首页 > 最新文献

Blood Cancer Discovery最新文献

英文 中文
Abstract P04: Role of glucocorticoid receptor in normal and malignant germinal center B cells 摘要 P04:糖皮质激素受体在正常和恶性生殖中心 B 细胞中的作用
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p04
Clarissa Corinaldesi, A. Holmes, Elodie Bal, L. Pasqualucci, K. Basso, R. Dalla-Favera
We have reported that super-enhancers (SEs) are specifically hypermutated by Activation-Induced Deaminase (AID)-induced Aberrant Somatic Hypermutation in >90% of diffuse large B cell lymphoma (DLBCL) cases (Bal et al. Nature 2022). Analysis of 122 primary DLBCL biopsies and cell lines identified more than 80 SEs that are recurrently hypermutated, with an average of 12 hypermutated SE/case. Hypermutated SEs are predominantly linked to genes encoding regulators of B cell development and well-known lymphoma oncogenes, including BCL6, BCL2 and CXCR4. We showed that specific hotspot mutations prevent binding and transcriptional downregulation by transcriptional repressors leading to target gene dysregulation. Among these, hypermutation of the SEs linked to BCL2 and CXCR4 abrogates the binding of the glucocorticoid receptor (GR)/transcription factor encoded by NR3C1. As a result, BCL2 and CXCR4 escape GR-mediated transcriptional repression leading to their de-regulated expression in germinal center (GC) B cells. Together with the observation that the NR3C1 gene is genetically inactivated in a small fraction of DLBCL, and the relevance of glucocorticoid-based therapy in DLBCL, these results prompted a comprehensive analysis of the role of GR in normal and malignant B cells. We report that GR is detectable in the nucleus of the great majority of GC B cells, suggesting that it is active as a transcription factor. Similarly, DLBCL primary cases (including GCB-, ABC- and unclassified DLBCL) display GR nuclear expression. Analysis of a conditional GC-specific Nr3c1 knock-out (KO) mouse model showed that GC B cells form normally in the absence of Nr3c1. However, the transcriptional profile of Nr3c1-KO GC B cells was significantly altered compared to wild-type cells. By integrating the GR binding profile obtained in normal human GC B cells with the transcripts differentially expressed in Nr3c1-KO GC B cells, we identified a set of about 2,000 genes that are directly modulated by GR during the GC reaction. Pathway enrichment analysis showed that GR controls several signaling pathways, suggesting a modulatory role on the BCR and CXCR4 pathways. In addition, GR targets display a significant overlap with the BCL6 transcriptional network, suggesting a cooperative role in GC B cell development. Notably, GR was shown to regulate pathways involved in plasma cell differentiation, while repressing targets associated with memory B cell development. In DLBCL cases, several NR3C1 binding sites are recurrently targeted by mutations, suggesting that genetic impairment of GR activity is heterogeneous and extends beyond CXCR4 and BCL2. Together, these data support a role for the glucocorticoid pathway in GC physiology. Recurrent inactivation of the NR3C1 gene or part of its transcriptional regulatory program may contribute to DLBCL pathogenesis, with implications for presently unknown specific roles of glucocorticoids in the therapeutic regimens for DLBCL. Citation Format:
我们曾报道过,在超过90%的弥漫大B细胞淋巴瘤(DLBCL)病例中,超级增强子(SE)会因活化诱导脱氨酶(AID)诱导的异常体细胞高突变而发生特异性高突变(Bal等人,Nature 2022)。对 122 例原发性 DLBCL 活检组织和细胞系的分析确定了 80 多个反复发生高突变的 SE,平均每个病例有 12 个高突变 SE。高突变SE主要与编码B细胞发育调控因子和著名淋巴瘤致癌基因(包括BCL6、BCL2和CXCR4)的基因有关。我们发现,特定的热点突变会阻止转录抑制因子的结合和转录下调,从而导致靶基因失调。结果,BCL2和CXCR4逃脱了GR介导的转录抑制,导致它们在生殖中心(GC)B细胞中的表达失调。观察发现,在一小部分 DLBCL 中,NR3C1 基因在基因上失活,而且糖皮质激素疗法与 DLBCL 相关,这些结果促使我们对 GR 在正常和恶性 B 细胞中的作用进行全面分析。我们报告说,绝大多数 GC B 细胞的细胞核中都能检测到 GR,这表明它作为一种转录因子非常活跃。同样,DLBCL 原始病例(包括 GCB-、ABC- 和未分类的 DLBCL)也有 GR 核表达。对条件性 GC 特异性 Nr3c1 基因敲除(KO)小鼠模型的分析表明,在缺乏 Nr3c1 的情况下,GC B 细胞可正常形成。然而,与野生型细胞相比,Nr3c1-KO GC B 细胞的转录谱发生了显著变化。通过整合在正常人GC B细胞中获得的GR结合谱和在Nr3c1-KO GC B细胞中差异表达的转录本,我们确定了一组在GC反应过程中直接受GR调控的约2000个基因。通路富集分析表明,GR 控制着几种信号通路,这表明它对 BCR 和 CXCR4 通路起着调节作用。此外,GR靶标与BCL6转录网络有明显重叠,表明其在GC B细胞发育中起着合作作用。值得注意的是,GR可调节浆细胞分化的通路,同时抑制与记忆性B细胞发育相关的靶点。在DLBCL病例中,几个NR3C1结合位点反复出现突变,这表明遗传对GR活性的损害是异质性的,而且超出了CXCR4和BCL2的范围。这些数据共同支持了糖皮质激素通路在 GC 生理学中的作用。NR3C1基因或其部分转录调控程序的复发性失活可能会导致DLBCL的发病机制,并对目前未知的糖皮质激素在DLBCL治疗方案中的特定作用产生影响。引用格式:Clarissa Corinaldesi, Antony B Holmes, Elodie Bal, Laura Pasqualucci, Katia Basso, Riccardo Dalla-Favera。糖皮质激素受体在正常和恶性生殖中心 B 细胞中的作用 [摘要]。见:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P04.
{"title":"Abstract P04: Role of glucocorticoid receptor in normal and malignant germinal center B cells","authors":"Clarissa Corinaldesi, A. Holmes, Elodie Bal, L. Pasqualucci, K. Basso, R. Dalla-Favera","doi":"10.1158/2643-3249.bcdsymp24-p04","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p04","url":null,"abstract":"\u0000 We have reported that super-enhancers (SEs) are specifically hypermutated by Activation-Induced Deaminase (AID)-induced Aberrant Somatic Hypermutation in >90% of diffuse large B cell lymphoma (DLBCL) cases (Bal et al. Nature 2022). Analysis of 122 primary DLBCL biopsies and cell lines identified more than 80 SEs that are recurrently hypermutated, with an average of 12 hypermutated SE/case. Hypermutated SEs are predominantly linked to genes encoding regulators of B cell development and well-known lymphoma oncogenes, including BCL6, BCL2 and CXCR4. We showed that specific hotspot mutations prevent binding and transcriptional downregulation by transcriptional repressors leading to target gene dysregulation. Among these, hypermutation of the SEs linked to BCL2 and CXCR4 abrogates the binding of the glucocorticoid receptor (GR)/transcription factor encoded by NR3C1. As a result, BCL2 and CXCR4 escape GR-mediated transcriptional repression leading to their de-regulated expression in germinal center (GC) B cells. Together with the observation that the NR3C1 gene is genetically inactivated in a small fraction of DLBCL, and the relevance of glucocorticoid-based therapy in DLBCL, these results prompted a comprehensive analysis of the role of GR in normal and malignant B cells. We report that GR is detectable in the nucleus of the great majority of GC B cells, suggesting that it is active as a transcription factor. Similarly, DLBCL primary cases (including GCB-, ABC- and unclassified DLBCL) display GR nuclear expression. Analysis of a conditional GC-specific Nr3c1 knock-out (KO) mouse model showed that GC B cells form normally in the absence of Nr3c1. However, the transcriptional profile of Nr3c1-KO GC B cells was significantly altered compared to wild-type cells. By integrating the GR binding profile obtained in normal human GC B cells with the transcripts differentially expressed in Nr3c1-KO GC B cells, we identified a set of about 2,000 genes that are directly modulated by GR during the GC reaction. Pathway enrichment analysis showed that GR controls several signaling pathways, suggesting a modulatory role on the BCR and CXCR4 pathways. In addition, GR targets display a significant overlap with the BCL6 transcriptional network, suggesting a cooperative role in GC B cell development. Notably, GR was shown to regulate pathways involved in plasma cell differentiation, while repressing targets associated with memory B cell development. In DLBCL cases, several NR3C1 binding sites are recurrently targeted by mutations, suggesting that genetic impairment of GR activity is heterogeneous and extends beyond CXCR4 and BCL2. Together, these data support a role for the glucocorticoid pathway in GC physiology. Recurrent inactivation of the NR3C1 gene or part of its transcriptional regulatory program may contribute to DLBCL pathogenesis, with implications for presently unknown specific roles of glucocorticoids in the therapeutic regimens for DLBCL.\u0000 Citation Format: ","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140080212","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}
引用次数: 0
Abstract P06: Characterization of the Immune Tumor Microenvironment in a Diverse Cohort of Patients with Multiple Myeloma 摘要 P06:多发性骨髓瘤患者免疫肿瘤微环境的特征描述
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p06
Suganti Shivaram, Hongwei Tang, Huihuang Yan, Shulan Tian, Guyu Qin, Emilie I. Anderson, Neeraj Sharma, Abiola Bolarinwa, Cinthya Zepeda-Mendoza, Stacey Lehman, Felicia Gomez, A. Mitra, Surendra Dasari, T. Kourelis, S. Kumar, Linda Baughn
Multiple myeloma (MM) is the most common blood cancer in African American (AA) individuals. AAs have an increased risk of developing Monoclonal Gammopathy of Undetermined Significance (MGUS) and MM compared to non-Hispanic White (NHW) individuals. While several studies have shown a pivotal role for the immune tumor microenvironment (iTME) in MM pathogenesis, the composition of the iTME in AAs with MM is under investigated. Viably frozen whole bone marrow (BM) aspirates from 128 individuals (53 AA and 75 NHW) with either MM (n=65), MGUS (n=28), smoldering MM (SMM, n=13), or healthy controls (n=22) were evaluated by single cell RNA-sequencing (scRNA-seq) or mass cytometry (CyTOF). We characterized 34 (14 AA and 20 NHW) by scRNA-seq and 94 (39 AA and 55 NHW) by CyTOF. CyTOF samples were stained with Maxpar Direct Immune Profiling Assay Kit (Fluidigm), acquired on a Helios mass cytometer and analyzed in Cytobank. Data were analyzed by logistic equivalent regression model with p<0.05 for significance. ScRNA-seq samples were processed as in Yao, 2022. Data were normalized using scvi-tools and clustered by Uniform manifold approximation and projection (UMAP). Differentially expressed markers between clusters were identified using Model-based Analysis of Single-cell Transcriptomics (MAST) in Seurat R package at FDR < 0.05 and log2 fold change > 0.55. ScRNA-seq of the CD45+ non-tumor lymphocytes revealed 17 immune populations (HSC, erythroblast, MDSC, GMP, CD1c+ DC, CD4+ and CD8+ T cell, CD14+ M1 macrophage, CD16+monocyte, monocyte, pDC, naïve B, pre-B, pro-B, memory B, NK/MAIT). CD3E/D+cells were clustered into naïve/central memory (CCR7+, LEF1+, SELL+), memory (GZMK+), effector (GZMB+, GZMH+, FGFBP2+), g/d (TRDC+), MAIT (SLC4A10+, KLRB1+) and IFN CD8+ T cells (IFIT1+, IFIT2+,IFIT3+). Disease progression was associated with increases in median CD4+ and CD8+ GZMK+ T cells, memory B cells and a reduction in all monocyte clusters. Within the diseased cohort in comparison to NHW patients, AA patients had increased CD3+ T cells (CD4 and CD8+) (AA 57.5% vs. NHW 44.3%) and a reduction in overall monocytes (AA 10.5% vs. 26.5%). These results were validated by CyTOF, which confirmed an increase in CD3+ T cells (AA 53.7% vs. NHW 35.7%, p<0.001) including increases in CD8+ (AA 7.0% vs. NHW 3.2%, p<0.05) and CD4+ terminal effector T cells (AA 5.7% vs. NHW 2.7%, p<0.05) and a reduction in monocytes (AA 18.3% vs. NHW 26.6%) among AAs. After adjusting for age and sex, AAs retained an increase in T cells (OR 1.59 (95%CI 1.2-2.01, p=0.00023)) and a reduction in monocytes (OR 0.88 (95%CI 0.78-1, p=0.045)). Despite the increase in CD8+ T cells in AA patients, we identified reduced GZMK+ T cells and reductions in exhaustion markers (TIGIT, LAG3, TOX) in CD8+ T cells in comparison to NHWs. Nevertheless, the distribution of the immune populations was similar between healthy AA and NHW individuals. Our multiomics approach to characterize the iTME identified significant diffe
多发性骨髓瘤(MM)是非裔美国人(AA)最常见的血癌。与非西班牙裔白人(NHW)相比,非裔美国人罹患意义未定的单克隆丙种球蛋白病(MGUS)和多发性骨髓瘤的风险更高。虽然多项研究表明免疫肿瘤微环境(iTME)在MM发病机制中起着关键作用,但对MM患者中AAs的iTME组成还在研究中。我们通过单细胞 RNA 序列测定(scRNA-seq)或质谱细胞计数法(CyTOF)对 128 例(53 例 AA 和 75 例 NHW)MM(n=65)、MGUS(n=28)、烟雾型 MM(SMM,n=13)或健康对照组(n=22)患者的活体冷冻全骨髓(BM)抽吸物进行了评估。我们通过 scRNA-seq 鉴定了 34 例(14 例 AA 和 20 例 NHW),通过 CyTOF 鉴定了 94 例(39 例 AA 和 55 例 NHW)。CyTOF样本用Maxpar Direct Immune Profiling Assay Kit (Fluidigm)染色,在Helios质谱仪上采集,并在Cytobank中进行分析。数据采用逻辑等效回归模型进行分析,P 值为 0.55。CD45+非肿瘤淋巴细胞的ScRNA-seq显示了17种免疫群体(造血干细胞、红细胞、MDSC、GMP、CD1c+ DC、CD4+和CD8+ T细胞、CD14+ M1巨噬细胞、CD16+单核细胞、单核细胞、pDC、幼稚B细胞、前B细胞、前B细胞、记忆B细胞、NK/MAIT)。CD3E/D+细胞分为幼稚/中心记忆细胞(CCR7+、LEF1+、SELL+)、记忆细胞(GZMK+)、效应细胞(GZMB+、GZMH+、FGFBP2+)、g/d细胞(TRDC+)、MAIT细胞(SLC4A10+、KLRB1+)和IFN CD8+T细胞(IFIT1+、IFIT2+、IFIT3+)。疾病进展与中位 CD4+ 和 CD8+ GZMK+ T 细胞、记忆 B 细胞的增加以及所有单核细胞群的减少有关。在患病队列中,与白血病患者相比,AA 患者的 CD3+ T 细胞(CD4 和 CD8+)增加(AA 57.5% 对白血病 44.3%),单核细胞总数减少(AA 10.5% 对白血病 26.5%)。CyTOF 验证了这些结果,证实 AAs 中 CD3+ T 细胞增加(AA 53.7% vs. NHW 35.7%,p<0.001),包括 CD8+ 细胞增加(AA 7.0% vs. NHW 3.2%,p<0.05)和 CD4+ 末端效应 T 细胞增加(AA 5.7% vs. NHW 2.7%,p<0.05),单核细胞减少(AA 18.3% vs. NHW 26.6%)。对年龄和性别进行调整后,AAs 保持了 T 细胞的增加(OR 1.59 (95%CI 1.2-2.01, p=0.00023))和单核细胞的减少(OR 0.88 (95%CI 0.78-1, p=0.045))。尽管 AA 患者的 CD8+ T 细胞有所增加,但与 NHWs 相比,我们发现 GZMK+ T 细胞减少,CD8+ T 细胞中的衰竭标记(TIGIT、LAG3、TOX)也有所减少。尽管如此,健康 AA 人和 NHW 人的免疫群体分布情况相似。我们的多组学方法确定了 iTME 的特征,发现 AA 与 NHW MM 患者之间存在显著差异。引用格式:Suganti Shivaram, Hongwei Tang, Huihuang Yan, Shulan Tian, Guyu Qin, Emilie Anderson, Neeraj Sharma, Abiola Bolarinwa, Cinthya Zepeda-Mendoza, Stacey Lehman, Felicia Gomez, Amit Mitra, Surendra Dasari, Taxiarchis Kourelis, Shaji Kumar, Linda Baughn.多发性骨髓瘤患者免疫肿瘤微环境的特征[摘要]。见:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P06.
{"title":"Abstract P06: Characterization of the Immune Tumor Microenvironment in a Diverse Cohort of Patients with Multiple Myeloma","authors":"Suganti Shivaram, Hongwei Tang, Huihuang Yan, Shulan Tian, Guyu Qin, Emilie I. Anderson, Neeraj Sharma, Abiola Bolarinwa, Cinthya Zepeda-Mendoza, Stacey Lehman, Felicia Gomez, A. Mitra, Surendra Dasari, T. Kourelis, S. Kumar, Linda Baughn","doi":"10.1158/2643-3249.bcdsymp24-p06","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p06","url":null,"abstract":"\u0000 Multiple myeloma (MM) is the most common blood cancer in African American (AA) individuals. AAs have an increased risk of developing Monoclonal Gammopathy of Undetermined Significance (MGUS) and MM compared to non-Hispanic White (NHW) individuals. While several studies have shown a pivotal role for the immune tumor microenvironment (iTME) in MM pathogenesis, the composition of the iTME in AAs with MM is under investigated. Viably frozen whole bone marrow (BM) aspirates from 128 individuals (53 AA and 75 NHW) with either MM (n=65), MGUS (n=28), smoldering MM (SMM, n=13), or healthy controls (n=22) were evaluated by single cell RNA-sequencing (scRNA-seq) or mass cytometry (CyTOF). We characterized 34 (14 AA and 20 NHW) by scRNA-seq and 94 (39 AA and 55 NHW) by CyTOF. CyTOF samples were stained with Maxpar Direct Immune Profiling Assay Kit (Fluidigm), acquired on a Helios mass cytometer and analyzed in Cytobank. Data were analyzed by logistic equivalent regression model with p<0.05 for significance. ScRNA-seq samples were processed as in Yao, 2022. Data were normalized using scvi-tools and clustered by Uniform manifold approximation and projection (UMAP). Differentially expressed markers between clusters were identified using Model-based Analysis of Single-cell Transcriptomics (MAST) in Seurat R package at FDR < 0.05 and log2 fold change > 0.55. ScRNA-seq of the CD45+ non-tumor lymphocytes revealed 17 immune populations (HSC, erythroblast, MDSC, GMP, CD1c+ DC, CD4+ and CD8+ T cell, CD14+ M1 macrophage, CD16+monocyte, monocyte, pDC, naïve B, pre-B, pro-B, memory B, NK/MAIT). CD3E/D+cells were clustered into naïve/central memory (CCR7+, LEF1+, SELL+), memory (GZMK+), effector (GZMB+, GZMH+, FGFBP2+), g/d (TRDC+), MAIT (SLC4A10+, KLRB1+) and IFN CD8+ T cells (IFIT1+, IFIT2+,IFIT3+). Disease progression was associated with increases in median CD4+ and CD8+ GZMK+ T cells, memory B cells and a reduction in all monocyte clusters. Within the diseased cohort in comparison to NHW patients, AA patients had increased CD3+ T cells (CD4 and CD8+) (AA 57.5% vs. NHW 44.3%) and a reduction in overall monocytes (AA 10.5% vs. 26.5%). These results were validated by CyTOF, which confirmed an increase in CD3+ T cells (AA 53.7% vs. NHW 35.7%, p<0.001) including increases in CD8+ (AA 7.0% vs. NHW 3.2%, p<0.05) and CD4+ terminal effector T cells (AA 5.7% vs. NHW 2.7%, p<0.05) and a reduction in monocytes (AA 18.3% vs. NHW 26.6%) among AAs. After adjusting for age and sex, AAs retained an increase in T cells (OR 1.59 (95%CI 1.2-2.01, p=0.00023)) and a reduction in monocytes (OR 0.88 (95%CI 0.78-1, p=0.045)). Despite the increase in CD8+ T cells in AA patients, we identified reduced GZMK+ T cells and reductions in exhaustion markers (TIGIT, LAG3, TOX) in CD8+ T cells in comparison to NHWs. Nevertheless, the distribution of the immune populations was similar between healthy AA and NHW individuals. Our multiomics approach to characterize the iTME identified significant diffe","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140079755","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}
引用次数: 0
Abstract P34: Bayesian networks modeling identifies a reliance of TP53 mutant AML on NF kappa B signaling 摘要 P34:贝叶斯网络建模确定了 TP53 突变型 AML 对 NF kappa B 信号的依赖性
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p34
Daniel Chang, Klara E Noble-Orcutt, Marie L Antony, Yoonkyu Lee, Fiona He, Karen Sachs, Chad L Myers, Z. Sachs
In acute myeloid leukemia (AML), TP53 mutations ( TP53Mut ) confer the worst prognosis. Leukemia stem cells (LSCs) are rare AML cells endowed with self-renewal capacity, allowing them to recapitulate leukemia after therapy and cause disease progression. In human AML, signaling pathways associated with inflammation have been implicated in the pathogenesis of TP53Mut AML, but the pathways that mediate self-renewal in human TP53Mut AML are not well-known. To define the signaling pathways that are activated in TP53Mut human LSCs, we used CyTOF, a form of flow cytometry that can measure up to 40 proteins simultaneously at single-cell resolution. We compared the levels of activated signaling molecules in the LSC-enriched (CD34+CD38−) subset of seven TP53Mut and seven TP53 wild-type ( TP53WT ) AML samples. Phosphorylated NF kappa B (pNFkB), pSTAT1, and pP38 are significantly higher in TP53Mut AMLs. In contrast, Ki67 and pMAPKAPKII are higher in TP53WT AML. These data demonstrate a unique signaling activation state in TP53Mut AML. The influence of signaling molecules on downstream targets can vary by cellular context. Therefore, we asked whether the influence and interaction of the activated signaling molecules vary between TP53Mut and TP53WT LSCs. To model the signaling architecture, we performed Bayesian networks modeling, a machine learning algorithm that has been validated as a method to discover statistical correlations and dependencies between signaling molecules. We found a similar global signaling network structure and hierarchy in TP53Mut and TP53WT AMLs. However, TP53Mut model showed NFkB have strong influences on phosphorylated Histone H3 (pHIS3) and Ki67, suggesting that proliferation depends on NFkB levels in TP53Mut LSCs. Furthermore, NFkB levels were highly dependent on pSTAT1 in TP53Mut LSCs. Next, we compared the transcriptional profiles of TP53Mut and TP53WT samples in the BEAT AML dataset and found that TP53Mut AML displayed significant enrichment of all the NFkB gene sets (n=19) in the molecular signatures database, providing more evidence for a unique reliance of TP53Mut human AML on NFkB signaling. Therefore, we used lentivirus transduced shRNA constructs to determine the influence of mutant p53 on signaling in human AML. We found that knockdown of TP53Mut reduced NFkB protein levels in Kasumi AML cells (which are TP53R248Q/ − ) but not in MOLM13 AML cells (which are TP53WT). RNA sequencing of transduced Kasumi cells showed that TP53Mut knockdown led to loss of NFkB and LSC self-renewal signatures. Knockdown of TP53Mut in primary human TP53Mut AMLs (S127F/- and S241Y/-) led to a similar loss of NFkB and LSC signatures in these primary samples as well. Colony formation in Kasumi cells and TP53Mut primary human AML were also abrogated after TP53Mut knockdown. These data show TP53Mut promotes NFkB activation in human AML and suggests that human TP53Mut LSCs may be dependent on NFkB for self-renewal. These data implicate NFkB as a pos
在急性髓性白血病(AML)中,TP53突变(TP53Mut)的预后最差。白血病干细胞(LSCs)是罕见的急性髓性白血病细胞,具有自我更新能力,可在治疗后再现白血病并导致疾病进展。在人类急性髓细胞性白血病中,与炎症相关的信号通路被认为与TP53Mut急性髓细胞性白血病的发病机制有关,但介导人类TP53Mut急性髓细胞性白血病自我更新的通路却不为人所知。为了确定在 TP53Mut 人类 LSCs 中被激活的信号通路,我们使用了 CyTOF,这是一种流式细胞术,能以单细胞分辨率同时测量多达 40 种蛋白质。我们比较了 7 个 TP53Mut 和 7 个 TP53 野生型(TP53WT)AML 样本的 LSC 富集(CD34+CD38-)亚组中活化信号分子的水平。在 TP53Mut AMLs 中,磷酸化 NF kappa B(pNFkB)、pSTAT1 和 pP38 明显升高。相比之下,TP53WT AML 中的 Ki67 和 pMAPKAPKII 则更高。这些数据证明了 TP53Mut AML 独特的信号激活状态。信号分子对下游靶点的影响可能因细胞环境而异。因此,我们想知道在 TP53Mut 和 TP53WT LSCs 之间,被激活的信号分子的影响和相互作用是否有所不同。为了建立信号结构模型,我们采用了贝叶斯网络建模,这是一种机器学习算法,已被证实是一种发现信号分子间统计相关性和依赖性的方法。我们在 TP53Mut 和 TP53WT AMLs 中发现了相似的全局信号网络结构和层次。然而,TP53Mut模型显示NFkB对磷酸化组蛋白H3(pHIS3)和Ki67有很大的影响,这表明TP53Mut LSCs的增殖依赖于NFkB水平。此外,TP53Mut LSCs 中的 NFkB 水平高度依赖 pSTAT1。接下来,我们比较了 BEAT AML 数据集中 TP53Mut 和 TP53WT 样本的转录谱,发现 TP53Mut AML 显示出分子特征数据库中所有 NFkB 基因组(n=19)的显著富集,为 TP53Mut 人类 AML 对 NFkB 信号的独特依赖提供了更多证据。因此,我们使用慢病毒转导的 shRNA 构建物来确定突变 p53 对人类 AML 信号转导的影响。我们发现,敲除 TP53Mut 能降低 Kasumi AML 细胞(TP53R248Q/-)的 NFkB 蛋白水平,但不能降低 MOLM13 AML 细胞(TP53WT)的 NFkB 蛋白水平。转导的 Kasumi 细胞的 RNA 测序显示,TP53Mut 基因敲除导致 NFkB 和 LSC 自我更新特征丧失。在原代人类 TP53Mut AMLs(S127F/- 和 S241Y/-)中敲除 TP53Mut 也会导致这些原代样本中 NFkB 和 LSC 特征的类似丧失。TP53Mut基因敲除后,Kasumi细胞和TP53Mut原代人类AML的集落形成也会减弱。这些数据显示 TP53Mut 促进了人类 AML 中 NFkB 的激活,并表明人类 TP53Mut LSCs 的自我更新可能依赖于 NFkB。这些数据表明,NFkB可能是针对TP53Mut人LSCs的一种治疗策略。引用格式:Daniel Chang, Klara Noble-Orcutt, Marie Lue Antony, Yoonkyu Lee, Fiona He, Karen Sachs, Chad L Myers, Zohar Sachs.贝叶斯网络建模确定了 TP53 突变 AML 对 NF kappa B 信号的依赖性 [摘要].在:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P34.
{"title":"Abstract P34: Bayesian networks modeling identifies a reliance of TP53 mutant AML on NF kappa B signaling","authors":"Daniel Chang, Klara E Noble-Orcutt, Marie L Antony, Yoonkyu Lee, Fiona He, Karen Sachs, Chad L Myers, Z. Sachs","doi":"10.1158/2643-3249.bcdsymp24-p34","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p34","url":null,"abstract":"\u0000 In acute myeloid leukemia (AML), TP53 mutations ( TP53Mut ) confer the worst prognosis. Leukemia stem cells (LSCs) are rare AML cells endowed with self-renewal capacity, allowing them to recapitulate leukemia after therapy and cause disease progression. In human AML, signaling pathways associated with inflammation have been implicated in the pathogenesis of TP53Mut AML, but the pathways that mediate self-renewal in human TP53Mut AML are not well-known. To define the signaling pathways that are activated in TP53Mut human LSCs, we used CyTOF, a form of flow cytometry that can measure up to 40 proteins simultaneously at single-cell resolution. We compared the levels of activated signaling molecules in the LSC-enriched (CD34+CD38−) subset of seven TP53Mut and seven TP53 wild-type ( TP53WT ) AML samples. Phosphorylated NF kappa B (pNFkB), pSTAT1, and pP38 are significantly higher in TP53Mut AMLs. In contrast, Ki67 and pMAPKAPKII are higher in TP53WT AML. These data demonstrate a unique signaling activation state in TP53Mut AML. The influence of signaling molecules on downstream targets can vary by cellular context. Therefore, we asked whether the influence and interaction of the activated signaling molecules vary between TP53Mut and TP53WT LSCs. To model the signaling architecture, we performed Bayesian networks modeling, a machine learning algorithm that has been validated as a method to discover statistical correlations and dependencies between signaling molecules. We found a similar global signaling network structure and hierarchy in TP53Mut and TP53WT AMLs. However, TP53Mut model showed NFkB have strong influences on phosphorylated Histone H3 (pHIS3) and Ki67, suggesting that proliferation depends on NFkB levels in TP53Mut LSCs. Furthermore, NFkB levels were highly dependent on pSTAT1 in TP53Mut LSCs. Next, we compared the transcriptional profiles of TP53Mut and TP53WT samples in the BEAT AML dataset and found that TP53Mut AML displayed significant enrichment of all the NFkB gene sets (n=19) in the molecular signatures database, providing more evidence for a unique reliance of TP53Mut human AML on NFkB signaling. Therefore, we used lentivirus transduced shRNA constructs to determine the influence of mutant p53 on signaling in human AML. We found that knockdown of TP53Mut reduced NFkB protein levels in Kasumi AML cells (which are TP53R248Q/ − ) but not in MOLM13 AML cells (which are TP53WT). RNA sequencing of transduced Kasumi cells showed that TP53Mut knockdown led to loss of NFkB and LSC self-renewal signatures. Knockdown of TP53Mut in primary human TP53Mut AMLs (S127F/- and S241Y/-) led to a similar loss of NFkB and LSC signatures in these primary samples as well. Colony formation in Kasumi cells and TP53Mut primary human AML were also abrogated after TP53Mut knockdown. These data show TP53Mut promotes NFkB activation in human AML and suggests that human TP53Mut LSCs may be dependent on NFkB for self-renewal. These data implicate NFkB as a pos","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140080060","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}
引用次数: 0
Abstract P21: VISTA a potential new Immuno-oncology therapeutic target to treat human Acute Myeloid Leukemia 摘要 P21:VISTA--治疗人类急性髓性白血病的潜在免疫肿瘤学新靶点
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p21
Thierry Guillaudeux, S. Sridhar, E. Frazier, Yulia Ovechkina, Shawn Iadonato
V-domain Immunoglobulin Suppressor of T cell Activation (VISTA/PD-1H) is a co-inhibitor molecule of the B7 family member highly expressed on circulating and intra-tumoral myeloid cells. VISTA is a negative checkpoint protein that inhibits anti-tumor T cell responses. VISTA expression has been associated with poor overall survival in multiple solid tumors and is also described as a potential mediator of resistance to anti-CLTA-4 and anti-PD1 therapies. VISTA expression was also investigated in hematological tumors and described as highly expressed on blasts including leukemic stem cells from AML patients. Therefore, VISTA is a potentially new therapeutic target for cancer immunotherapy in this indication. Kineta has developed a fully human monoclonal antibody targeting VISTA, KVA12123, currently evaluated in a Phase 1/2 clinical trial alone and in combination with pembrolizumab in cancer patients with advanced solid tumors. Supported with previous published data on VISTA expression and function in myeloid leukemia, we also believe that our lead therapeutic anti-VISTA antibody could be a promising drug for AML patients. Therefore, we have tested in vivo, in a Kasumi-3 AML disseminated tumor model, the efficacy of our antibody alone or in combination with standard of care therapy, cytarabine and doxorubicin. The Kasumi-3 cell line exhibits a high level of VISTA expression on its cell surface, like leukemic cells in AML patients. Female NOD-scid mice were first inoculated with Kasumi cells via tail vein injection, and tumor engraftment was evaluated at different time points in the blood, spleen and bone marrow to monitor engraftment using CD34, CD33, CD45 and HLA-DR cell surface expression markers. Then, when tumor burden was high, standard of care treatment (5 doses of 16mg/kg AraC by IV injections and 3 doses of 0.5mg/kg Dox by IV injections) was initiated in indicated groups for 5 cycles, before starting treatment with KVA12 for the rest of the study. Tumor engraftment was tested after chemotherapy alone to evaluate the efficacy before starting treatment with our antibody. Our results showed that untreated mice had higher number of Kasumi-3 cells in the blood when compared to chemotherapy treated mice. Two backbone versions of our antibody were then tested, a human IgG1 – KVA12123 and a human IgG4 - KVA12402, at 30mg/kg IP injections twice a week for 5 weeks. Our data showed that KVA12123 and KVA12402 as single agents and in combination with chemotherapy reduced Kasumi-3 tumor load in the blood, spleen and bone marrow within the groups and significantly improved survival when compared to IgG1 control groups. These data indicate that anti-VISTA targeted therapy should be evaluated in myeloproliferative disorders like AML alone or in combination with standard of care. Citation Format: Thierry Guillaudeux, Shaarwari Sridhar, Emily Frazier, Yulia Ovechkina, Shawn Iadonato. VISTA a potential new Immuno-oncology therapeutic target to treat human Acut
V 域免疫球蛋白 T 细胞活化抑制因子(VISTA/PD-1H)是 B7 家族成员的共同抑制分子,在循环和肿瘤内髓系细胞上高度表达。VISTA 是一种抑制抗肿瘤 T 细胞反应的阴性检查点蛋白。VISTA 的表达与多种实体瘤的总生存率低下有关,也被描述为抗CLTA-4 和抗 PD1 疗法耐药性的潜在介质。VISTA 在血液肿瘤中的表达也得到了研究,并被描述为在急性髓细胞性白血病患者的囊泡包括白血病干细胞中高度表达。因此,在这一适应症中,VISTA 有可能成为癌症免疫疗法的新治疗靶点。Kineta 已开发出一种靶向 VISTA 的全人源单克隆抗体 KVA12123,目前正在 1 1/2 期临床试验中进行评估,该抗体可单独使用,也可与 pembrolizumab 联合使用,用于晚期实体瘤癌症患者。根据之前发表的有关 VISTA 在髓细胞白血病中的表达和功能的数据,我们也相信我们的先导治疗性抗 VISTA 抗体可能是治疗急性髓细胞白血病患者的一种很有前景的药物。因此,我们在 Kasumi-3 AML 播散性肿瘤模型中测试了我们的抗体单独使用或与标准疗法(阿糖胞苷和多柔比星)联合使用的疗效。Kasumi-3 细胞系与急性髓细胞白血病患者的白血病细胞一样,其细胞表面也有高水平的 VISTA 表达。首先通过尾静脉注射将 Kasumi 细胞接种到雌性 NOD-scid 小鼠体内,然后在不同的时间点评估血液、脾脏和骨髓中的肿瘤吞噬情况,利用 CD34、CD33、CD45 和 HLA-DR 细胞表面表达标记监测吞噬情况。然后,当肿瘤负荷较高时,在指定组开始标准治疗(静脉注射5次16毫克/千克的AraC和静脉注射3次0.5毫克/千克的Dox),共5个周期,然后开始使用KVA12治疗研究的其余部分。在开始使用我们的抗体进行治疗之前,先对单纯化疗后的肿瘤移植情况进行检测,以评估疗效。结果显示,与接受化疗的小鼠相比,未接受化疗的小鼠血液中的 Kasumi-3 细胞数量更高。随后,我们测试了两种骨架抗体,一种是人 IgG1 - KVA12123,另一种是人 IgG4 - KVA12402,每周两次,每次 30 毫克/千克 IP 注射,持续 5 周。我们的数据显示,与 IgG1 对照组相比,KVA12123 和 KVA12402 作为单药或与化疗联合使用可减少组内血液、脾脏和骨髓中的 Kasumi-3 肿瘤负荷,并显著提高生存率。这些数据表明,抗VISTA靶向疗法应单独或与标准疗法联合用于评估骨髓增生性疾病(如急性髓细胞性白血病)。引用格式:Thierry Guillaudeux, Shaarwari Sridhar, Emily Frazier, Yulia Ovechkina, Shawn Iadonato.治疗人类急性髓性白血病的潜在新免疫肿瘤治疗靶点 VISTA [摘要]。在:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P21.
{"title":"Abstract P21: VISTA a potential new Immuno-oncology therapeutic target to treat human Acute Myeloid Leukemia","authors":"Thierry Guillaudeux, S. Sridhar, E. Frazier, Yulia Ovechkina, Shawn Iadonato","doi":"10.1158/2643-3249.bcdsymp24-p21","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p21","url":null,"abstract":"\u0000 V-domain Immunoglobulin Suppressor of T cell Activation (VISTA/PD-1H) is a co-inhibitor molecule of the B7 family member highly expressed on circulating and intra-tumoral myeloid cells. VISTA is a negative checkpoint protein that inhibits anti-tumor T cell responses. VISTA expression has been associated with poor overall survival in multiple solid tumors and is also described as a potential mediator of resistance to anti-CLTA-4 and anti-PD1 therapies. VISTA expression was also investigated in hematological tumors and described as highly expressed on blasts including leukemic stem cells from AML patients. Therefore, VISTA is a potentially new therapeutic target for cancer immunotherapy in this indication. Kineta has developed a fully human monoclonal antibody targeting VISTA, KVA12123, currently evaluated in a Phase 1/2 clinical trial alone and in combination with pembrolizumab in cancer patients with advanced solid tumors. Supported with previous published data on VISTA expression and function in myeloid leukemia, we also believe that our lead therapeutic anti-VISTA antibody could be a promising drug for AML patients. Therefore, we have tested in vivo, in a Kasumi-3 AML disseminated tumor model, the efficacy of our antibody alone or in combination with standard of care therapy, cytarabine and doxorubicin. The Kasumi-3 cell line exhibits a high level of VISTA expression on its cell surface, like leukemic cells in AML patients. Female NOD-scid mice were first inoculated with Kasumi cells via tail vein injection, and tumor engraftment was evaluated at different time points in the blood, spleen and bone marrow to monitor engraftment using CD34, CD33, CD45 and HLA-DR cell surface expression markers. Then, when tumor burden was high, standard of care treatment (5 doses of 16mg/kg AraC by IV injections and 3 doses of 0.5mg/kg Dox by IV injections) was initiated in indicated groups for 5 cycles, before starting treatment with KVA12 for the rest of the study. Tumor engraftment was tested after chemotherapy alone to evaluate the efficacy before starting treatment with our antibody. Our results showed that untreated mice had higher number of Kasumi-3 cells in the blood when compared to chemotherapy treated mice. Two backbone versions of our antibody were then tested, a human IgG1 – KVA12123 and a human IgG4 - KVA12402, at 30mg/kg IP injections twice a week for 5 weeks. Our data showed that KVA12123 and KVA12402 as single agents and in combination with chemotherapy reduced Kasumi-3 tumor load in the blood, spleen and bone marrow within the groups and significantly improved survival when compared to IgG1 control groups. These data indicate that anti-VISTA targeted therapy should be evaluated in myeloproliferative disorders like AML alone or in combination with standard of care.\u0000 Citation Format: Thierry Guillaudeux, Shaarwari Sridhar, Emily Frazier, Yulia Ovechkina, Shawn Iadonato. VISTA a potential new Immuno-oncology therapeutic target to treat human Acut","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140266100","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}
引用次数: 0
Abstract P13: Genetic and epigenetic characterization of B-cell chronic lymphocytic leukemia with IG::BCL3-translocation provides evidence for a distinct biological entity 摘要 P13:IG::BCL3转位的B细胞慢性淋巴细胞白血病的遗传学和表观遗传学特征为一个独特的生物学实体提供了证据
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p13
Cosima Drewes, Cristina López, Nnamdi Okeke, S. Hillebrecht, Petra Schütz, Anja Fischer, Anja Mottok, S. Bens, C. Schneider, S. Stilgenbauer, E. Tausch, Reiner Siebert
IG translocations occur in various B-cell neoplasms and result in the deregulation of (onco)genes due to their juxtaposition with regulatory elements in the immunoglobulin loci, predominantly the immunoglobulin heavy chain (IGH) locus (14q32). Some of these IGH translocations are pathognomonic for distinct disease entities, like those involving MYC, BCL2 or CCND1 in Burkitt, Follicular or Mantle Cell Lymphomas, respectively. In Chronic Lymphocytic Leukemia (CLL), the BCL3 gene (19q13), a transcriptional coactivator of the nuclear factor-kappaB (NF-κB) family, is a known partner gene in these translocations. Two subsets of IG::BCL3 translocation-positive B-cell neoplasms have been described that exhibit differences in chromosomal aberrations, IGHV mutation status, and histopathology [Martín-Subero et al. 2007]. In CLL, IG::BCL3 translocation is linked to a younger age at diagnosis, a more aggressive clinical course, and an atypical tumor cell phenotype [Michaux et al. 1997; Au et al. 2002]. A total of 89 B-cell neoplasms, primarily diagnosed as CLL, displaying IG::BCL3 translocation detected by fluorescence in situ hybridization (FISH), were examined. Analysis of DNA methylation and copy number aberrations (CNA) was conducted for 86 IGH::BCL3-translocated B-cell neoplasms using 850K EPIC BeadChip array data. Evaluation of chromosomal aberrations typical for CLL was moreover performed by FISH and the IGHV mutation status was determined. IGH::BCL3 breakpoint junctions were sequenced in 23 samples using targeted capture and whole-genome sequencing approaches. RNA expression of BCL3 was assessed using the HTG mRNA pan B-cell panel. In UMAP analysis of DNA methylation, CLLs with IG::BCL3 translocation exhibited segregation from other CLLs, irrespective of IGH translocations. Supervised analysis revealed global hypomethylation of B-cell neoplasms with IG::BCL3 translocation compared to other CLL. A total of 66/69 CLLs with IG::BCL3 translocation showed an unmutated IGHV status (96%). Copy number determination using BeadChip data (n=86) and FISH analysis (n=85) showed a significantly higher incidence of trisomy 12 in IG::BCL3-translocated cases compared to the general CLL population (p=0.011). Breakpoint sequencing indicated IG::BCL3 junctions to be recurrently associated with aberrant class-switch recombination at the IGH locus, involving IGHA (n=11/23), IGHG (n=5/23) and IGHM segments (n=5/23). Breakpoints at the BCL3 locus displayed two groups. 80 cases (all CLL) were located upstream of BCL3 and two other B-cell neoplasms (NHL, DLBCL) were located downstream of BCL3. BCL3 expression analysis in 15 IG::BCL3-translocated CLLs confirmed its transcriptional upregulation compared to the general CLL population. Our genetic and epigenetic analyses support the view that CLL with IG::BCL3 translocation may constitute a genetically and epigenetically distinct subtype of B-cell neoplasms, diverging from the typical CLL. Citation Format: Cosima Drewes, Cristi
IG 易位发生在各种 B 细胞肿瘤中,由于它们与免疫球蛋白基因座(主要是免疫球蛋白重链(IGH)基因座(14q32))中的调控元件并置,导致(onco)基因失调。其中一些 IGH 易位是不同疾病实体的病理标志,如伯基特淋巴瘤、滤泡细胞淋巴瘤或套细胞淋巴瘤中分别涉及 MYC、BCL2 或 CCND1 的易位。在慢性淋巴细胞白血病(CLL)中,BCL3 基因(19q13)是核因子-卡巴(NF-κB)家族的转录辅激活因子,是这些易位的已知伙伴基因。目前已描述了两个 IG::BCL3 易位阳性 B 细胞肿瘤亚群,它们在染色体畸变、IGHV 突变状态和组织病理学方面表现出差异[Martín-Subero 等人,2007 年]。在 CLL 中,IG::BCL3 易位与较年轻的诊断年龄、更具侵袭性的临床过程和非典型肿瘤细胞表型有关 [Michaux 等人,1997 年;Au 等人,2002 年]。本文共研究了 89 例 B 细胞肿瘤,主要诊断为 CLL,通过荧光原位杂交(FISH)检测到 IG::BCL3 易位。利用 850K EPIC BeadChip 阵列数据对 86 例 IGH::BCL3 易位的 B 细胞肿瘤进行了 DNA 甲基化和拷贝数畸变(CNA)分析。此外,还通过 FISH 对 CLL 典型的染色体畸变进行了评估,并确定了 IGHV 突变状态。采用靶向捕获和全基因组测序方法对 23 个样本中的 IGH::BCL3 断点连接进行了测序。使用 HTG mRNA pan B 细胞面板评估了 BCL3 的 RNA 表达。在DNA甲基化的UMAP分析中,无论是否存在IGH易位,具有IG::BCL3易位的CLL都表现出与其他CLL的分离。监督分析显示,与其他 CLL 相比,IG::BCL3 易位的 B 细胞肿瘤的甲基化水平较低。在66/69例有IG::BCL3易位的CLL中,共有96%显示出未突变的IGHV状态。利用 BeadChip 数据(n=86)和 FISH 分析(n=85)进行的拷贝数测定显示,与普通 CLL 患者相比,IG::BCL3 易位病例中 12 三体综合征的发生率明显更高(p=0.011)。断点测序显示,IG::BCL3连接点经常与IGH基因座的异常类开关重组相关,涉及IGHA(n=11/23)、IGHG(n=5/23)和IGHM片段(n=5/23)。BCL3 基因座上的断点显示为两组。80例(均为CLL)位于BCL3的上游,另外两例B细胞肿瘤(NHL、DLBCL)位于BCL3的下游。在15例IG::BCL3转移的CLL中进行的BCL3表达分析证实,与普通CLL人群相比,BCL3转录上调。我们的遗传学和表观遗传学分析支持这样一种观点,即带有IG::BCL3易位的CLL可能是B细胞肿瘤中遗传学和表观遗传学上不同的亚型,有别于典型的CLL。引用格式:Cosima Drewes, Cristina López, Nnamdi Okeke, Sina Hillebrecht, Petra Schütz, Anja Fischer, Anja Mottok, Susanne Bens, Christof Schneider, Stephan Stilgenbauer, Eugen Tausch, Reiner Siebert.具有IG::BCL3转位的B细胞慢性淋巴细胞白血病的遗传学和表观遗传学特征为一个独特的生物学实体提供了证据[摘要]。见:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P13.
{"title":"Abstract P13: Genetic and epigenetic characterization of B-cell chronic lymphocytic leukemia with IG::BCL3-translocation provides evidence for a distinct biological entity","authors":"Cosima Drewes, Cristina López, Nnamdi Okeke, S. Hillebrecht, Petra Schütz, Anja Fischer, Anja Mottok, S. Bens, C. Schneider, S. Stilgenbauer, E. Tausch, Reiner Siebert","doi":"10.1158/2643-3249.bcdsymp24-p13","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p13","url":null,"abstract":"\u0000 IG translocations occur in various B-cell neoplasms and result in the deregulation of (onco)genes due to their juxtaposition with regulatory elements in the immunoglobulin loci, predominantly the immunoglobulin heavy chain (IGH) locus (14q32). Some of these IGH translocations are pathognomonic for distinct disease entities, like those involving MYC, BCL2 or CCND1 in Burkitt, Follicular or Mantle Cell Lymphomas, respectively. In Chronic Lymphocytic Leukemia (CLL), the BCL3 gene (19q13), a transcriptional coactivator of the nuclear factor-kappaB (NF-κB) family, is a known partner gene in these translocations. Two subsets of IG::BCL3 translocation-positive B-cell neoplasms have been described that exhibit differences in chromosomal aberrations, IGHV mutation status, and histopathology [Martín-Subero et al. 2007]. In CLL, IG::BCL3 translocation is linked to a younger age at diagnosis, a more aggressive clinical course, and an atypical tumor cell phenotype [Michaux et al. 1997; Au et al. 2002]. A total of 89 B-cell neoplasms, primarily diagnosed as CLL, displaying IG::BCL3 translocation detected by fluorescence in situ hybridization (FISH), were examined. Analysis of DNA methylation and copy number aberrations (CNA) was conducted for 86 IGH::BCL3-translocated B-cell neoplasms using 850K EPIC BeadChip array data. Evaluation of chromosomal aberrations typical for CLL was moreover performed by FISH and the IGHV mutation status was determined. IGH::BCL3 breakpoint junctions were sequenced in 23 samples using targeted capture and whole-genome sequencing approaches. RNA expression of BCL3 was assessed using the HTG mRNA pan B-cell panel. In UMAP analysis of DNA methylation, CLLs with IG::BCL3 translocation exhibited segregation from other CLLs, irrespective of IGH translocations. Supervised analysis revealed global hypomethylation of B-cell neoplasms with IG::BCL3 translocation compared to other CLL. A total of 66/69 CLLs with IG::BCL3 translocation showed an unmutated IGHV status (96%). Copy number determination using BeadChip data (n=86) and FISH analysis (n=85) showed a significantly higher incidence of trisomy 12 in IG::BCL3-translocated cases compared to the general CLL population (p=0.011). Breakpoint sequencing indicated IG::BCL3 junctions to be recurrently associated with aberrant class-switch recombination at the IGH locus, involving IGHA (n=11/23), IGHG (n=5/23) and IGHM segments (n=5/23). Breakpoints at the BCL3 locus displayed two groups. 80 cases (all CLL) were located upstream of BCL3 and two other B-cell neoplasms (NHL, DLBCL) were located downstream of BCL3. BCL3 expression analysis in 15 IG::BCL3-translocated CLLs confirmed its transcriptional upregulation compared to the general CLL population. Our genetic and epigenetic analyses support the view that CLL with IG::BCL3 translocation may constitute a genetically and epigenetically distinct subtype of B-cell neoplasms, diverging from the typical CLL.\u0000 Citation Format: Cosima Drewes, Cristi","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140265498","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}
引用次数: 0
Abstract P01: Impact of Germline and Somatic ATM Variants in Chronic Lymphocytic Leukemia (CLL): Clinical Implications and Response to PARP Inhibition 摘要 P01:种系和体细胞 ATM 变异对慢性淋巴细胞白血病 (CLL) 的影响:临床意义及对PARP抑制剂的反应
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p01
Kiyomi Mashima, Nicholas S. Moore, M. Mikhaleva, Anna Petráčková, Samantha Shupe, Stacey M Fernandes, A. Taylor-Weiner, R. Gillani, Hoyin Chu, Seunghun Han, S. Camp, Eric Kofman, G. Getz, Catherine J. Wu, S. Tyekucheva, M. Davids, E. V. Van Allen, S. AlDubayan, Jennifer R. Brown
Background: Next-generation sequencing has revealed numerous Variants of Unknown Significance (VUS) in the ATM gene, important in DNA repair and chronic lymphocytic leukemia (CLL) pathogenesis. This study evaluates the effect of rare ATM germline variants, including VUS, on features of CLL, ATM kinase activity, and response to PARP inhibition. Methods:ATM variants were detected in 885 CLL patients (327 DFCI; 276 German CLL Study Group; 282 ICGC) using DeepVariant, and compared to 5310 ancestry-matched controls. Concurrently, 278 CLL patients with ATM aberrations from clinical sequencing were retrospectively analyzed, annotated by population allele frequency (JCO 2023, Lampson et al.). ATM kinase activity was measured by ATM and KAP1 phosphorylation following gamma-irradiation in primary CLL samples. The impact of talazoparib, a PARP inhibitor, on ATM aberrant and wild-type CLL cell proliferation was also assessed. Results: Among the 885 CLL cases, 130 ATM germline variants were identified by DeepVariant, including 87 missense variants. The top 5 most frequent rare germline missense variants with high odds ratio for association with CLL were ATM p.L2332P (AF cases vs controls, 0.328% vs 0.0188%, OR 17.46), p.L2307F (OR 16.88), p.F763L (OR 18.89), p.Y1442H (OR 35.39) and p.S99G (OR 16.28). In our retrospective clinical cohort, we identified 294 ATM variants with 146 unique variants. Our previously proposed algorithm for classifying germline vs somatic identified 74 germline, with 70 missense, and 72 somatic. Phosphorylation of ATM and KAP1 (pATM, pKAP1) in response to IR was reduced in 13 patients assessed with germline ATM variants (9 rare and 4 non-rare; ATM variant alone group) compared to those without any somatic or germline ATM aberrancy (WT group) (P<0.01). Phosphorylation levels were also significantly lower in 17 patients with rare germline ATM variants with concurrent del(11q) (del11q + ATM germline variant group) compared to 15 with del(11q) alone (del 11q group) (P<0.01). We obtained similar results from Western blot analysis. To assess the sensitivity of ATM deficient cells to talazoparib, we compared proliferation inhibition across groups cocultured with talazoparib for 14 days. The del11q with ATM-somatic group exhibited significantly greater sensitivity to talazoparib at lower dosages compared to ATM WT (talazoparib 0.1 μM, P<0.05; 0.5μM, P=0.098). The monoallelic ATM deficient groups (del11q alone or ATM-germline alone) tended to have lower proliferation, but not significantly. Higher concentrations of talazoparib (2.5μM) strongly inhibited proliferation of all CLLs including WT. Conclusion: This study highlights the prevalence of rare ATM germline variants in CLL and their impact on ATM function in CLL. Our findings also suggest that these variants affect the DNA damage response in primary CLL cells and that ATM biallelic deficiency influences sensitivity to PARP inhibitors. Our evidence could lead to more personalized therapeut
背景:下一代测序发现了ATM基因中的许多未知意义变异(VUS),这些变异在DNA修复和慢性淋巴细胞白血病(CLL)发病机制中非常重要。本研究评估了包括VUS在内的罕见ATM种系变异对CLL特征、ATM激酶活性以及对PARP抑制剂反应的影响。方法:使用 DeepVariant 检测了 885 例 CLL 患者(327 例 DFCI;276 例德国 CLL 研究组;282 例 ICGC)的 ATM 变异,并与 5310 例血缘匹配对照进行了比较。同时,对临床测序中出现 ATM 畸变的 278 例 CLL 患者进行了回顾性分析,并按人群等位基因频率进行了注释(JCO 2023,Lampson 等人)。在原发性 CLL 样本中,通过伽马射线照射后的 ATM 和 KAP1 磷酸化测量 ATM 激酶活性。还评估了 PARP 抑制剂 Talazoparib 对 ATM 畸变和野生型 CLL 细胞增殖的影响。研究结果在 885 个 CLL 病例中,DeepVariant 发现了 130 个 ATM 基因变异,包括 87 个错义变异。与 CLL 关联几率最高的前 5 个罕见种系错义变异是:ATM p.L2332P(AF 病例 vs 对照,0.328% vs 0.0188%,OR 17.46)、p.L2307F(OR 16.88)、p.F763L(OR 18.89)、p.Y1442H(OR 35.39)和 p.S99G(OR 16.28)。在我们的回顾性临床队列中,我们发现了 294 个 ATM 变异,其中有 146 个独特变异。我们之前提出的种系变异与体细胞变异分类算法发现了 74 个种系变异,其中 70 个为错义变异,72 个为体细胞变异。与没有任何体细胞或种系ATM变异的患者(WT组)相比,13例评估为ATM种系变异的患者(9例罕见,4例非罕见;仅有ATM变异组)的ATM和KAP1(pATM、pKAP1)对IR的磷酸化反应降低(P<0.01)。17例罕见种系ATM变异同时伴有del(11q)的患者(del11q + ATM种系变异组)的磷酸化水平也明显低于15例仅伴有del(11q)的患者(del 11q组)(P<0.01)。我们从 Western 印迹分析中得到了类似的结果。为了评估ATM缺陷细胞对talazoparib的敏感性,我们比较了与talazoparib共培养14天的各组细胞的增殖抑制情况。与ATM WT相比,del11q伴ATM异常组在较低剂量下对talazoparib的敏感性明显更高(talazoparib 0.1μM,P<0.05;0.5μM,P=0.098)。单倍性ATM缺陷组(单独del11q或单独ATM-种系)的增殖往往较低,但并不显著。较高浓度的talazoparib(2.5μM)可强烈抑制包括WT在内的所有CLL的增殖。结论这项研究强调了罕见的ATM种系变异在CLL中的普遍性及其对CLL中ATM功能的影响。我们的研究结果还表明,这些变异会影响原发性 CLL 细胞的 DNA 损伤反应,ATM 双倍序列缺陷会影响对 PARP 抑制剂的敏感性。我们的证据可为具有不同ATM变异的CLL患者提供更个性化的治疗策略。引用格式:Kiyomi Mashima, Nicholas Moore, Mariia Mikhaleva, Anna Petráčková, Samantha Shupe, Stacey M Fernandes, Amaro Taylor-Weiner, Riaz Gillani, Hoyin Chu, Seunghun Han, Sabrina Camp, Eric Kofman, Gad Getz, Catherine J. Wu, Svitlana Tyekucheva, Matthew S Davids, Eliezer Mendel Van Allen, Saud AlDubayan, Jennifer R Brown.慢性淋巴细胞白血病(CLL)中种系和体细胞 ATM 变异的影响:临床意义及对 PARP 抑制的反应 [摘要]。In:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P01.
{"title":"Abstract P01: Impact of Germline and Somatic ATM Variants in Chronic Lymphocytic Leukemia (CLL): Clinical Implications and Response to PARP Inhibition","authors":"Kiyomi Mashima, Nicholas S. Moore, M. Mikhaleva, Anna Petráčková, Samantha Shupe, Stacey M Fernandes, A. Taylor-Weiner, R. Gillani, Hoyin Chu, Seunghun Han, S. Camp, Eric Kofman, G. Getz, Catherine J. Wu, S. Tyekucheva, M. Davids, E. V. Van Allen, S. AlDubayan, Jennifer R. Brown","doi":"10.1158/2643-3249.bcdsymp24-p01","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p01","url":null,"abstract":"\u0000 Background: Next-generation sequencing has revealed numerous Variants of Unknown Significance (VUS) in the ATM gene, important in DNA repair and chronic lymphocytic leukemia (CLL) pathogenesis. This study evaluates the effect of rare ATM germline variants, including VUS, on features of CLL, ATM kinase activity, and response to PARP inhibition. Methods:ATM variants were detected in 885 CLL patients (327 DFCI; 276 German CLL Study Group; 282 ICGC) using DeepVariant, and compared to 5310 ancestry-matched controls. Concurrently, 278 CLL patients with ATM aberrations from clinical sequencing were retrospectively analyzed, annotated by population allele frequency (JCO 2023, Lampson et al.). ATM kinase activity was measured by ATM and KAP1 phosphorylation following gamma-irradiation in primary CLL samples. The impact of talazoparib, a PARP inhibitor, on ATM aberrant and wild-type CLL cell proliferation was also assessed. Results: Among the 885 CLL cases, 130 ATM germline variants were identified by DeepVariant, including 87 missense variants. The top 5 most frequent rare germline missense variants with high odds ratio for association with CLL were ATM p.L2332P (AF cases vs controls, 0.328% vs 0.0188%, OR 17.46), p.L2307F (OR 16.88), p.F763L (OR 18.89), p.Y1442H (OR 35.39) and p.S99G (OR 16.28). In our retrospective clinical cohort, we identified 294 ATM variants with 146 unique variants. Our previously proposed algorithm for classifying germline vs somatic identified 74 germline, with 70 missense, and 72 somatic. Phosphorylation of ATM and KAP1 (pATM, pKAP1) in response to IR was reduced in 13 patients assessed with germline ATM variants (9 rare and 4 non-rare; ATM variant alone group) compared to those without any somatic or germline ATM aberrancy (WT group) (P<0.01). Phosphorylation levels were also significantly lower in 17 patients with rare germline ATM variants with concurrent del(11q) (del11q + ATM germline variant group) compared to 15 with del(11q) alone (del 11q group) (P<0.01). We obtained similar results from Western blot analysis. To assess the sensitivity of ATM deficient cells to talazoparib, we compared proliferation inhibition across groups cocultured with talazoparib for 14 days. The del11q with ATM-somatic group exhibited significantly greater sensitivity to talazoparib at lower dosages compared to ATM WT (talazoparib 0.1 μM, P<0.05; 0.5μM, P=0.098). The monoallelic ATM deficient groups (del11q alone or ATM-germline alone) tended to have lower proliferation, but not significantly. Higher concentrations of talazoparib (2.5μM) strongly inhibited proliferation of all CLLs including WT. Conclusion: This study highlights the prevalence of rare ATM germline variants in CLL and their impact on ATM function in CLL. Our findings also suggest that these variants affect the DNA damage response in primary CLL cells and that ATM biallelic deficiency influences sensitivity to PARP inhibitors. Our evidence could lead to more personalized therapeut","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140080275","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}
引用次数: 0
Abstract P25: Identification of biological components and novel clinical subsets of NPM1-mutated AML using an integrated, multi-omics approach 摘要 P25:利用综合多组学方法鉴定 NPM1 基因突变急性髓细胞性白血病的生物成分和新型临床亚群
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p25
Salma Abdelbaky, Kyoko Yamaguchi, Yue-Zhong Wu, Kevin R. Coombes, Lianbo Yu, Christopher C Oakes
Acute myeloid leukemia is the most common acute leukemia in adults with a 5-year overall survival rate of only 28%. Relapse occurs in almost half of adults within 3 years. Due to a high degree of heterogeneity in biological and genetic features in AML tumor cells, current prognostic markers (including genetic aberrations) do not fully explain the ranges of phenotype and outcomes observed in AML patients. Recently, we characterized 13 DNA methylation signatures (epitypes) and a STAT hypomethylation signature (SHS) to be predictive of outcome and stable at relapse. Here, we used the integrative Multi-omics Factor Analysis (MOFA) approach to combine genetic, cytogenetic, transcriptomic, and our novel epigenetic information in an extensive multi-omics analysis to infer latent factors capable of explaining independent signatures comprising NPM1-mutated AML biology. The study used the well-annotated CALGB/Alliance AML cohort encompassing 581 NPM1-mutated patients. The analysis showed that RNA-sequencing contributed the most to explaining the variance between patients followed by RNA splicing, highlighting the depth of information present in the transcriptome. Surprisingly, genetic mutations contributed to only approximately 3% of the overall variance, while DNA methylation captured more than 15% of the total variance. We were able to infer 15 latent factors, 5 of which were associated with overall survival, event-free survival, and attainment of complete remission. All factors were significant after adjusting for established prognostic features, such as FLT3-ITD, sex, and age. Factors 2, 7 and 11 were associated with unfavorable outcomes, and variably included increased HOX signatures, suppressed TP53 activation, a stem cell-like phenotype, the epigenetic SHS signature, triple NPM1/FLT3-ITD/DNMT3A mutations, and alternative splicing, including NUP98. Factors 4 and 13 had a favorable prognostic value. Factor 13 uncovered a unique expression pattern of X-linked cancer testis antigen gene family members dividing NPM1 patients independent of sex, age, or common mutations. The factor was inversely associated with the expression of CD34, MN1 and BAALC – prognostic genes related to stemness. Analysis of immune cell subsets indicated differences in proportions of CD8 effector T cell populations between patients with high vs. low factor 13. This feature pattern and clinical significance were validated in an independent cohort (Beat-AML). Lastly, we clustered the 15 generated factors and identified 5 subsets of NPM1-mutated patients – 2 clusters with a favorable prognostic value, 2 clusters with unfavorable prognostic value, and one intermediate cluster. Overall, we identify 15 main sources of variance within NPM1-mutated patients, including 5 clinically-relevant factors associated with unique biological features, which together generate unique independent biomarker signatures not observed when individually considering separate omics data types. Citation Form
急性髓性白血病是成人中最常见的急性白血病,5 年总生存率仅为 28%。几乎一半的成人在 3 年内会复发。由于急性髓细胞白血病肿瘤细胞的生物和遗传特征具有高度异质性,目前的预后标志物(包括遗传畸变)并不能完全解释急性髓细胞白血病患者的表型和预后范围。最近,我们鉴定了 13 个 DNA 甲基化特征(表型)和一个 STAT 低甲基化特征(SHS),它们可预测预后并在复发时保持稳定。在这里,我们采用了多组学因素分析(MOFA)的综合方法,在广泛的多组学分析中结合了遗传学、细胞遗传学、转录组学和我们的新型表观遗传学信息,推断出能够解释 NPM1 突变 AML 生物学独立特征的潜在因素。该研究使用了CALGB/Alliance急性髓细胞性白血病队列(CALGB/Alliance AML cohort),其中包括581名NPM1突变患者。分析表明,RNA测序对解释患者之间的差异贡献最大,其次是RNA剪接,突出了转录组信息的深度。令人惊讶的是,基因突变只占总变异的约3%,而DNA甲基化则占总变异的15%以上。我们能够推断出 15 个潜在因素,其中 5 个与总生存率、无事件生存率和完全缓解率相关。在对 FLT3-ITD、性别和年龄等既定预后特征进行调整后,所有因素均具有显著性。因素2、7和11与不良预后相关,不同的因素包括HOX特征增加、TP53激活受抑制、干细胞样表型、表观遗传学SHS特征、三重NPM1/FLT3-ITD/DNMT3A突变以及包括NUP98在内的替代剪接。因子 4 和 13 具有良好的预后价值。因子 13 发现了一种独特的 X 连锁癌睾丸抗原基因家族成员表达模式,这种模式将 NPM1 患者区分开来,与性别、年龄或常见突变无关。该因子与CD34、MN1和BAALC--与干性有关的预后基因--的表达成反比。对免疫细胞亚群的分析表明,在13因子高与13因子低的患者中,CD8效应T细胞群的比例存在差异。这一特征模式和临床意义在一个独立队列(Beat-AML)中得到了验证。最后,我们对生成的 15 个因子进行了聚类,并确定了 NPM1 突变患者的 5 个亚群--2 个具有有利预后价值的群组、2 个具有不利预后价值的群组和 1 个中间群组。总之,我们在 NPM1 基因突变患者中发现了 15 个主要的变异来源,其中包括 5 个与独特生物特征相关的临床相关因素,这些因素共同产生了独特的独立生物标志物特征,而单独考虑不同的 omics 数据类型时则无法观察到这些特征。引用格式:Salma B. Abdelbaky, Kyoko Yamaguchi, Yue-Zhong Wu, Kevin R. Coombes, Lianbo Yu, Christopher C. Oakes.使用综合多组学方法鉴定 NPM1 基因突变急性髓细胞性白血病的生物成分和新型临床亚群 [摘要].In:血癌发现研讨会论文集;2024 年 3 月 4-6 日;马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P25.
{"title":"Abstract P25: Identification of biological components and novel clinical subsets of NPM1-mutated AML using an integrated, multi-omics approach","authors":"Salma Abdelbaky, Kyoko Yamaguchi, Yue-Zhong Wu, Kevin R. Coombes, Lianbo Yu, Christopher C Oakes","doi":"10.1158/2643-3249.bcdsymp24-p25","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p25","url":null,"abstract":"\u0000 Acute myeloid leukemia is the most common acute leukemia in adults with a 5-year overall survival rate of only 28%. Relapse occurs in almost half of adults within 3 years. Due to a high degree of heterogeneity in biological and genetic features in AML tumor cells, current prognostic markers (including genetic aberrations) do not fully explain the ranges of phenotype and outcomes observed in AML patients. Recently, we characterized 13 DNA methylation signatures (epitypes) and a STAT hypomethylation signature (SHS) to be predictive of outcome and stable at relapse. Here, we used the integrative Multi-omics Factor Analysis (MOFA) approach to combine genetic, cytogenetic, transcriptomic, and our novel epigenetic information in an extensive multi-omics analysis to infer latent factors capable of explaining independent signatures comprising NPM1-mutated AML biology. The study used the well-annotated CALGB/Alliance AML cohort encompassing 581 NPM1-mutated patients. The analysis showed that RNA-sequencing contributed the most to explaining the variance between patients followed by RNA splicing, highlighting the depth of information present in the transcriptome. Surprisingly, genetic mutations contributed to only approximately 3% of the overall variance, while DNA methylation captured more than 15% of the total variance. We were able to infer 15 latent factors, 5 of which were associated with overall survival, event-free survival, and attainment of complete remission. All factors were significant after adjusting for established prognostic features, such as FLT3-ITD, sex, and age. Factors 2, 7 and 11 were associated with unfavorable outcomes, and variably included increased HOX signatures, suppressed TP53 activation, a stem cell-like phenotype, the epigenetic SHS signature, triple NPM1/FLT3-ITD/DNMT3A mutations, and alternative splicing, including NUP98. Factors 4 and 13 had a favorable prognostic value. Factor 13 uncovered a unique expression pattern of X-linked cancer testis antigen gene family members dividing NPM1 patients independent of sex, age, or common mutations. The factor was inversely associated with the expression of CD34, MN1 and BAALC – prognostic genes related to stemness. Analysis of immune cell subsets indicated differences in proportions of CD8 effector T cell populations between patients with high vs. low factor 13. This feature pattern and clinical significance were validated in an independent cohort (Beat-AML). Lastly, we clustered the 15 generated factors and identified 5 subsets of NPM1-mutated patients – 2 clusters with a favorable prognostic value, 2 clusters with unfavorable prognostic value, and one intermediate cluster. Overall, we identify 15 main sources of variance within NPM1-mutated patients, including 5 clinically-relevant factors associated with unique biological features, which together generate unique independent biomarker signatures not observed when individually considering separate omics data types.\u0000 Citation Form","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140080491","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}
引用次数: 0
Abstract P17: Epigenomic fingerprinting of limited primary immune cells using automated CUT&RUN 摘要 P17:利用自动 CUT&RUN 对有限的原始免疫细胞进行表观基因组指纹分析
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p17
Ellen N Weinzapfel, Keith E. Maier, M. Marunde, V. U. Kumary, Carolina P. Lin Windham, Danielle N. Maryanski, Liz Albertorio-Saez, Dughan J. Ahimovic, Michael J Bale, Juliana J Lee, Bryan J. Venters, Michael-Christopher Keogh
Chromatin structure drives gene expression programs during immune cell development and in cancer, making it central to the advancement of precision medicine. Indeed, the study of chromatin regulation has provided valuable insight on effector cell differentiation and function, anti-tumor immune responses, and therapeutic resistance. Epigenomic features – such as histone post-translational modifications (PTMs) and chromatin-associated proteins – mark distinct genomic compartments (e.g., promoters, enhancers) and regulate chromatin function/gene expression. Mapping the genomic distribution of these may uncover new biomarkers or drug targets and provides a rich context for exploring immunology. Despite widespread interest, integration of epigenomics in large-scale immunotherapy and/or drug research has been hampered by the poor sensitivity, high background, and low throughput of traditional chromatin mapping technologies, most notably ChIP-seq. Instead, efforts to characterize chromatin dynamics have focused on DNA methylation (bisulfite-seq) or chromatin accessibility (ATAC-seq). However, these assays provide an incomplete view of the chromatin landscape, limiting their ability to define cell differentiation pathways or derive mechanistic insight into disease etiology. Here, we present autoCUT&RUN, a high-throughput assay for rapid, ultra-sensitive profiling of epigenomic features from FACS-isolated primary cells or tissues. This workflow generates reliable profiles from ~10,000 cells per reaction, and is supported by a rigorous optimization strategy, high-quality antibodies, and quantitative spike-in controls. As part of a multi-site collaboration with the Immunological Genome Consortium, we used our autoCUT&RUN platform to build a comprehensive epigenomic database of primary mouse immune cells - composed of >1,500 epigenomic profiles from >100 different FACS-isolated immune cell types. These studies set the stage to leverage high-throughput epigenomics in immunotherapy and precision medicine applications. Citation Format: Ellen N. Weinzapfel, Keith E. Maier, Matthew R. Marunde, Vishnu U. Sunitha Kumary, Carolina P. Lin Windham, Danielle N Maryanski, Liz Albertorio-Saez, Dughan J. Ahimovic, Michael J. Bale, Juliana J. Lee, Bryan J. Venters, Michael-Christopher Keogh, Immunological Genome Consortium. Epigenomic fingerprinting of limited primary immune cells using automated CUT&RUN [abstract]. In: Proceedings of the Blood Cancer Discovery Symposium; 2024 Mar 4-6; Boston, MA. Philadelphia (PA): AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P17.
染色质结构驱动着免疫细胞发育和癌症中的基因表达程序,使其成为精准医学发展的核心。事实上,染色质调控研究为效应细胞分化和功能、抗肿瘤免疫反应和治疗耐药性提供了宝贵的见解。表观基因组特征--如组蛋白翻译后修饰(PTM)和染色质相关蛋白--标志着不同的基因组区隔(如启动子、增强子),并调控染色质功能/基因表达。绘制这些物质的基因组分布图可能会发现新的生物标记物或药物靶点,并为探索免疫学提供丰富的背景。尽管表观基因组学受到广泛关注,但由于传统染色质图谱绘制技术(尤其是 ChIP-seq)的灵敏度低、背景高、通量低,表观基因组学在大规模免疫疗法和/或药物研究中的整合一直受到阻碍。相反,研究染色质动态特征的工作主要集中在 DNA 甲基化(亚硫酸氢盐-质谱)或染色质可及性(ATAC-质谱)上。然而,这些检测方法提供的染色质景观并不完整,限制了它们定义细胞分化途径或从机理上深入了解疾病病因学的能力。在这里,我们介绍了 autoCUT&RUN,这是一种从 FACS 分离的原代细胞或组织中快速、超灵敏地分析表观基因组特征的高通量检测方法。该工作流程每次反应可从约 10,000 个细胞中生成可靠的图谱,并有严格的优化策略、高质量抗体和定量尖峰对照支持。作为与免疫基因组联盟(Immunological Genome Consortium)多点合作的一部分,我们利用 autoCUT&RUN 平台建立了一个全面的小鼠原代免疫细胞表观基因组数据库,该数据库由来自 >100 种不同 FACS 分离免疫细胞类型的 >1,500 份表观基因组图谱组成。这些研究为在免疫疗法和精准医疗应用中利用高通量表观基因组学奠定了基础。引用格式:Ellen N. Weinzapfel、Keith E. Maier、Matthew R. Marunde、Vishnu U. Sunitha Kumary、Carolina P.Lin Windham、Danielle N Maryanski、Liz Albertorio-Saez、Dughan J. Ahimovic、Michael J. Bale、Juliana J. Lee、Bryan J. Venters、Michael-Christopher Keogh、免疫基因组联盟。利用自动 CUT&RUN 对有限的原代免疫细胞进行表观基因组指纹分析 [摘要]。In:血癌发现研讨会论文集;2024 年 3 月 4-6 日;马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P17.
{"title":"Abstract P17: Epigenomic fingerprinting of limited primary immune cells using automated CUT&RUN","authors":"Ellen N Weinzapfel, Keith E. Maier, M. Marunde, V. U. Kumary, Carolina P. Lin Windham, Danielle N. Maryanski, Liz Albertorio-Saez, Dughan J. Ahimovic, Michael J Bale, Juliana J Lee, Bryan J. Venters, Michael-Christopher Keogh","doi":"10.1158/2643-3249.bcdsymp24-p17","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p17","url":null,"abstract":"\u0000 Chromatin structure drives gene expression programs during immune cell development and in cancer, making it central to the advancement of precision medicine. Indeed, the study of chromatin regulation has provided valuable insight on effector cell differentiation and function, anti-tumor immune responses, and therapeutic resistance. Epigenomic features – such as histone post-translational modifications (PTMs) and chromatin-associated proteins – mark distinct genomic compartments (e.g., promoters, enhancers) and regulate chromatin function/gene expression. Mapping the genomic distribution of these may uncover new biomarkers or drug targets and provides a rich context for exploring immunology. Despite widespread interest, integration of epigenomics in large-scale immunotherapy and/or drug research has been hampered by the poor sensitivity, high background, and low throughput of traditional chromatin mapping technologies, most notably ChIP-seq. Instead, efforts to characterize chromatin dynamics have focused on DNA methylation (bisulfite-seq) or chromatin accessibility (ATAC-seq). However, these assays provide an incomplete view of the chromatin landscape, limiting their ability to define cell differentiation pathways or derive mechanistic insight into disease etiology. Here, we present autoCUT&RUN, a high-throughput assay for rapid, ultra-sensitive profiling of epigenomic features from FACS-isolated primary cells or tissues. This workflow generates reliable profiles from ~10,000 cells per reaction, and is supported by a rigorous optimization strategy, high-quality antibodies, and quantitative spike-in controls. As part of a multi-site collaboration with the Immunological Genome Consortium, we used our autoCUT&RUN platform to build a comprehensive epigenomic database of primary mouse immune cells - composed of >1,500 epigenomic profiles from >100 different FACS-isolated immune cell types. These studies set the stage to leverage high-throughput epigenomics in immunotherapy and precision medicine applications.\u0000 Citation Format: Ellen N. Weinzapfel, Keith E. Maier, Matthew R. Marunde, Vishnu U. Sunitha Kumary, Carolina P. Lin Windham, Danielle N Maryanski, Liz Albertorio-Saez, Dughan J. Ahimovic, Michael J. Bale, Juliana J. Lee, Bryan J. Venters, Michael-Christopher Keogh, Immunological Genome Consortium. Epigenomic fingerprinting of limited primary immune cells using automated CUT&RUN [abstract]. In: Proceedings of the Blood Cancer Discovery Symposium; 2024 Mar 4-6; Boston, MA. Philadelphia (PA): AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P17.","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140080629","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}
引用次数: 0
Abstract P22: PVR (CD155) epigenetic status mediates adoptive cell therapy and anti-TIGIT response in Multiple Myeloma 摘要 P22:PVR(CD155)的表观遗传学状态介导多发性骨髓瘤的采纳细胞疗法和抗 TIGIT 反应
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p22
L. Martinez-Verbo, L. Villanueva, P. Llinàs-Arias, C. García-Prieto, D. Piñeyro, A. Oliver-Caldés, A. García-Ortiz, A. Valeri, C. F. de Larrea, J. Martínez-López, Gerardo Ferrer, Manel Esteller
DNA promoter methylation acts as a key regulator for gene expression and is deeply altered in tumorigenesis. In the context of tumor clearance, cytotoxic cells (i.e. T and NK cells) play a critical role. Their function is mainly controlled by immune checkpoint (IC) synapse events, where known inhibitory markers are detrimental for cytotoxic activation. TIGIT, an inhibitory receptor on immune cells, interacts with PVR on malignant cells, precluding recognition and clearance. Multiple Myeloma (MM) remains and incurable disease and represents 10% of all hematological malignancies. In this project, we have proved PVR is regulated by promoter methylation and the importance of PVR/TIGIT axis inhibition on current immunotherapeutic options for MM. Studying newly diagnosed MM patients (n=793) we observed that higher expression of PVR is associated with shorter overall survival (median overall survival of PVR high: 56 months; PVR low: not reached; Log-rank P<0.001). In silico analysis of the promoter region of PVR, suggested its regulation by DNA methylation, so we characterized the epigenetic regulation of PVR at DNA, RNA and protein levels on a panel of MM cell lines (two methylated: AMO-1, KMS-12_BM and four unmethylated: EJM, JJN-3, MM.1S and RPMI-8226) and validated its epigenetic regulation. We cocultured cytotoxic cells with PVR unmethylated (expressing) models as well as PVR depleted ones. This depletion was obtained by shRNA interference and at least four different donors were evaluated in all the coculture experiments. Three different cell lines (EJM, JJN-3 and RPMI-8226) were tested and all expressing controls showed resistance to cytotoxicity (Mann-Whitney P<0.05). To validate the PVR/TIGIT role, we incorporated 10μg/ml of neutralizing anti-TIGIT into the system. We observed an increase in cytotoxicity in the expressing models as observed in the depleted models (Mann-Whitney P<0.05). We then tested PVR/TIGIT effect on anti-BCMA/CD3 BiTE, anti-BCMA (ARI0002h) CAR-T, and anti-BCMA (ARI0002h) CAR-NK cells. In the presence of BiTE, PVR depletion had a significant impact on cytotoxicity (Mann-Whitney P<0.05) when compared to controls. Coculturing CAR-T cells and MM models showed similar effects (Mann-Whitney P<0.05). As for CAR-NK coculture, only depleting PVR on RPMI-8226 showed a significant increase in cytotoxicity (Mann-Whitney P=0.01) when compared with the control, whereas JJN-3 remained unaffected. In summary, in the context of MM, PVR's expression is regulated by its promoter region's methylation status. Expression of PVR effectively reduces cytotoxic cell response, and the addition of anti-TIGIT validated that the PVR cytotoxic cell inhibition is mediated by TIGIT interaction. Furthermore, immunotherapies benefit from PVR depletion. Finally, PVR/TIGIT interaction seems to impact patients treated with current therapies since their outcome correlates with PVR expression. These results warrant further investigation on PVR as a biomarker and
DNA 启动子甲基化是基因表达的关键调节因子,在肿瘤发生过程中会发生深刻变化。在清除肿瘤的过程中,细胞毒性细胞(即 T 细胞和 NK 细胞)发挥着至关重要的作用。它们的功能主要受免疫检查点(IC)突触事件的控制,其中已知的抑制性标记物不利于细胞毒性的激活。TIGIT是免疫细胞上的一种抑制性受体,它与恶性细胞上的PVR相互作用,阻碍识别和清除。多发性骨髓瘤(MM)仍然是一种无法治愈的疾病,占所有血液恶性肿瘤的 10%。在该项目中,我们证明了 PVR 受启动子甲基化调控,以及抑制 PVR/TIGIT 轴对当前 MM 免疫治疗方案的重要性。通过研究新诊断的 MM 患者(793 人),我们发现 PVR 的高表达与较短的总生存期相关(PVR 高的中位总生存期:56 个月;PVR 低的:未达到;Log-rank P<0.001)。对 PVR 启动子区域的硅学分析表明,PVR 受 DNA 甲基化的调控,因此我们在一组 MM 细胞系(两种甲基化细胞系:AMO-1、KMS-12_MM-1 和 KMS-12_MM-1)中对 PVR 在 DNA、RNA 和蛋白质水平的表观遗传调控进行了表征:AMO-1、KMS-12_BM 和四个未甲基化细胞系:EJM、JJN-3、MM.1S 和 RPMI-8226),并验证了其表观遗传调控。我们将细胞毒性细胞与 PVR 未甲基化(表达)模型和 PVR 缺失模型进行了共培养。这种缺失是通过 shRNA 干扰获得的,在所有共培养实验中至少评估了四种不同的供体。测试了三种不同的细胞系(EJM、JJN-3 和 RPMI-8226),所有表达对照都显示出细胞毒性抵抗力(Mann-Whitney P<0.05)。为了验证 PVR/TIGIT 的作用,我们在系统中加入了 10μg/ml 的中和性抗 TIGIT。我们在表达模型中观察到细胞毒性的增加,与在耗竭模型中观察到的相同(Mann-Whitney P<0.05)。然后,我们测试了 PVR/TIGIT 对抗 BCMA/CD3 BiTE、抗 BCMA (ARI0002h) CAR-T 和抗 BCMA (ARI0002h) CAR-NK 细胞的影响。与对照组相比,在存在 BiTE 的情况下,PVR 的耗竭对细胞毒性有显著影响(Mann-Whitney P<0.05)。CAR-T 细胞与 MM 模型的共培养显示出相似的效果(Mann-Whitney P<0.05)。至于 CAR-NK 共培养,与对照组相比,只有在 RPMI-8226 上消耗 PVR 才会显示细胞毒性显著增加(Mann-Whitney P=0.01),而 JJN-3 则不受影响。总之,在 MM 中,PVR 的表达受其启动子区域甲基化状态的调控。PVR 的表达能有效降低细胞毒性反应,而抗 TIGIT 的加入则验证了 PVR 的细胞毒性细胞抑制作用是由 TIGIT 相互作用介导的。此外,免疫疗法也能从 PVR 的消耗中获益。最后,PVR/TIGIT 相互作用似乎会影响接受现有疗法的患者,因为他们的治疗结果与 PVR 表达相关。这些结果值得进一步研究作为生物标记物和新型免疫疗法靶点的 PVR。引用格式:Laura Martinez-Verbo、Lorea Villanueva、Pere Llinàs-Arias、Carlos A. García-Prieto、David Piñeyro、Aina Oliver-Caldes、Almudena García-Ortiz、Antonio Valeri、Carlos Fernández de Larrea、Joaquín Martínez-López、Gerardo Ferrer、Manel Esteller。PVR(CD155)的表观遗传学状态介导多发性骨髓瘤的采纳细胞疗法和抗TIGIT反应[摘要]。见:血癌发现研讨会论文集》,2024 年 3 月 4-6 日,马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P22.
{"title":"Abstract P22: PVR (CD155) epigenetic status mediates adoptive cell therapy and anti-TIGIT response in Multiple Myeloma","authors":"L. Martinez-Verbo, L. Villanueva, P. Llinàs-Arias, C. García-Prieto, D. Piñeyro, A. Oliver-Caldés, A. García-Ortiz, A. Valeri, C. F. de Larrea, J. Martínez-López, Gerardo Ferrer, Manel Esteller","doi":"10.1158/2643-3249.bcdsymp24-p22","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p22","url":null,"abstract":"\u0000 DNA promoter methylation acts as a key regulator for gene expression and is deeply altered in tumorigenesis. In the context of tumor clearance, cytotoxic cells (i.e. T and NK cells) play a critical role. Their function is mainly controlled by immune checkpoint (IC) synapse events, where known inhibitory markers are detrimental for cytotoxic activation. TIGIT, an inhibitory receptor on immune cells, interacts with PVR on malignant cells, precluding recognition and clearance. Multiple Myeloma (MM) remains and incurable disease and represents 10% of all hematological malignancies. In this project, we have proved PVR is regulated by promoter methylation and the importance of PVR/TIGIT axis inhibition on current immunotherapeutic options for MM. Studying newly diagnosed MM patients (n=793) we observed that higher expression of PVR is associated with shorter overall survival (median overall survival of PVR high: 56 months; PVR low: not reached; Log-rank P<0.001). In silico analysis of the promoter region of PVR, suggested its regulation by DNA methylation, so we characterized the epigenetic regulation of PVR at DNA, RNA and protein levels on a panel of MM cell lines (two methylated: AMO-1, KMS-12_BM and four unmethylated: EJM, JJN-3, MM.1S and RPMI-8226) and validated its epigenetic regulation. We cocultured cytotoxic cells with PVR unmethylated (expressing) models as well as PVR depleted ones. This depletion was obtained by shRNA interference and at least four different donors were evaluated in all the coculture experiments. Three different cell lines (EJM, JJN-3 and RPMI-8226) were tested and all expressing controls showed resistance to cytotoxicity (Mann-Whitney P<0.05). To validate the PVR/TIGIT role, we incorporated 10μg/ml of neutralizing anti-TIGIT into the system. We observed an increase in cytotoxicity in the expressing models as observed in the depleted models (Mann-Whitney P<0.05). We then tested PVR/TIGIT effect on anti-BCMA/CD3 BiTE, anti-BCMA (ARI0002h) CAR-T, and anti-BCMA (ARI0002h) CAR-NK cells. In the presence of BiTE, PVR depletion had a significant impact on cytotoxicity (Mann-Whitney P<0.05) when compared to controls. Coculturing CAR-T cells and MM models showed similar effects (Mann-Whitney P<0.05). As for CAR-NK coculture, only depleting PVR on RPMI-8226 showed a significant increase in cytotoxicity (Mann-Whitney P=0.01) when compared with the control, whereas JJN-3 remained unaffected. In summary, in the context of MM, PVR's expression is regulated by its promoter region's methylation status. Expression of PVR effectively reduces cytotoxic cell response, and the addition of anti-TIGIT validated that the PVR cytotoxic cell inhibition is mediated by TIGIT interaction. Furthermore, immunotherapies benefit from PVR depletion. Finally, PVR/TIGIT interaction seems to impact patients treated with current therapies since their outcome correlates with PVR expression. These results warrant further investigation on PVR as a biomarker and","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140265743","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}
引用次数: 0
Abstract P29: Zotatifin, the eukaryotic translation initiation factor 4A (eIF4A) inhibitor, synergizes with Venetoclax to inhibit acute myeloid leukemia cell growth 摘要P29:真核翻译起始因子4A(eIF4A)抑制剂佐他替芬与Venetoclax协同抑制急性髓性白血病细胞的生长
IF 11.2 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2643-3249.bcdsymp24-p29
Y. Lee, Jonathan Good, Noha Awais, Benjamin Eschle, Jenny Noel, Jean Acosta, Erica DePasquale, David Cucchi, Prafulla Gokhale, Fadi Najm, P. van Galen
New targeted therapies such as the selective and potent BCL-2 inhibitor Venetoclax have revolutionized the treatment of acute myeloid leukemia (AML). However, due to the cellular heterogeneity of the disease, drug-resistant cell types often emerge and drive relapse. We found that the eukaryotic translation initiation factor 4A (EIF4A) is highly expressed in multiple primitive AML cell types compared to their healthy counterparts, suggesting its potential as a therapeutic target. By unwinding complex structures in the 5’ UTR of transcripts, eIF4A promotes the translation of oncogene transcripts. A first-in-class drug, Zotatifin, inhibits the helicase activity of eIF4A and curtails tumor growth in receptor tyrosine kinase-driven tumors. Zotatifin is currently being evaluated in phase 1/2 trials across multiple solid tumors (NCT04092673). However, its effectiveness in blood cancers such as AML remains unexplored, and it is unknown whether Zotatifin may work synergistically with Venetoclax. Here, we treated AML cell lines in vitro and primary healthy or AML bone marrow ex vivo with Zotatifin or Venetoclax alone and in combination, and demonstrate synergistic killing of AML cells by the combination therapy (ZIP synergy score = 57.0 in MOLM-13, 19.3 in MV4-11, P < 0.0001). Mechanistically, we show that Zotatifin decreases eIF4A and AKT expression, and its combination with Venetoclax drives increased BAK and cleaved BAK expression, as well as increased cleavage of MCL-1 and BCL-2, consistent with drug synergy by convergence on the apoptotic pathway. In pilot colony-forming unit (CFU) assays, we found the combination to be tolerable in healthy CD34+ human hematopoietic stem and progenitor cells (HSPCs). To test drug efficacy in vivo, we generated a patient-derived xenograft (PDX) AML model from a patient who experienced relapse with complex cytogenetics, FLT3 and NPM1 mutations. Only the combination of Zotatifin and Venetoclax but not monotherapies effectively suppressed tumor burden as assessed by human CD45 engraftment in the peripheral blood. Further, the combination of Zotatifin and Venetoclax significantly prolonged survival of xenografted mice (median survival after treatment: vehicle 11 days, Venetoclax 13 days, Zotatifin 15 days, combination 42 days, P = 0.0002). Together, our data indicates that Zotatifin is synergistic with Venetoclax and may represent an effective treatment option for patients who are either primary refractory or have developed resistance to existing Venetoclax-containing regimens. Citation Format: Yoke Seng Lee, Jonathan Good, Noha Awais, Benjamin Eschle, Jenny Noel, Jean Acosta, Erica DePasquale, David Cucchi, Prafulla Gokhale, Fadi Najm, Peter van Galen. Zotatifin, the eukaryotic translation initiation factor 4A (eIF4A) inhibitor, synergizes with Venetoclax to inhibit acute myeloid leukemia cell growth [abstract]. In: Proceedings of the Blood Cancer Discovery Symposium; 2024 Mar 4-6; Boston, MA. Philadelphia (PA): AACR;
新型靶向疗法,如选择性强效BCL-2抑制剂Venetoclax,彻底改变了急性髓性白血病(AML)的治疗方法。然而,由于这种疾病的细胞异质性,耐药细胞类型经常出现并导致复发。我们发现,与健康细胞相比,真核翻译起始因子 4A(EIF4A)在多种原始急性髓性白血病细胞类型中高度表达,这表明它有可能成为治疗靶点。通过解开转录本 5' UTR 中的复杂结构,EIF4A 促进了癌基因转录本的翻译。首创药物佐他替芬(Zotatifin)可抑制 eIF4A 的螺旋酶活性,抑制受体酪氨酸激酶驱动的肿瘤的生长。佐他替芬目前正在多种实体瘤的1/2期试验中进行评估(NCT04092673)。然而,它对血癌(如急性髓细胞白血病)的疗效仍有待探索,而且尚不清楚佐他替芬是否能与 Venetoclax 发挥协同作用。在此,我们用Zotatifin或Venetoclax单独或联合治疗体外AML细胞系和体外原发性健康或AML骨髓,并证明了联合疗法对AML细胞的协同杀伤作用(在MOLM-13中,ZIP协同作用得分=57.0;在MV4-11中,ZIP协同作用得分=19.3,P<0.0001)。从机理上讲,我们发现Zotatifin会降低eIF4A和AKT的表达,而它与Venetoclax的联用会增加BAK和裂解BAK的表达,以及增加MCL-1和BCL-2的裂解,这与通过汇聚凋亡途径产生药物协同作用是一致的。在试验性集落形成单位(CFU)测定中,我们发现这种组合在健康的 CD34+ 人类造血干细胞和祖细胞(HSPCs)中是可耐受的。为了测试药物在体内的疗效,我们从一名复发并伴有复杂细胞遗传学、FLT3 和 NPM1 基因突变的患者身上建立了患者衍生异种移植(PDX)急性髓细胞性白血病模型。根据外周血中人类 CD45 细胞的吞噬情况评估,只有佐他替芬和 Venetoclax 的联合疗法能有效抑制肿瘤负荷,而单一疗法则不能。此外,Zotatifin 和 Venetoclax 的联合用药显著延长了异种移植小鼠的生存期(治疗后中位生存期:载体 11 天,Venetoclax 13 天,Zotatifin 15 天,联合用药 42 天,P = 0.0002)。总之,我们的数据表明,Zotatifin与Venetoclax具有协同作用,对于初治难治或对现有含Venetoclax方案产生耐药性的患者来说,Zotatifin可能是一种有效的治疗选择。引用格式:Yoke Seng Lee, Jonathan Good, Noha Awais, Benjamin Eschle, Jenny Noel, Jean Acosta, Erica DePasquale, David Cucchi, Prafulla Gokhale, Fadi Najm, Peter van Galen.真核翻译起始因子 4A(eIF4A)抑制剂佐他替芬与 Venetoclax 协同抑制急性髓性白血病细胞生长 [摘要].见:血癌发现研讨会论文集;2024 年 3 月 4-6 日;马萨诸塞州波士顿。费城(宾夕法尼亚州):AACR; Blood Cancer Discov 2024;5(2_Suppl):Abstract nr P29.
{"title":"Abstract P29: Zotatifin, the eukaryotic translation initiation factor 4A (eIF4A) inhibitor, synergizes with Venetoclax to inhibit acute myeloid leukemia cell growth","authors":"Y. Lee, Jonathan Good, Noha Awais, Benjamin Eschle, Jenny Noel, Jean Acosta, Erica DePasquale, David Cucchi, Prafulla Gokhale, Fadi Najm, P. van Galen","doi":"10.1158/2643-3249.bcdsymp24-p29","DOIUrl":"https://doi.org/10.1158/2643-3249.bcdsymp24-p29","url":null,"abstract":"\u0000 New targeted therapies such as the selective and potent BCL-2 inhibitor Venetoclax have revolutionized the treatment of acute myeloid leukemia (AML). However, due to the cellular heterogeneity of the disease, drug-resistant cell types often emerge and drive relapse. We found that the eukaryotic translation initiation factor 4A (EIF4A) is highly expressed in multiple primitive AML cell types compared to their healthy counterparts, suggesting its potential as a therapeutic target. By unwinding complex structures in the 5’ UTR of transcripts, eIF4A promotes the translation of oncogene transcripts. A first-in-class drug, Zotatifin, inhibits the helicase activity of eIF4A and curtails tumor growth in receptor tyrosine kinase-driven tumors. Zotatifin is currently being evaluated in phase 1/2 trials across multiple solid tumors (NCT04092673). However, its effectiveness in blood cancers such as AML remains unexplored, and it is unknown whether Zotatifin may work synergistically with Venetoclax. Here, we treated AML cell lines in vitro and primary healthy or AML bone marrow ex vivo with Zotatifin or Venetoclax alone and in combination, and demonstrate synergistic killing of AML cells by the combination therapy (ZIP synergy score = 57.0 in MOLM-13, 19.3 in MV4-11, P < 0.0001). Mechanistically, we show that Zotatifin decreases eIF4A and AKT expression, and its combination with Venetoclax drives increased BAK and cleaved BAK expression, as well as increased cleavage of MCL-1 and BCL-2, consistent with drug synergy by convergence on the apoptotic pathway. In pilot colony-forming unit (CFU) assays, we found the combination to be tolerable in healthy CD34+ human hematopoietic stem and progenitor cells (HSPCs). To test drug efficacy in vivo, we generated a patient-derived xenograft (PDX) AML model from a patient who experienced relapse with complex cytogenetics, FLT3 and NPM1 mutations. Only the combination of Zotatifin and Venetoclax but not monotherapies effectively suppressed tumor burden as assessed by human CD45 engraftment in the peripheral blood. Further, the combination of Zotatifin and Venetoclax significantly prolonged survival of xenografted mice (median survival after treatment: vehicle 11 days, Venetoclax 13 days, Zotatifin 15 days, combination 42 days, P = 0.0002). Together, our data indicates that Zotatifin is synergistic with Venetoclax and may represent an effective treatment option for patients who are either primary refractory or have developed resistance to existing Venetoclax-containing regimens.\u0000 Citation Format: Yoke Seng Lee, Jonathan Good, Noha Awais, Benjamin Eschle, Jenny Noel, Jean Acosta, Erica DePasquale, David Cucchi, Prafulla Gokhale, Fadi Najm, Peter van Galen. Zotatifin, the eukaryotic translation initiation factor 4A (eIF4A) inhibitor, synergizes with Venetoclax to inhibit acute myeloid leukemia cell growth [abstract]. In: Proceedings of the Blood Cancer Discovery Symposium; 2024 Mar 4-6; Boston, MA. Philadelphia (PA): AACR; ","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":null,"pages":null},"PeriodicalIF":11.2,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140266282","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}
引用次数: 0
期刊
Blood Cancer Discovery
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