Pub Date : 2025-10-27DOI: 10.1158/2643-3230.BCD-25-0130
Paul Lin, Sunil Acharya, Francia Reyes-Silva, Rafet Basar, Nadima Uprety, Luz Yurany Moreno Rueda, Pei Lin, April L Gilbert, Pinaki P Banerjee, Dexing Fang, Chenyu Zhang, Ana Karen Nunez Cortes, Luciana Melo Garcia, May Daher, Luis Muniz-Feliciano, Gary M Deyter, Vernikka Woods, Seema Rawal, Ping Li, Corry M Jones, Rejeena Shrestha, Muzaffar H Qazilbash, Krina K Patel, Hans C Lee, Richard E Champlin, David Marin, Elizabeth J Shpall, Robert Z Orlowski, Katayoun Rezvani
CD70 is highly expressed in many cancers, including multiple myeloma (MM). We show in two cohorts of patients with MM that CD70 is elevated in several high-risk disease categories and correlates with poor survival. These findings were validated using single-cell RNA sequencing, flow cytometry and immunohistochemistry. Moreover, we demonstrate the feasibility of targeting CD70 in myeloma using natural killer (NK) cells engineered with a chimeric antigen receptor (CAR) incorporating the CD70 cognate receptor CD27 and IL-15. CAR27/IL-15 NK cells exerted potent in vitro and in vivo cytotoxicity against CD70+ MM cells, comparable to CAR27/IL-15 T cells, and remained effective in BCMA-knockout models. Collectively, these results establish CD70 as a promising therapeutic target for high-risk MM, particularly for patients who relapse after BCMA-directed therapy, providing preclinical support for the ongoing phase I/II clinical trial of CD70-targeting CAR-NK cells (NCT05092451).
{"title":"CD70-Targeting CAR-NK Cells Overcome BCMA Downregulation and Improve Survival in High-Risk Multiple Myeloma Models.","authors":"Paul Lin, Sunil Acharya, Francia Reyes-Silva, Rafet Basar, Nadima Uprety, Luz Yurany Moreno Rueda, Pei Lin, April L Gilbert, Pinaki P Banerjee, Dexing Fang, Chenyu Zhang, Ana Karen Nunez Cortes, Luciana Melo Garcia, May Daher, Luis Muniz-Feliciano, Gary M Deyter, Vernikka Woods, Seema Rawal, Ping Li, Corry M Jones, Rejeena Shrestha, Muzaffar H Qazilbash, Krina K Patel, Hans C Lee, Richard E Champlin, David Marin, Elizabeth J Shpall, Robert Z Orlowski, Katayoun Rezvani","doi":"10.1158/2643-3230.BCD-25-0130","DOIUrl":"https://doi.org/10.1158/2643-3230.BCD-25-0130","url":null,"abstract":"<p><p>CD70 is highly expressed in many cancers, including multiple myeloma (MM). We show in two cohorts of patients with MM that CD70 is elevated in several high-risk disease categories and correlates with poor survival. These findings were validated using single-cell RNA sequencing, flow cytometry and immunohistochemistry. Moreover, we demonstrate the feasibility of targeting CD70 in myeloma using natural killer (NK) cells engineered with a chimeric antigen receptor (CAR) incorporating the CD70 cognate receptor CD27 and IL-15. CAR27/IL-15 NK cells exerted potent in vitro and in vivo cytotoxicity against CD70+ MM cells, comparable to CAR27/IL-15 T cells, and remained effective in BCMA-knockout models. Collectively, these results establish CD70 as a promising therapeutic target for high-risk MM, particularly for patients who relapse after BCMA-directed therapy, providing preclinical support for the ongoing phase I/II clinical trial of CD70-targeting CAR-NK cells (NCT05092451).</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145379117","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 : 2025-09-03DOI: 10.1158/2643-3230.BCD-25-0221
Adam J de Smith
In this issue of Blood Cancer Discovery, Newman and colleagues investigated the impact of genetic ancestry on molecular subtypes, genomic alterations, and survival outcomes in a diverse cohort of patients with T-cell acute lymphoblastic leukemia. They demonstrated that the prognostic utility of genomic alterations varied by ancestry, in particular, with NOTCH1 mutations having no prognostic utility in patients of predominantly African ancestry, and their findings highlight the importance of considering genetic ancestry in the risk stratification of patients with T-ALL. See related article by Newman et al., p. 412.
{"title":"Toward Equitable Risk Classification for All Patients with T-ALL.","authors":"Adam J de Smith","doi":"10.1158/2643-3230.BCD-25-0221","DOIUrl":"10.1158/2643-3230.BCD-25-0221","url":null,"abstract":"<p><p>In this issue of Blood Cancer Discovery, Newman and colleagues investigated the impact of genetic ancestry on molecular subtypes, genomic alterations, and survival outcomes in a diverse cohort of patients with T-cell acute lymphoblastic leukemia. They demonstrated that the prognostic utility of genomic alterations varied by ancestry, in particular, with NOTCH1 mutations having no prognostic utility in patients of predominantly African ancestry, and their findings highlight the importance of considering genetic ancestry in the risk stratification of patients with T-ALL. See related article by Newman et al., p. 412.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"400-402"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405836/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144800431","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-25-0174
Simon Renders, Alexander Waclawiczek, Andreas Trumpp
In a large retrospective multicenter study of patients with acute myeloid leukemia treated with hypomethylating agents and venetoclax, Lachowiez and colleagues examine the interplay among genetic mutations, disease phenotype, and clinical outcomes. They show that within genetic subgroups-particularly NPM1 wild-type cases-monocytic differentiation is linked to poor treatment response and inferior long-term prognosis, adding a phenotypic layer to genetics-based response prediction. See related article by Lachowiez et al., p. 437.
{"title":"A FABulous Couple: Adding PHENotype to GENotype Improves HMA/VEN Response Prediction in AML.","authors":"Simon Renders, Alexander Waclawiczek, Andreas Trumpp","doi":"10.1158/2643-3230.BCD-25-0174","DOIUrl":"10.1158/2643-3230.BCD-25-0174","url":null,"abstract":"<p><p>In a large retrospective multicenter study of patients with acute myeloid leukemia treated with hypomethylating agents and venetoclax, Lachowiez and colleagues examine the interplay among genetic mutations, disease phenotype, and clinical outcomes. They show that within genetic subgroups-particularly NPM1 wild-type cases-monocytic differentiation is linked to poor treatment response and inferior long-term prognosis, adding a phenotypic layer to genetics-based response prediction. See related article by Lachowiez et al., p. 437.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"403-405"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405864/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144800477","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-25-0047
Curtis A Lachowiez, Georgios Asimomitis, Elsa Bernard, Sean M Devlin, Yanis Tazi, Maria Creignou, Ulrich Germing, Norbert Gattermann, Amanda Gilkes, Ian Thomas, Lars Bullinger, Konstanze Döhner, Luca Malcovati, Jad Othman, Richard Dillon, Ann-Kathrin Eisfeld, Deedra Nicolet, Ghayas C Issa, Naval Daver, Tapan M Kadia, Courtney D DiNardo, Farhad Ravandi, Guillermo Garcia-Manero, Guillermo Montalban-Bravo, Nigel Russell, Mario Cazzola, Hartmut Döhner, Brian J P Huntly, Robert P Hasserjian, Eva Hellström-Lindberg, Elli Papaemmanuil, Sanam Loghavi
The World Health Organization fifth edition and International Consensus Classification for myeloid neoplasms both incorporate empirical numerical thresholds to morphologic and molecular features defining certain disease entities. However, the clinical implications of these thresholds remain unclear. We analyzed a large cohort (N = 6,976) of patients with myeloid neoplasms to evaluate the impact of proposed yet different numerical thresholds for variant allele frequency of genetic mutations or hematologic parameters set forth by the World Health Organization fifth edition and International Consensus Classification for classification of SF3B1-mutated myelodysplastic neoplasms, NPM1-mutated acute myeloid leukemia (AML), and oligomonocytic chronic myelomonocytic leukemia. Our analysis demonstrated that the clonal burden of SF3B1 mutation in myelodysplastic neoplasms informs classification and prognosis. Our findings support the notion that NPM1 mutation should be AML-defining regardless of blast percentage and highlight the adverse prognostic impact of the cumulative number of myelodysplasia-related mutations in NPM1-mutated AML. Finally, we provide evidence that integrating specific molecular signatures could improve the accuracy of oligomonocytic chronic myelomonocytic leukemia classification.
Significance: Using comprehensive clinical and molecular profiling, this study provides a data-driven approach for evaluating numerical thresholds of variant allele frequency or hematologic parameters (i.e., blast percentage and absolute monocyte count) included in current classification schemas across a spectrum of myeloid malignancies, enabling refinement of disease classification and prognostication.
{"title":"Multimodal and Data-Driven Assessment of Myeloid Neoplasms Refines Classification across Disease States.","authors":"Curtis A Lachowiez, Georgios Asimomitis, Elsa Bernard, Sean M Devlin, Yanis Tazi, Maria Creignou, Ulrich Germing, Norbert Gattermann, Amanda Gilkes, Ian Thomas, Lars Bullinger, Konstanze Döhner, Luca Malcovati, Jad Othman, Richard Dillon, Ann-Kathrin Eisfeld, Deedra Nicolet, Ghayas C Issa, Naval Daver, Tapan M Kadia, Courtney D DiNardo, Farhad Ravandi, Guillermo Garcia-Manero, Guillermo Montalban-Bravo, Nigel Russell, Mario Cazzola, Hartmut Döhner, Brian J P Huntly, Robert P Hasserjian, Eva Hellström-Lindberg, Elli Papaemmanuil, Sanam Loghavi","doi":"10.1158/2643-3230.BCD-25-0047","DOIUrl":"10.1158/2643-3230.BCD-25-0047","url":null,"abstract":"<p><p>The World Health Organization fifth edition and International Consensus Classification for myeloid neoplasms both incorporate empirical numerical thresholds to morphologic and molecular features defining certain disease entities. However, the clinical implications of these thresholds remain unclear. We analyzed a large cohort (N = 6,976) of patients with myeloid neoplasms to evaluate the impact of proposed yet different numerical thresholds for variant allele frequency of genetic mutations or hematologic parameters set forth by the World Health Organization fifth edition and International Consensus Classification for classification of SF3B1-mutated myelodysplastic neoplasms, NPM1-mutated acute myeloid leukemia (AML), and oligomonocytic chronic myelomonocytic leukemia. Our analysis demonstrated that the clonal burden of SF3B1 mutation in myelodysplastic neoplasms informs classification and prognosis. Our findings support the notion that NPM1 mutation should be AML-defining regardless of blast percentage and highlight the adverse prognostic impact of the cumulative number of myelodysplasia-related mutations in NPM1-mutated AML. Finally, we provide evidence that integrating specific molecular signatures could improve the accuracy of oligomonocytic chronic myelomonocytic leukemia classification.</p><p><strong>Significance: </strong>Using comprehensive clinical and molecular profiling, this study provides a data-driven approach for evaluating numerical thresholds of variant allele frequency or hematologic parameters (i.e., blast percentage and absolute monocyte count) included in current classification schemas across a spectrum of myeloid malignancies, enabling refinement of disease classification and prognostication.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"425-436"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12294350/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144561337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-24-0256
Curtis A Lachowiez, Mael Heiblig, Gaspar Aspas Requena, Emmanuelle Tavernier-Tardy, Fangyan Dai, Amenech B Ashango, Daniel T Peters, Jacob Fang, Andy Kaempf, Nicola Long, Christopher A Eide, Stephen E Kurtz, Wei Xie, Anupriya Agarwal, Aishwarya Sahasrabudhe, Christine M McMahon, Maria L Amaya, Gabrielle Meyers, Arpita Gandhi, Jessica Leonard, Brandon Hayes-Lattin, Richard T Maziarz, Elie Traer, Rachel J Cook, Ronan Swords, Theodore P Braun, Jennifer N Saultz, Ashley M Eckel, Michael R Loken, Joshua F Zeidner, Jeffrey W Tyner, Daniel A Pollyea
Resistance to venetoclax (VEN)-based therapy in acute myeloid leukemia (AML) includes genetic (i.e., mutations in N/KRAS, FLT3-ITD, TP53) and phenotypic (i.e., monocytic differentiation) features. Whether monocytic differentiation contributes to clinical VEN resistance secondary to a genetic bias remains unknown. This multimodal, multicenter, international analysis, inclusive of 678 patients, comprehensively characterized the prognostic role of monocytic differentiation in patients with AML treated with hypomethylating agents combined with VEN. AML genetics and monocytic differentiation (HR = 1.89; 95% confidence interval, 1.35-2.66; P < 0.001) in NPM1 wild-type cases correlated with an increased risk of death, clustering of centralized quantitative multiparameter flow cytometry data, evaluation of RNA sequencing-derived AML maturation stage, and single-cell proteogenomics linked driver mutations with AML phenotype and antiapoptotic gene expression. This comprehensive analysis of AML genetics, phenotype, and antiapoptotic protein expression highlights the complementary role these factors impart following VEN-based therapy.
Significance: AML with monocytic differentiation often occurs in the context of co-occurring mutations within signaling pathways. In certain AML subgroups (such as NPM1 wild-type and signaling pathway gene-mutated), a monocytic phenotype is associated with decreased overall survival following VEN-based therapy. See related commentary by Renders, p. 403.
急性髓性白血病(AML)对venetoclax为基础的治疗的耐药性包括遗传(即N/KRAS, FLT3-ITD, TP53的突变)和表型(即单核细胞分化)特征。是否单核细胞分化有助于继发于遗传偏倚的临床venetoclax耐药仍然未知。这项多模式、多中心、包括678例患者的国际分析全面表征了单核细胞分化在接受低甲基化药物联合venetoclax治疗的AML患者中的预后作用。NPM1野生型病例的AML遗传和单核细胞分化(HR: 1.89, 95% CI: 1.35-2.66, p < 0.001)与死亡风险增加相关。集中定量多参数流式细胞术数据的聚类,RNA测序衍生的AML成熟阶段的评估,以及与AML表型和抗凋亡基因表达相关的驱动突变的单细胞蛋白质基因组学。这项对AML遗传学、表型和抗凋亡蛋白表达的综合分析强调了这些因素在venetoclax为基础的治疗后所具有的互补作用。
{"title":"Genetic and Phenotypic Correlates of Clinical Outcomes with Venetoclax in Acute Myeloid Leukemia: The GEN-PHEN-VEN Study.","authors":"Curtis A Lachowiez, Mael Heiblig, Gaspar Aspas Requena, Emmanuelle Tavernier-Tardy, Fangyan Dai, Amenech B Ashango, Daniel T Peters, Jacob Fang, Andy Kaempf, Nicola Long, Christopher A Eide, Stephen E Kurtz, Wei Xie, Anupriya Agarwal, Aishwarya Sahasrabudhe, Christine M McMahon, Maria L Amaya, Gabrielle Meyers, Arpita Gandhi, Jessica Leonard, Brandon Hayes-Lattin, Richard T Maziarz, Elie Traer, Rachel J Cook, Ronan Swords, Theodore P Braun, Jennifer N Saultz, Ashley M Eckel, Michael R Loken, Joshua F Zeidner, Jeffrey W Tyner, Daniel A Pollyea","doi":"10.1158/2643-3230.BCD-24-0256","DOIUrl":"10.1158/2643-3230.BCD-24-0256","url":null,"abstract":"<p><p>Resistance to venetoclax (VEN)-based therapy in acute myeloid leukemia (AML) includes genetic (i.e., mutations in N/KRAS, FLT3-ITD, TP53) and phenotypic (i.e., monocytic differentiation) features. Whether monocytic differentiation contributes to clinical VEN resistance secondary to a genetic bias remains unknown. This multimodal, multicenter, international analysis, inclusive of 678 patients, comprehensively characterized the prognostic role of monocytic differentiation in patients with AML treated with hypomethylating agents combined with VEN. AML genetics and monocytic differentiation (HR = 1.89; 95% confidence interval, 1.35-2.66; P < 0.001) in NPM1 wild-type cases correlated with an increased risk of death, clustering of centralized quantitative multiparameter flow cytometry data, evaluation of RNA sequencing-derived AML maturation stage, and single-cell proteogenomics linked driver mutations with AML phenotype and antiapoptotic gene expression. This comprehensive analysis of AML genetics, phenotype, and antiapoptotic protein expression highlights the complementary role these factors impart following VEN-based therapy.</p><p><strong>Significance: </strong>AML with monocytic differentiation often occurs in the context of co-occurring mutations within signaling pathways. In certain AML subgroups (such as NPM1 wild-type and signaling pathway gene-mutated), a monocytic phenotype is associated with decreased overall survival following VEN-based therapy. See related commentary by Renders, p. 403.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"437-449"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405866/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144112165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-24-0328
Benjamin Diamond, Dhanvantri Chahar, Michael D Jain, Alexandra M Poos, Michael A Durante, Bachisio Ziccheddu, Marcella Kaddoura, Marios Papadimitriou, Kylee H Maclachlan, Tomas Jelinek, Faith E Davies, Nicholas B Figura, Gareth J Morgan, Elias K Mai, Katja Weisel, Roland Fenk, Marc S Raab, Saad Usmani, Ola Landgren, Frederick L Locke, Hartmut Goldschmidt, Jonathan Harry Schatz, Niels Weinhold, Francesco Maura
Ionizing radiotherapy (RT) is a widely used treatment strategy for malignancies. In solid tumors, RT-induced double-strand breaks lead to the accumulation of insertion-deletions (indels; ID), and their repair by nonhomologous end joining has been linked to the ID8 mutational signature in surviving cells. However, the extent of RT-induced mutagenesis in hematologic malignancies and its impact on their mutational profiles and interplay with commonly used chemotherapies has not yet been explored. In this study, we interrogated 580 whole-genome sequence (WGS) samples from patients with large B-cell lymphoma, multiple myeloma, and myeloid neoplasms and identified ID8 only in relapsed disease. Yet ID8 was detected after exposure to both RT and mutagenic chemotherapy (i.e., platinum and melphalan). Using WGS of single-cell colonies derived from treated lymphoma cells, we revealed a dose-response relationship between RT and platinum and ID8. Finally, using ID8 as a genomic barcode, we demonstrate that a single RT-surviving cell may seed distant relapse.
Significance: RT and the ID8 indel signature are related, but their genomic impact on hematologic malignancies is unclear. Leveraging WGS, we linked ID8 to both RT and mutagenic chemotherapy and validated that platinum can induce ID8. We used ID8 as a genomic barcode to reveal that RT-resistant cells may seed systemic relapse.
{"title":"Mutagenic Impact and Evolutionary Influence of Chemoradiotherapy in Hematologic Malignancies.","authors":"Benjamin Diamond, Dhanvantri Chahar, Michael D Jain, Alexandra M Poos, Michael A Durante, Bachisio Ziccheddu, Marcella Kaddoura, Marios Papadimitriou, Kylee H Maclachlan, Tomas Jelinek, Faith E Davies, Nicholas B Figura, Gareth J Morgan, Elias K Mai, Katja Weisel, Roland Fenk, Marc S Raab, Saad Usmani, Ola Landgren, Frederick L Locke, Hartmut Goldschmidt, Jonathan Harry Schatz, Niels Weinhold, Francesco Maura","doi":"10.1158/2643-3230.BCD-24-0328","DOIUrl":"10.1158/2643-3230.BCD-24-0328","url":null,"abstract":"<p><p>Ionizing radiotherapy (RT) is a widely used treatment strategy for malignancies. In solid tumors, RT-induced double-strand breaks lead to the accumulation of insertion-deletions (indels; ID), and their repair by nonhomologous end joining has been linked to the ID8 mutational signature in surviving cells. However, the extent of RT-induced mutagenesis in hematologic malignancies and its impact on their mutational profiles and interplay with commonly used chemotherapies has not yet been explored. In this study, we interrogated 580 whole-genome sequence (WGS) samples from patients with large B-cell lymphoma, multiple myeloma, and myeloid neoplasms and identified ID8 only in relapsed disease. Yet ID8 was detected after exposure to both RT and mutagenic chemotherapy (i.e., platinum and melphalan). Using WGS of single-cell colonies derived from treated lymphoma cells, we revealed a dose-response relationship between RT and platinum and ID8. Finally, using ID8 as a genomic barcode, we demonstrate that a single RT-surviving cell may seed distant relapse.</p><p><strong>Significance: </strong>RT and the ID8 indel signature are related, but their genomic impact on hematologic malignancies is unclear. Leveraging WGS, we linked ID8 to both RT and mutagenic chemotherapy and validated that platinum can induce ID8. We used ID8 as a genomic barcode to reveal that RT-resistant cells may seed systemic relapse.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"450-463"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405863/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144121083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-24-0327
Cuijuan Han, Zhiping Zhang, Edie I Crosse, Sogand Sajedi, Bin Lu, Xiyue Wang, Sadik Karma, Mitch Kostich, Sakthi Harini Rajendran, Dylan B Udy, Steven Chen, Alexander Arnuk, Abimbola Eunice Lawal, Kayla R Koenig, Meryl McKenna, Patrick K Reville, Hussein A Abbas, Omar Abdel-Wahab, Pedro Miura, Robert K Bradley, Eric Wang
Aberrant levels or structures of RNA isoforms are a hallmark of many cancers, including acute myeloid leukemia (AML), yet their role in AML chemoresistance remains unclear. We conducted a paired analysis of RNA isoform changes in patients with AML before therapy and at relapse after chemotherapy and identified intragenic DNA methylation at the proximal promoter of the transcription factor RUNX1, which resulted in elevated expression of the long-isoform RUNX1C through its alternative distal promoter. The unique N-terminal region of RUNX1C orchestrated an isoform-specific transcriptional program that promoted chemoresistance, with its direct target BTG2 playing a role in chemotherapy resistance. BTG2 promoted rRNA deadenylation, resulting in decreased mRNA expression and stability. Deletion of rRNAs increased cellular quiescence. Moreover, RNA-based targeting of RUNX1C reactivated quiescent leukemia cells and enhanced chemotherapy efficacy. These findings delineated an isoform-specific transcriptional circuit that governed chemotherapy response, providing a potential therapeutic strategy to mitigate AML recurrence.
Significance: This study identifies RUNX1C as a contributor to AML chemoresistance and an inducer of quiescence through BTG2. Targeting RUNX1C with RNA-based approaches disrupts this state and improves chemotherapy response, highlighting RUNX1C inhibition as a promising strategy to overcome resistance and enhance treatment efficacy in AML.
{"title":"An Isoform-Specific RUNX1C-BTG2 Axis Governs AML Quiescence and Chemoresistance.","authors":"Cuijuan Han, Zhiping Zhang, Edie I Crosse, Sogand Sajedi, Bin Lu, Xiyue Wang, Sadik Karma, Mitch Kostich, Sakthi Harini Rajendran, Dylan B Udy, Steven Chen, Alexander Arnuk, Abimbola Eunice Lawal, Kayla R Koenig, Meryl McKenna, Patrick K Reville, Hussein A Abbas, Omar Abdel-Wahab, Pedro Miura, Robert K Bradley, Eric Wang","doi":"10.1158/2643-3230.BCD-24-0327","DOIUrl":"10.1158/2643-3230.BCD-24-0327","url":null,"abstract":"<p><p>Aberrant levels or structures of RNA isoforms are a hallmark of many cancers, including acute myeloid leukemia (AML), yet their role in AML chemoresistance remains unclear. We conducted a paired analysis of RNA isoform changes in patients with AML before therapy and at relapse after chemotherapy and identified intragenic DNA methylation at the proximal promoter of the transcription factor RUNX1, which resulted in elevated expression of the long-isoform RUNX1C through its alternative distal promoter. The unique N-terminal region of RUNX1C orchestrated an isoform-specific transcriptional program that promoted chemoresistance, with its direct target BTG2 playing a role in chemotherapy resistance. BTG2 promoted rRNA deadenylation, resulting in decreased mRNA expression and stability. Deletion of rRNAs increased cellular quiescence. Moreover, RNA-based targeting of RUNX1C reactivated quiescent leukemia cells and enhanced chemotherapy efficacy. These findings delineated an isoform-specific transcriptional circuit that governed chemotherapy response, providing a potential therapeutic strategy to mitigate AML recurrence.</p><p><strong>Significance: </strong>This study identifies RUNX1C as a contributor to AML chemoresistance and an inducer of quiescence through BTG2. Targeting RUNX1C with RNA-based approaches disrupts this state and improves chemotherapy response, highlighting RUNX1C inhibition as a promising strategy to overcome resistance and enhance treatment efficacy in AML.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"464-483"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405853/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144592464","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-24-0279
Jaeyong Jung, Sining Zhu, Almin Lalani, Judith Shakarchi, Brygida Matracz, Guojun Gary Wu, Wei-Xing Zong, Liping Zhao, Ping Xie
Myeloid cells are central players in innate immunity and inflammation. Their function is regulated by the adapter protein TRAF3. We previously reported that aging myeloid cell-specific TRAF3-deficient (M-Traf3-/-) mice spontaneously develop chronic inflammation and B-cell lymphoma (BCL). In this study, we aimed to identify the internal trigger of this disease phenotype in these mice. We first detected gut microbiota dysbiosis and transmigration of commensal bacteria (CB) to the liver in aging M-Traf3-/- mice. Interestingly, depletion of CB using antibiotics effectively prevented BCL development in these mice. Systemic IgG responses against CB were induced and the IgH CDR3 sequences of malignant B-cell clones of M-Traf3-/- mice showed high homology to prevalent bacteria-reactive Ig clonotypes. Furthermore, M-Traf3-/- mice with BCL exhibited high serum titers of antibodies against CB. Together, our findings offer insights into the mechanisms underlying increased risks of B-cell lymphomagenesis observed in patients with compromised innate immunity.
Significance: We present evidence that microbiota dysbiosis in animals with compromised innate immunity increases risk of intestinal bacteria transmigration to internal organs, which subsequently induces malignant transformation of CB-reactive B-cell clones. Accordingly, antibiotic treatment or blocking CB transmigration may serve as a strategy for preventing BCL in patients with innate immunodeficiency.
{"title":"Commensal Bacteria Drive B-cell Lymphomagenesis in the Setting of Innate Immunodeficiency.","authors":"Jaeyong Jung, Sining Zhu, Almin Lalani, Judith Shakarchi, Brygida Matracz, Guojun Gary Wu, Wei-Xing Zong, Liping Zhao, Ping Xie","doi":"10.1158/2643-3230.BCD-24-0279","DOIUrl":"10.1158/2643-3230.BCD-24-0279","url":null,"abstract":"<p><p>Myeloid cells are central players in innate immunity and inflammation. Their function is regulated by the adapter protein TRAF3. We previously reported that aging myeloid cell-specific TRAF3-deficient (M-Traf3-/-) mice spontaneously develop chronic inflammation and B-cell lymphoma (BCL). In this study, we aimed to identify the internal trigger of this disease phenotype in these mice. We first detected gut microbiota dysbiosis and transmigration of commensal bacteria (CB) to the liver in aging M-Traf3-/- mice. Interestingly, depletion of CB using antibiotics effectively prevented BCL development in these mice. Systemic IgG responses against CB were induced and the IgH CDR3 sequences of malignant B-cell clones of M-Traf3-/- mice showed high homology to prevalent bacteria-reactive Ig clonotypes. Furthermore, M-Traf3-/- mice with BCL exhibited high serum titers of antibodies against CB. Together, our findings offer insights into the mechanisms underlying increased risks of B-cell lymphomagenesis observed in patients with compromised innate immunity.</p><p><strong>Significance: </strong>We present evidence that microbiota dysbiosis in animals with compromised innate immunity increases risk of intestinal bacteria transmigration to internal organs, which subsequently induces malignant transformation of CB-reactive B-cell clones. Accordingly, antibiotic treatment or blocking CB transmigration may serve as a strategy for preventing BCL in patients with innate immunodeficiency.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"505-525"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12279395/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144561336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-25-0141
Akash Patel, Shireen Kassam, Urvi A Shah
There is growing interest from both patients and clinicians in understanding the role nutrition plays across hematologic malignancies. In this study, we highlight key unanswered questions related to studying dietary interventions in hematologic malignancies, including questions about how to conduct research, trial design considerations, and integrating dietary interventions into hematologic cancer care and policy.
{"title":"Food for Thought: Addressing a Research Gap for Dietary Trials in Hematologic Malignancies.","authors":"Akash Patel, Shireen Kassam, Urvi A Shah","doi":"10.1158/2643-3230.BCD-25-0141","DOIUrl":"10.1158/2643-3230.BCD-25-0141","url":null,"abstract":"<p><p>There is growing interest from both patients and clinicians in understanding the role nutrition plays across hematologic malignancies. In this study, we highlight key unanswered questions related to studying dietary interventions in hematologic malignancies, including questions about how to conduct research, trial design considerations, and integrating dietary interventions into hematologic cancer care and policy.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"406-411"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405862/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144800478","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-03DOI: 10.1158/2643-3230.BCD-24-0203
Mireia Uribe-Herranz, Aina Oliver-Caldés, Neus Martínez-Micaelo, Marta Español-Rego, Maria Val-Casals, Roberto Martínez-Soler, Elisa Rubio-Garcia, Valeria Brunello, Erik Z Mihelic, Nela Klein-González, Daniel Benítez-Ribas, Núria Amigó, Andrea Vergara, Valentin Ortiz-Maldonado, Luis Gerardo Rodríguez-Lobato, Julio Delgado, Iñaki Ortiz de Landazuri, Verónica González-Calle, Valentín Cabañas, Beatriz Martin-Antonio, Lorena Pérez-Amill, Juan Luis Reguera-Ortega, Paula Rodríguez-Otero, Bruno Paiva, Joaquín Martínez-López, Maria-Victoria Mateos, Mariona Pascal, Álvaro Urbano-Ispizua, Europa Azucena González-Navarro, Carlos Fernández de Larrea, Manel Juan
Multiple myeloma remains incurable despite advances in immunotherapies like chimeric antigen receptor (CAR) T-cell therapy. This study investigates the role of metabolites and gut microbiota in clinical outcomes in patients treated with the humanized B-cell maturation antigen (BCMA)-directed CAR-T therapy ARI0002h. Stool metabolites, particularly succinate, were associated with CAR T-cell phenotypes and persistence in patients. In CAR T-cell culture, succinate supplementation enhanced CD4+ central memory phenotype and respiratory capacity. In a murine myeloma model, a succinate-enhancing diet significantly improved CAR T-cell persistence and showed a trend toward better tumor control. Furthermore, Acidaminococcaceae, Monoglobaceae, or Akkermansiaceae, along with specific metabolites, were associated with CAR T-cell clinical outcomes. These multimodal profiles were integrated into response models, including one that identified patients likely to achieve a complete response by days 100 and 180 after infusion. These findings suggest that metabolites and gut microbiota correlate with CAR T-cell therapy responses and can be a valuable tool for risk assessment.
Significance: This study integrates microbial profiles into response models, providing a tool to identify patients with multiple myeloma who may benefit from BCMA-directed CAR T-cell therapy optimization by identifying bacterial taxa and metabolites associated with CAR T-cell persistence and therapeutic outcomes.
{"title":"Microbiota Shape Metabolic and Immune Determinants of CAR-T Therapy and Correlate with Outcomes in Myeloma.","authors":"Mireia Uribe-Herranz, Aina Oliver-Caldés, Neus Martínez-Micaelo, Marta Español-Rego, Maria Val-Casals, Roberto Martínez-Soler, Elisa Rubio-Garcia, Valeria Brunello, Erik Z Mihelic, Nela Klein-González, Daniel Benítez-Ribas, Núria Amigó, Andrea Vergara, Valentin Ortiz-Maldonado, Luis Gerardo Rodríguez-Lobato, Julio Delgado, Iñaki Ortiz de Landazuri, Verónica González-Calle, Valentín Cabañas, Beatriz Martin-Antonio, Lorena Pérez-Amill, Juan Luis Reguera-Ortega, Paula Rodríguez-Otero, Bruno Paiva, Joaquín Martínez-López, Maria-Victoria Mateos, Mariona Pascal, Álvaro Urbano-Ispizua, Europa Azucena González-Navarro, Carlos Fernández de Larrea, Manel Juan","doi":"10.1158/2643-3230.BCD-24-0203","DOIUrl":"10.1158/2643-3230.BCD-24-0203","url":null,"abstract":"<p><p>Multiple myeloma remains incurable despite advances in immunotherapies like chimeric antigen receptor (CAR) T-cell therapy. This study investigates the role of metabolites and gut microbiota in clinical outcomes in patients treated with the humanized B-cell maturation antigen (BCMA)-directed CAR-T therapy ARI0002h. Stool metabolites, particularly succinate, were associated with CAR T-cell phenotypes and persistence in patients. In CAR T-cell culture, succinate supplementation enhanced CD4+ central memory phenotype and respiratory capacity. In a murine myeloma model, a succinate-enhancing diet significantly improved CAR T-cell persistence and showed a trend toward better tumor control. Furthermore, Acidaminococcaceae, Monoglobaceae, or Akkermansiaceae, along with specific metabolites, were associated with CAR T-cell clinical outcomes. These multimodal profiles were integrated into response models, including one that identified patients likely to achieve a complete response by days 100 and 180 after infusion. These findings suggest that metabolites and gut microbiota correlate with CAR T-cell therapy responses and can be a valuable tool for risk assessment.</p><p><strong>Significance: </strong>This study integrates microbial profiles into response models, providing a tool to identify patients with multiple myeloma who may benefit from BCMA-directed CAR T-cell therapy optimization by identifying bacterial taxa and metabolites associated with CAR T-cell persistence and therapeutic outcomes.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":"484-504"},"PeriodicalIF":11.5,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12405848/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144609689","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}