tumors showed moderate to complete histopathological response. Interestingly, all three showed a mucinous phenotype. Eight (36.4%) EBV+ and 114 (29.9%) EBV-/MSS tumors showed moderate to complete histopathological response. None of the EBV+ GCs showed mucinous differentiation. Conclusions: The favorable outcome of GC patients with resectable EBV+ or MSI-high tumors compared to EBV-/MSS tumors remains after perioperative chemotherapy. In MSI-high tumors significant histopathological response to neoadjuvant chemotherapy was found only in those with a mucinous phenotype. Citation Format: H.D. Biesma, A.T.T.D. Soeratram, K. Sikorska, I.A. Caspers, H.F. van Essen, J.M.P. Egthuijsen, A. Mookhoek, D.M. Hoek, W. Vos, H.W.M. van Laarhoven, M. Nordsmark, D.L. van der Peet, F.A.R.M. Warmerdam, M.M. Geenen, O.J.L. Loosveld, J.E.A. Portielje, M. Los, E. Meershoek - Klein Kranenbarg, H.H. Hartgrink, J. van Sandick, C.J.H. van de Velde, M. Verheij, A. Cats, B. Ylstra, N.C.T. van Grieken, On behalf of the CRITICS investigators. Mucinous phenotype is associated with response to neoadjuvant chemotherapy in microsatellite instable resectable gastric cancer [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 354.","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"6 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90149471","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.AM2021-633
Fan Yang, Samuel W. Brady, Huiying Sun, Chao Tang, L. Du, M. Barz, Xiaotu Ma, Yao Chen, Houshun Fang, Xiaomeng Li, Pandurang Kolekar, Omkar Pathak, J. Cai, Lixia Ding, Tianyi Wang, A. Stackelberg, S. Shen, C. Duan, C. Eckert, Hongzhuan Chen, Yu Liu, J. Klco, Hui Li, Ben-shang Li, Jinghui Zhang, R. Kirschner-Schwabe, Bin-Bing S. Zhou
Chemotherapy is curative for most children with acute lymphoblastic leukemia (ALL). Here we provide direct evidence that thiopurine chemotherapeutics can also directly induce drug resistance mutations leading to relapse. Using a large relapsed ALL cohort assembled from Chinese, US and German patients, we found that TP53 R248Q mutations were highly enriched at relapse compared to diagnosis. Relapse-specific TP53 R248Q was associated with the acquisition of MMR deficiency mutations in MSH2, MSH6, or PMS2 and a novel relapse-specific mutational signature. Using isogenic MCF10A cells with or without engineered MSH2 knockout, and the Nalm6 ALL cell line which has native MMR deficiency, we found that this novel signature was caused by a synergistic mutagenic interaction between thiopurine treatment and mismatch repair (MMR) deficiency (called the thio-dMMR signature) that contributes to a hypermutator phenotype and acquisition of TP53 R248Q in residual ALL during remission. Treatment-induced TP53-mutant clones then expand due to broad chemoresistance, leading to eventual relapse. Indeed, thiopurines preferentially induced C>T mutations at the center of NCG trinucleotides, which can lead to TP53 R248Q, and the thiopurine mutation rate was accelerated 2- to 10-fold in MMR-deficient ALL and cell lines. Thiopurine treatment induced C>T mutations preferentially on the transcribed strand, rather than the untranscribed strand, of mRNAs, which further increased the likelihood of TP53 R248Q induction. Further, experimental thiopurine treatment was able to directly induce TP53 R248Q variants in MMR-deficient cultured cells, including Nalm6 and MCF10A MSH2-/-, by activating the thio-dMMR mutational signature, while MMR-proficient MCF10A cells did not experience R248Q induction. The sequential acquisition of MMR deficiency mutations, followed by TP53 mutations, during post-diagnosis ALL evolution was supported by clonal evolution analysis of serial patient samples. p53 R248Q promoted resistance to multiple ALL chemotherapeutic agents, and was associated with on-treatment relapse and poor relapse-treatment response. Our findings indicate that the enrichment of TP53 R248Q in relapsed ALL is due to synergistic mutagenesis from thiopurine treatment and MMR deficiency, followed by selection for TP53 R248Q9s chemoresistance phenotype. This suggests that cancer drug resistance mutations may not always pre-exist subclonally at diagnosis, but may be therapy-induced in some patients. Additionally, the qualitative and quantitative mutational signature output of a mutagen (e.g., thiopurines) can vary based on the genetic background. Finally, our findings suggest potential therapeutic strategies, including avoiding thiopurine treatment in MMR-deficient relapses, and therapeutic p53 mutant reactivation, to deal with this genetically-unstable, chemoresistant disease. Citation Format: Fan Yang, Samuel W. Brady, Huiying Sun, Chao Tang, Lijuan Du, Malwine Barz, Xiaotu Ma, Yao Chen,
化疗是治疗大多数儿童急性淋巴细胞白血病(ALL)。在这里,我们提供了直接的证据,证明硫嘌呤化疗也可以直接诱导耐药突变导致复发。通过对来自中国、美国和德国的复发性ALL患者的大型队列研究,我们发现与诊断相比,复发时TP53 R248Q突变高度富集。复发特异性TP53 R248Q与MSH2、MSH6或PMS2中MMR缺陷突变的获得以及一种新的复发特异性突变特征相关。通过对等基因MCF10A细胞进行或不进行MSH2基因敲除,以及对具有天然MMR缺陷的Nalm6 ALL细胞系进行研究,我们发现这种新的特征是由硫嘌呤治疗和错配修复(MMR)缺陷(称为硫代- dmmr特征)之间的协同诱变相互作用引起的,这种相互作用导致了缓解期间残留ALL的超突变表型和TP53 R248Q的获得。治疗诱导的tp53突变克隆随后由于广泛的化疗耐药而扩增,导致最终复发。事实上,硫嘌呤优先诱导NCG三核苷酸中心的C>T突变,从而导致TP53 R248Q,并且在mmr缺陷ALL和细胞系中,硫嘌呤突变率加快了2- 10倍。硫嘌呤处理优先诱导mrna转录链上的C>T突变,而不是非转录链上的突变,这进一步增加了TP53 R248Q诱导的可能性。此外,实验硫嘌呤处理能够通过激活硫代dmmr突变特征,在mmr缺陷培养细胞(包括Nalm6和MCF10A MSH2-/-)中直接诱导TP53 R248Q变异,而mmr熟练的MCF10A细胞则没有R248Q诱导。在诊断后ALL进化过程中,MMR缺陷突变和TP53突变的顺序获取得到了一系列患者样本克隆进化分析的支持。p53 R248Q促进对多种ALL化疗药物的耐药,并与治疗中复发和复发治疗不良反应相关。我们的研究结果表明,复发性ALL中TP53 R248Q的富集是由于硫嘌呤治疗和MMR缺乏的协同诱变,然后选择TP53 R248Q9s化疗耐药表型。这表明癌症耐药突变可能并不总是在诊断时预先存在亚克隆,但在一些患者中可能是治疗诱导的。此外,诱变原(例如,硫嘌呤)的定性和定量突变特征输出可以根据遗传背景而变化。最后,我们的研究结果提出了潜在的治疗策略,包括避免在mmr缺陷复发时使用硫嘌呤治疗,以及治疗性p53突变体再激活,以应对这种遗传不稳定,耐药的疾病。引用格式:杨帆,Samuel W. Brady,孙慧颖,唐超,杜丽娟,Malwine Barz,马晓图,陈瑶,方厚顺,李晓萌,Pandurang Kolekar, Omkar Pathak,蔡焦阳,丁丽霞,王天一,Arend von Stackelberg,沈淑红,段彩文,Cornelia Eckert,陈红转,刘宇,Jeffery M. Klco,李辉,李本尚,张景辉,Renate Kirschner-Schwabe,周斌斌硫嘌呤和错配修复缺陷共同促进TP53突变和ALL复发[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633期。
{"title":"Abstract 633: Thiopurines and mismatch repair deficiency cooperate to fuel TP53 mutagenesis and ALL relapse","authors":"Fan Yang, Samuel W. Brady, Huiying Sun, Chao Tang, L. Du, M. Barz, Xiaotu Ma, Yao Chen, Houshun Fang, Xiaomeng Li, Pandurang Kolekar, Omkar Pathak, J. Cai, Lixia Ding, Tianyi Wang, A. Stackelberg, S. Shen, C. Duan, C. Eckert, Hongzhuan Chen, Yu Liu, J. Klco, Hui Li, Ben-shang Li, Jinghui Zhang, R. Kirschner-Schwabe, Bin-Bing S. Zhou","doi":"10.1158/1538-7445.AM2021-633","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-633","url":null,"abstract":"Chemotherapy is curative for most children with acute lymphoblastic leukemia (ALL). Here we provide direct evidence that thiopurine chemotherapeutics can also directly induce drug resistance mutations leading to relapse. Using a large relapsed ALL cohort assembled from Chinese, US and German patients, we found that TP53 R248Q mutations were highly enriched at relapse compared to diagnosis. Relapse-specific TP53 R248Q was associated with the acquisition of MMR deficiency mutations in MSH2, MSH6, or PMS2 and a novel relapse-specific mutational signature. Using isogenic MCF10A cells with or without engineered MSH2 knockout, and the Nalm6 ALL cell line which has native MMR deficiency, we found that this novel signature was caused by a synergistic mutagenic interaction between thiopurine treatment and mismatch repair (MMR) deficiency (called the thio-dMMR signature) that contributes to a hypermutator phenotype and acquisition of TP53 R248Q in residual ALL during remission. Treatment-induced TP53-mutant clones then expand due to broad chemoresistance, leading to eventual relapse. Indeed, thiopurines preferentially induced C>T mutations at the center of NCG trinucleotides, which can lead to TP53 R248Q, and the thiopurine mutation rate was accelerated 2- to 10-fold in MMR-deficient ALL and cell lines. Thiopurine treatment induced C>T mutations preferentially on the transcribed strand, rather than the untranscribed strand, of mRNAs, which further increased the likelihood of TP53 R248Q induction. Further, experimental thiopurine treatment was able to directly induce TP53 R248Q variants in MMR-deficient cultured cells, including Nalm6 and MCF10A MSH2-/-, by activating the thio-dMMR mutational signature, while MMR-proficient MCF10A cells did not experience R248Q induction. The sequential acquisition of MMR deficiency mutations, followed by TP53 mutations, during post-diagnosis ALL evolution was supported by clonal evolution analysis of serial patient samples. p53 R248Q promoted resistance to multiple ALL chemotherapeutic agents, and was associated with on-treatment relapse and poor relapse-treatment response. Our findings indicate that the enrichment of TP53 R248Q in relapsed ALL is due to synergistic mutagenesis from thiopurine treatment and MMR deficiency, followed by selection for TP53 R248Q9s chemoresistance phenotype. This suggests that cancer drug resistance mutations may not always pre-exist subclonally at diagnosis, but may be therapy-induced in some patients. Additionally, the qualitative and quantitative mutational signature output of a mutagen (e.g., thiopurines) can vary based on the genetic background. Finally, our findings suggest potential therapeutic strategies, including avoiding thiopurine treatment in MMR-deficient relapses, and therapeutic p53 mutant reactivation, to deal with this genetically-unstable, chemoresistant disease. Citation Format: Fan Yang, Samuel W. Brady, Huiying Sun, Chao Tang, Lijuan Du, Malwine Barz, Xiaotu Ma, Yao Chen,","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"32 4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90608470","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.AM2021-644
S. Giacchetti, D. Vlachou, G. Bjarnason, David A. Rand, Francis Lévi
{"title":"Abstract 644: Tumor circadian dysfunction is associated with improved survival in breast cancer patients on neoadjuvant chemotherapy (NAC)","authors":"S. Giacchetti, D. Vlachou, G. Bjarnason, David A. Rand, Francis Lévi","doi":"10.1158/1538-7445.AM2021-644","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-644","url":null,"abstract":"","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"87 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85982641","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.AM2021-515
Assaf Magen, Pauline Hamon, Étienne Humblin, Alessandra S. Schanoski, Joel Kim, J. Berichel, E. Kenigsberg, M. Schwartz, T. Marron, A. Kamphorst, M. Merad
{"title":"Abstract 515: Heterogeneity of PD-1hiCD8 T cells associates with response to PD-1 blockade in hepatocellular carcinoma","authors":"Assaf Magen, Pauline Hamon, Étienne Humblin, Alessandra S. Schanoski, Joel Kim, J. Berichel, E. Kenigsberg, M. Schwartz, T. Marron, A. Kamphorst, M. Merad","doi":"10.1158/1538-7445.AM2021-515","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-515","url":null,"abstract":"","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"14 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86050074","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-LB071
N. Jahchan, M. Binnewies, Joshua L. Pollack, R. Mehta, S. Dash, Christina Tun, Erick Lu, Xiaoyan Du, K. Baker, L. Reyno, V. Sriram
The tumor microenvironment (TME) often contains high levels of suppressive myeloid cells that may contribute to innate checkpoint inhibitor (CPI) resistance. Pionyr9s Myeloid Tuning approach involves altering the composition and/or the function of myeloid cells in the TME. To this end, therapeutic targeting of tumor-associated macrophages (TAMs) is a promising strategy to increase CPI response rates in solid tumor indications, as well as to overcome resistance to CPI therapies. Pionyr and others identified the transmembrane protein triggering receptor expressed on myeloid cells-2 (TREM2) as a highly enriched TAMs target. Furthermore, TREM2 mRNA expression negatively correlates with patient survival in a variety of tumor types, supporting the involvement of TAMs in tumor progression. Pionyr developed a lead anti-TREM2 monoclonal antibody (mAb), termed PY314, as well as a murinized version of PY314, termed PY314m. PY314m demonstrated significant anti-tumor activity either as single agent in CPI-sensitive syngeneic tumor models or in combination with anti-PD-1 in CPI-resistant syngeneic tumor models. Mechanistically, PY314m reduced the pro-tumorigenic MHC class II-low, M2-like TAMs, induced pro-inflammatory cytokine production, significant increased CD8+ T cell infiltration into the TME. These findings suggest that PY314 therapy could be used to overcome CPI resistance in humans. To select patients most likely to benefit from PY314 therapy, Pionyr developed a qualitative IHC assay that detects TREM2 expression levels in formalin-fixed, paraffin-embedded human tumor tissues. Screening for TREM2 expression in tumor tissues demonstrated that TREM2+ TAMs were present in multiple solid tumor indications and their number increased with disease grade in a selected set of indications. Ongoing efforts are aimed at better understanding localization of TREM2+ TAMs within the TME, and spatial relationship of the TREM2+ TAMs to other immune cells present in the TME. The TREM2 IHC assay will be used to test our hypothesis that patients with tumors with high level of TREM2+ TAMs are most likely to benefit from PY314 treatment. Citation Format: Nadine S. Jahchan, Mikhail Binnewies, Joshua L. Pollack, Ranna Mehta, Subhadra Dash, Christine Tun, Erick Lu, Xiaoyan Du, Kevin P. Baker, Len Reyno, Venkataraman Sriram. Tuning the tumor myeloid microenvironment (TME) by targeting TREM2+ tumor-associated macrophages to overcome resistance to immune checkpoint inhibitors [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 LB071.
肿瘤微环境(TME)通常含有高水平的抑制性骨髓细胞,这可能有助于先天检查点抑制剂(CPI)的抵抗。Pionyr9s髓系调节方法涉及改变TME中髓系细胞的组成和/或功能。为此,肿瘤相关巨噬细胞(tam)的治疗靶向是一种很有前景的策略,可以提高实体肿瘤适应症中CPI的反应率,并克服对CPI治疗的耐药性。Pionyr等人发现髓样细胞-2表达的跨膜蛋白触发受体(TREM2)是高度富集的tam靶标。此外,在多种肿瘤类型中,TREM2 mRNA的表达与患者生存呈负相关,支持tam参与肿瘤进展。Pionyr开发了一种抗trem2单克隆抗体(mAb),称为PY314,以及PY314的鼠化版本,称为PY314m。PY314m无论是在cpi敏感的同基因肿瘤模型中单独使用,还是在cpi耐药的同基因肿瘤模型中与抗pd -1合用均显示出显著的抗肿瘤活性。在机制上,PY314m减少致瘤性MHC ii类低,m2样tam,诱导促炎细胞因子的产生,显著增加CD8+ T细胞浸润到TME。这些发现表明PY314疗法可用于克服人类的CPI耐药性。为了选择最有可能从PY314治疗中获益的患者,Pionyr开发了一种定性免疫组化检测方法,用于检测福尔马林固定石蜡包埋的人肿瘤组织中TREM2的表达水平。对肿瘤组织中TREM2表达的筛查表明,TREM2+ tam存在于多种实体肿瘤适应症中,并且在选定的一组适应症中,其数量随着疾病级别的增加而增加。正在进行的工作旨在更好地了解TREM2+ tam在TME中的定位,以及TREM2+ tam与TME中存在的其他免疫细胞的空间关系。TREM2 IHC检测将用于验证我们的假设,即具有高水平TREM2+ tam的肿瘤患者最有可能从PY314治疗中受益。引文格式:Nadine S. Jahchan, Mikhail Binnewies, Joshua L. Pollack, Ranna Mehta, Subhadra Dash, Christine Tun, Erick Lu, Xiaoyan Du, Kevin P. Baker, Len Reyno, Venkataraman Sriram通过靶向TREM2+肿瘤相关巨噬细胞来调节肿瘤髓系微环境(TME)以克服对免疫检查点抑制剂的抗性[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 LB071。
{"title":"Abstract LB071: Tuning the tumor myeloid microenvironment (TME) by targeting TREM2+tumor-associated macrophages to overcome resistance to immune checkpoint inhibitors","authors":"N. Jahchan, M. Binnewies, Joshua L. Pollack, R. Mehta, S. Dash, Christina Tun, Erick Lu, Xiaoyan Du, K. Baker, L. Reyno, V. Sriram","doi":"10.1158/1538-7445.AM2021-LB071","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-LB071","url":null,"abstract":"The tumor microenvironment (TME) often contains high levels of suppressive myeloid cells that may contribute to innate checkpoint inhibitor (CPI) resistance. Pionyr9s Myeloid Tuning approach involves altering the composition and/or the function of myeloid cells in the TME. To this end, therapeutic targeting of tumor-associated macrophages (TAMs) is a promising strategy to increase CPI response rates in solid tumor indications, as well as to overcome resistance to CPI therapies. Pionyr and others identified the transmembrane protein triggering receptor expressed on myeloid cells-2 (TREM2) as a highly enriched TAMs target. Furthermore, TREM2 mRNA expression negatively correlates with patient survival in a variety of tumor types, supporting the involvement of TAMs in tumor progression. Pionyr developed a lead anti-TREM2 monoclonal antibody (mAb), termed PY314, as well as a murinized version of PY314, termed PY314m. PY314m demonstrated significant anti-tumor activity either as single agent in CPI-sensitive syngeneic tumor models or in combination with anti-PD-1 in CPI-resistant syngeneic tumor models. Mechanistically, PY314m reduced the pro-tumorigenic MHC class II-low, M2-like TAMs, induced pro-inflammatory cytokine production, significant increased CD8+ T cell infiltration into the TME. These findings suggest that PY314 therapy could be used to overcome CPI resistance in humans. To select patients most likely to benefit from PY314 therapy, Pionyr developed a qualitative IHC assay that detects TREM2 expression levels in formalin-fixed, paraffin-embedded human tumor tissues. Screening for TREM2 expression in tumor tissues demonstrated that TREM2+ TAMs were present in multiple solid tumor indications and their number increased with disease grade in a selected set of indications. Ongoing efforts are aimed at better understanding localization of TREM2+ TAMs within the TME, and spatial relationship of the TREM2+ TAMs to other immune cells present in the TME. The TREM2 IHC assay will be used to test our hypothesis that patients with tumors with high level of TREM2+ TAMs are most likely to benefit from PY314 treatment. Citation Format: Nadine S. Jahchan, Mikhail Binnewies, Joshua L. Pollack, Ranna Mehta, Subhadra Dash, Christine Tun, Erick Lu, Xiaoyan Du, Kevin P. Baker, Len Reyno, Venkataraman Sriram. Tuning the tumor myeloid microenvironment (TME) by targeting TREM2+ tumor-associated macrophages to overcome resistance to immune checkpoint inhibitors [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 LB071.","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"54 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90934408","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.AM2021-380
D. Ren, Bo Zhang, Renwang Liu, Fuyu Gong, Yue-zong Bai, W. Xie, Huandong Huo, Hao Zhang, Zuoqing Song
Background: Immune checkpoint inhibitors targeting the programmed cell death-1 receptor (PD-1) improve survival in a subset of patients with NSCLC. There are still lots of patients could not reach the clinical benefit, even though with the positive expression of the programmed cell death 1 ligand 1 (PD-L1). Here, we aimed to research the immune resistance mechanism in NSCLC. Methods: The genomic data and clinical data of the discovery cohort was obtained from The Cancer Genome Atlas (TCGA). The clinical data of validation cohort in NSCLC treated by immunotherapy was retrospective collected. And tissue from patients with NSCLC were performed to whole exome sequencing in a College of American Pathologists-certified and Clinical Laboratory Improvement Amendments-accredited lab. Results: Activation of WNT signaling was inferred that somatic mutations or somatic copy number alterations in WNT signaling elements including APC, WNT16, AXIN2, WNT4, AXIN1, CTNNB1, BCL9L and SMAD4. Two cohort have been enrolled in this study, the discovery cohort from TCGA, and the validation cohort from the Chinese patients with NSCLC. The frequency of WNT pathway alterations from the TCGA cohort was 16%, and which was represented mutually exclusive molecular subsets. In TCGA cohort, activating alteration WNT signaling were associated with shorter median PFS (18.8 vs 3.9 months, p=0.036, HR=0.24 (0.09-0.68)). Conclusions: Mutations predicted to activate the WNT pathway were associated with innate resistance to immune checkpoint blockade in NSCLC Citation Format: Dian Ren, Bo Zhang, Renwang Liu, Fuyu Gong, Yuezong Bai, Wenzhuan Xie, Huandong Huo, Hao Zhang, Zuoqing Song. WNT signal pathway activation correlates with innate resistant to immune checkpoint therapies in NSCLC [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 380.
背景:靶向程序性细胞死亡-1受体(PD-1)的免疫检查点抑制剂可提高部分非小细胞肺癌患者的生存率。尽管程序性细胞死亡1配体1 (PD-L1)阳性表达,仍有许多患者未能达到临床获益。在此,我们旨在研究非小细胞肺癌的免疫抵抗机制。方法:从癌症基因组图谱(TCGA)中获取发现队列的基因组数据和临床数据。回顾性收集非小细胞肺癌免疫治疗验证队列的临床资料。非小细胞肺癌患者的组织在美国病理学家学会认证和临床实验室改进修订认可的实验室进行全外显子组测序。结果:通过APC、WNT16、AXIN2、WNT4、AXIN1、CTNNB1、BCL9L和SMAD4等WNT信号元件的体细胞突变或体细胞拷贝数改变,推测WNT信号的激活。本研究纳入了两个队列,来自TCGA的发现队列和来自中国NSCLC患者的验证队列。来自TCGA队列的WNT通路改变的频率为16%,这代表了相互排斥的分子亚群。在TCGA队列中,激活改变WNT信号与较短的中位PFS相关(18.8 vs 3.9个月,p=0.036, HR=0.24(0.09-0.68))。结论:预测激活WNT通路的突变与非小细胞肺癌(NSCLC)免疫checkpoint阻断的先天耐药有关。引用本文:任dian,张博,刘仁旺,龚福玉,白跃宗,谢文转,霍焕东,张昊,宋佐清。WNT信号通路激活与非小细胞肺癌免疫检查点疗法的先天耐药相关[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380期。
{"title":"Abstract 380: WNT signal pathway activation correlates with innate resistant to immune checkpoint therapies in NSCLC","authors":"D. Ren, Bo Zhang, Renwang Liu, Fuyu Gong, Yue-zong Bai, W. Xie, Huandong Huo, Hao Zhang, Zuoqing Song","doi":"10.1158/1538-7445.AM2021-380","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-380","url":null,"abstract":"Background: Immune checkpoint inhibitors targeting the programmed cell death-1 receptor (PD-1) improve survival in a subset of patients with NSCLC. There are still lots of patients could not reach the clinical benefit, even though with the positive expression of the programmed cell death 1 ligand 1 (PD-L1). Here, we aimed to research the immune resistance mechanism in NSCLC. Methods: The genomic data and clinical data of the discovery cohort was obtained from The Cancer Genome Atlas (TCGA). The clinical data of validation cohort in NSCLC treated by immunotherapy was retrospective collected. And tissue from patients with NSCLC were performed to whole exome sequencing in a College of American Pathologists-certified and Clinical Laboratory Improvement Amendments-accredited lab. Results: Activation of WNT signaling was inferred that somatic mutations or somatic copy number alterations in WNT signaling elements including APC, WNT16, AXIN2, WNT4, AXIN1, CTNNB1, BCL9L and SMAD4. Two cohort have been enrolled in this study, the discovery cohort from TCGA, and the validation cohort from the Chinese patients with NSCLC. The frequency of WNT pathway alterations from the TCGA cohort was 16%, and which was represented mutually exclusive molecular subsets. In TCGA cohort, activating alteration WNT signaling were associated with shorter median PFS (18.8 vs 3.9 months, p=0.036, HR=0.24 (0.09-0.68)). Conclusions: Mutations predicted to activate the WNT pathway were associated with innate resistance to immune checkpoint blockade in NSCLC Citation Format: Dian Ren, Bo Zhang, Renwang Liu, Fuyu Gong, Yuezong Bai, Wenzhuan Xie, Huandong Huo, Hao Zhang, Zuoqing Song. WNT signal pathway activation correlates with innate resistant to immune checkpoint therapies in NSCLC [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 380.","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"38 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91100015","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.AM2021-376
H. Kang, I. Kim, Jin Woo Kim
Introduction: The Glasgow prognostic score (GPS) reflects the host systemic inflammatory response and is validated prognostic factor in lung cancer. However, little is known about the prognostic role in non-small cell lung cancer (NSCLC) patients treated with immunotherapy after platinum-based cytotoxic chemotherapy. Patient and Methods: This study used a lung cancer cohort of the Catholic Medical Center of Korea between January 2018 and June 2020. We included the patients who were diagnosed with unresectable advanced stage NSCLC or recurrent disease after pulmonary resection and had received at least one regimen of platinum-based chemotherapy before being administered immunotherapy. The patients with NSCLC treated with anti-PD1 or anti-PD-L1 (pembrolizumab, nivolumab, or atezolizumab) and assessed the prognostic value of the GPS. The GPS was calculated using C-reactive protein and albumin concentrations within 1 week before starting anti-PD1 or anti-PD-L1 treatment. Results: A total of 78 patients with NSCLC treated with immunotherapy as 2nd or 3rd line therapy after platinum-based chemotherapy. Kaplan-Meier analyses revealed that higher GPS was significant predictors of shorter progression free survival (PFS) (Log rank Conclusion: Higher GPS were identified as one of poor prognostic factor for OS and PFS in NSCLC patients received immunotherapy as 2nd or 3rd line therapy after platinum-based chemotherapy. Citation Format: Hye Seon Kang, In Kyoung Kim, Jin Woo Kim. Prognostic significance of Glasgow prognostic score in NSCLC patients treated with immunotherapy after platinum-based cytotoxic chemotherapy [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 376.
格拉斯哥预后评分(GPS)反映了宿主的全身炎症反应,是肺癌的有效预后因素。然而,对于非小细胞肺癌(NSCLC)患者在铂基细胞毒性化疗后接受免疫治疗的预后作用知之甚少。患者和方法:本研究使用了2018年1月至2020年6月期间韩国天主教医疗中心的肺癌队列。我们纳入了在肺切除术后被诊断为不可切除的晚期非小细胞肺癌或复发性疾病的患者,并在给予免疫治疗之前接受了至少一种基于铂的化疗方案。NSCLC患者接受抗pd1或抗pd - l1(派姆单抗、纳武单抗或阿特唑单抗)治疗,并评估GPS的预后价值。在开始抗pd1或抗pd - l1治疗前1周内,使用c反应蛋白和白蛋白浓度计算GPS。结果:共有78例NSCLC患者在铂类化疗后接受免疫治疗作为二线或三线治疗。Kaplan-Meier分析显示,较高的GPS是较短无进展生存期(PFS)的重要预测因子(Log rank)。结论:较高的GPS被确定为非小细胞肺癌患者在铂基化疗后接受免疫治疗作为第二或三线治疗的OS和PFS的不良预后因素之一。引文格式:Hye Seon Kang, In Kyoung Kim, Jin Woo Kim。基于铂的细胞毒化疗后免疫治疗的NSCLC患者格拉斯哥预后评分的预后意义[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376期。
{"title":"Abstract 376: Prognostic significance of Glasgow prognostic score in NSCLC patients treated with immunotherapy after platinum-based cytotoxic chemotherapy","authors":"H. Kang, I. Kim, Jin Woo Kim","doi":"10.1158/1538-7445.AM2021-376","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-376","url":null,"abstract":"Introduction: The Glasgow prognostic score (GPS) reflects the host systemic inflammatory response and is validated prognostic factor in lung cancer. However, little is known about the prognostic role in non-small cell lung cancer (NSCLC) patients treated with immunotherapy after platinum-based cytotoxic chemotherapy. Patient and Methods: This study used a lung cancer cohort of the Catholic Medical Center of Korea between January 2018 and June 2020. We included the patients who were diagnosed with unresectable advanced stage NSCLC or recurrent disease after pulmonary resection and had received at least one regimen of platinum-based chemotherapy before being administered immunotherapy. The patients with NSCLC treated with anti-PD1 or anti-PD-L1 (pembrolizumab, nivolumab, or atezolizumab) and assessed the prognostic value of the GPS. The GPS was calculated using C-reactive protein and albumin concentrations within 1 week before starting anti-PD1 or anti-PD-L1 treatment. Results: A total of 78 patients with NSCLC treated with immunotherapy as 2nd or 3rd line therapy after platinum-based chemotherapy. Kaplan-Meier analyses revealed that higher GPS was significant predictors of shorter progression free survival (PFS) (Log rank Conclusion: Higher GPS were identified as one of poor prognostic factor for OS and PFS in NSCLC patients received immunotherapy as 2nd or 3rd line therapy after platinum-based chemotherapy. Citation Format: Hye Seon Kang, In Kyoung Kim, Jin Woo Kim. Prognostic significance of Glasgow prognostic score in NSCLC patients treated with immunotherapy after platinum-based cytotoxic chemotherapy [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 376.","PeriodicalId":10518,"journal":{"name":"Clinical Research (Excluding Clinical Trials)","volume":"36 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91189799","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}