Pub Date : 2026-01-05DOI: 10.1182/blood.2025031455
Juan Pablo Alderuccio,Diva Baggio,Sunwoo Han,Paola Ghione,Imran A Nizamuddin,Jahanzaib Khwaja,Aditi Saha,Ning Dong,Yucai Wang,Hua-Jay J Cherng,Seda S Tolu,Nina D Wagner-Johnston,Thomas A Ollila,Natalie S Grover,Jean L Koff,Amrita Desai,Praveen Ramakrishnan Geethakumari,Tamara K Moyo,Jose Sandoval-Sus,Narendranath Epperla,Danielle S Wallace,Manali Kamdar,Rita Tavarozzi,Alexey V Danilov,Han W Tun,Javier Munoz,Mayur S Narkhede,Joanna M Rhodes,Anca Prica,Andrea Kuhnl,Adrian Matthew Maraj,Jessica Okosun,Jeffery Smith,Wendy Osborne,Victoria Calvert,Dima El-Sharkawi,Ammar Hilali,Graham P Collins,Kim Linton,Nagah Elmusharaf,Anna Santarsieri,Farheen Karim,Firas Baidoun Firas,Sarah Monick,Iris Margalit Trutzer,Jones Can,Amy A Ayers,Jacopo Calabrese De Feo,John Sharp,Nilanjan Ghosh,Rachel Treitman,Avyakta Kallam,Izel Okcu,Chathuri Abeyakoon,William Hann,Aisling Barrett,Vismay Deshani,Brad S Kahl,Julio C Chavez,Adam J Olszewski,Kate Cwynarski
Secondary central nervous system (CNS) large B-cell lymphoma (SCNSL) occurs in the de novo setting, as a CNS-isolated relapse, or synchronous (concomitant CNS and systemic) relapse. SCNSL is a devastating event without therapeutic consensus. Thus, we aimed to evaluate treatment outcomes in an international cohort. Progression-free survival (PFS), overall survival (OS) and cumulative incidence of relapse (CIR, estimated using competing-risk models) were reported. Prognostic factors were identified in a 6-month landmark multivariate analysis. Outcomes following thiotepa autologous stem cell transplant (ASCT) and chimeric antigen receptor T-cell therapy (CAR-T) delivered at relapse were compared following propensity score matching (PSM). A total of 1139 patients were included in the analysis (de novo: 537; relapsed SCNSL: 602). 2-year PFS estimates were 40.4%, 43.9% and 16.2% for de novo SCNSL, CNS-isolated relapse, and synchronous relapse respectively. Patients with CNS-isolated relapse demonstrated low rates of systemic recurrence (24-month CIR 6%). Thiotepa-ASCT correlated with longer survival in de novo SCNSL (PFS: HR=0.57; P=0.005; and OS: HR=0.62; P=0.023) and CNS-isolated relapses (PFS: HR=0.55; P=0.002; and OS: HR=0.39; P<.0001) in 6-month multivariable landmark analysis. ASCT (thiotepa or non-thiotepa) also associated with improved survival in synchronous relapses (PFS: HR=0.57; P=0.023; and OS: HR=0.48; P=0.019). Higher survival with thiotepa-ASCT compared to CAR-T was observed in survival analyses following PSM (PFS: HR=0.45; P=0.005 and OS: HR=0.41; P=0.014). These data support thiotepa-ASCT in eligible patients, particularly de novo disease and CNS-isolated relapses. CNS-isolated relapse was infrequently associated with systemic recurrence, supporting treatment regimens adopted from primary CNS lymphoma.
{"title":"Treatment-related Outcomes and Patterns of Relapse in Secondary CNS Involvement by Large B-cell Lymphoma.","authors":"Juan Pablo Alderuccio,Diva Baggio,Sunwoo Han,Paola Ghione,Imran A Nizamuddin,Jahanzaib Khwaja,Aditi Saha,Ning Dong,Yucai Wang,Hua-Jay J Cherng,Seda S Tolu,Nina D Wagner-Johnston,Thomas A Ollila,Natalie S Grover,Jean L Koff,Amrita Desai,Praveen Ramakrishnan Geethakumari,Tamara K Moyo,Jose Sandoval-Sus,Narendranath Epperla,Danielle S Wallace,Manali Kamdar,Rita Tavarozzi,Alexey V Danilov,Han W Tun,Javier Munoz,Mayur S Narkhede,Joanna M Rhodes,Anca Prica,Andrea Kuhnl,Adrian Matthew Maraj,Jessica Okosun,Jeffery Smith,Wendy Osborne,Victoria Calvert,Dima El-Sharkawi,Ammar Hilali,Graham P Collins,Kim Linton,Nagah Elmusharaf,Anna Santarsieri,Farheen Karim,Firas Baidoun Firas,Sarah Monick,Iris Margalit Trutzer,Jones Can,Amy A Ayers,Jacopo Calabrese De Feo,John Sharp,Nilanjan Ghosh,Rachel Treitman,Avyakta Kallam,Izel Okcu,Chathuri Abeyakoon,William Hann,Aisling Barrett,Vismay Deshani,Brad S Kahl,Julio C Chavez,Adam J Olszewski,Kate Cwynarski","doi":"10.1182/blood.2025031455","DOIUrl":"https://doi.org/10.1182/blood.2025031455","url":null,"abstract":"Secondary central nervous system (CNS) large B-cell lymphoma (SCNSL) occurs in the de novo setting, as a CNS-isolated relapse, or synchronous (concomitant CNS and systemic) relapse. SCNSL is a devastating event without therapeutic consensus. Thus, we aimed to evaluate treatment outcomes in an international cohort. Progression-free survival (PFS), overall survival (OS) and cumulative incidence of relapse (CIR, estimated using competing-risk models) were reported. Prognostic factors were identified in a 6-month landmark multivariate analysis. Outcomes following thiotepa autologous stem cell transplant (ASCT) and chimeric antigen receptor T-cell therapy (CAR-T) delivered at relapse were compared following propensity score matching (PSM). A total of 1139 patients were included in the analysis (de novo: 537; relapsed SCNSL: 602). 2-year PFS estimates were 40.4%, 43.9% and 16.2% for de novo SCNSL, CNS-isolated relapse, and synchronous relapse respectively. Patients with CNS-isolated relapse demonstrated low rates of systemic recurrence (24-month CIR 6%). Thiotepa-ASCT correlated with longer survival in de novo SCNSL (PFS: HR=0.57; P=0.005; and OS: HR=0.62; P=0.023) and CNS-isolated relapses (PFS: HR=0.55; P=0.002; and OS: HR=0.39; P<.0001) in 6-month multivariable landmark analysis. ASCT (thiotepa or non-thiotepa) also associated with improved survival in synchronous relapses (PFS: HR=0.57; P=0.023; and OS: HR=0.48; P=0.019). Higher survival with thiotepa-ASCT compared to CAR-T was observed in survival analyses following PSM (PFS: HR=0.45; P=0.005 and OS: HR=0.41; P=0.014). These data support thiotepa-ASCT in eligible patients, particularly de novo disease and CNS-isolated relapses. CNS-isolated relapse was infrequently associated with systemic recurrence, supporting treatment regimens adopted from primary CNS lymphoma.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"30 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903566","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-05DOI: 10.1182/blood.2025032277
Hiroshi Kimura,Jeffrey I Cohen
Chronic active Epstein-Barr virus (CAEBV) disease is an uncommon, often lethal T and/or NK (T/NK)-cell lymphoproliferative disorder that remains underrecognized outside Asia. Recent advances in molecular and immunopathologic studies-together with the 2022 International Consensus Classification and the 5th World Health Organization lymphoma classification-have consolidated the disease concept and diagnostic framework. Recent studies support a model wherein mutated EBV infects hematopoietic stem or lymphoid progenitor cells in the bone marrow, establishing latent infection that persists as these progenitors differentiate into T/NK cells. The infected lymphocytes subsequently undergo clonal expansion, immune evasion, and progressive accumulation of genetic and epigenetic alterations, giving rise to systemic CAEBV disease and, in some cases, transformation into EBV-positive lymphomas or leukemias. We review the clinical spectrum and differential diagnosis in relation to EBV-associated hemophagocytic lymphohistiocytosis and EBV-positive lymphomas or leukemias, and highlight geographic differences between Asian and non-Asian cohorts. Despite progress, diagnosis remains hampered by the lack of standardized and commercially available assays to identify infected cell subsets. Hematopoietic stem cell transplantation remains the only curative option, yet transplant-related mortality, relapse, and suboptimal outcomes in adult-onset disease underscore the need for optimized conditioning and pre-transplant disease control. We review emerging therapeutic strategies-including PD-1 blockade, JAK inhibition, and EBV-specific cytotoxic T-lymphocyte therapy-and outline priorities for prospective international trials. This review aims to raise global awareness among hematologists and foster collaborative studies to improve outcomes for patients with CAEBV disease.
{"title":"Chronic Active Epstein-Barr Virus Disease: Molecular Pathogenesis, Evolving Concepts, and Emerging Therapies.","authors":"Hiroshi Kimura,Jeffrey I Cohen","doi":"10.1182/blood.2025032277","DOIUrl":"https://doi.org/10.1182/blood.2025032277","url":null,"abstract":"Chronic active Epstein-Barr virus (CAEBV) disease is an uncommon, often lethal T and/or NK (T/NK)-cell lymphoproliferative disorder that remains underrecognized outside Asia. Recent advances in molecular and immunopathologic studies-together with the 2022 International Consensus Classification and the 5th World Health Organization lymphoma classification-have consolidated the disease concept and diagnostic framework. Recent studies support a model wherein mutated EBV infects hematopoietic stem or lymphoid progenitor cells in the bone marrow, establishing latent infection that persists as these progenitors differentiate into T/NK cells. The infected lymphocytes subsequently undergo clonal expansion, immune evasion, and progressive accumulation of genetic and epigenetic alterations, giving rise to systemic CAEBV disease and, in some cases, transformation into EBV-positive lymphomas or leukemias. We review the clinical spectrum and differential diagnosis in relation to EBV-associated hemophagocytic lymphohistiocytosis and EBV-positive lymphomas or leukemias, and highlight geographic differences between Asian and non-Asian cohorts. Despite progress, diagnosis remains hampered by the lack of standardized and commercially available assays to identify infected cell subsets. Hematopoietic stem cell transplantation remains the only curative option, yet transplant-related mortality, relapse, and suboptimal outcomes in adult-onset disease underscore the need for optimized conditioning and pre-transplant disease control. We review emerging therapeutic strategies-including PD-1 blockade, JAK inhibition, and EBV-specific cytotoxic T-lymphocyte therapy-and outline priorities for prospective international trials. This review aims to raise global awareness among hematologists and foster collaborative studies to improve outcomes for patients with CAEBV disease.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"391 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903571","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-05DOI: 10.1182/blood.2025030722
Hartmut Döhner,Daniela Späth,Maral Saadati,Walter Fiedler,Katharina S Götze,Elisabeth Koller,Jörg Westermann,Wichard Vogel,Michael Heuser,Michael Lübbert,Hans-Joachim Tischler,Ulrich Germing,Lino Lars Teichmann,Lars Fransecky,Albert Wölfler,David Nachbaur,Bernd Hertenstein,Roland Schroers,Uwe M Martens,Stephanie von Harsdorf,Markus P Radsak,Gregor Aschauer,Stefanie Weißhaar,Andrea Corbacioglu,Anika Schrade,Verena I Gaidzik,Felicitas R Thol,Peter Paschka,Lars Bullinger,Axel Benner,Konstanze Döhner,Arnold Ganser
BACKGROUNDCore-binding factor acute myeloid leukemia (CBF-AML) is associated with KIT mutations and deregulated expression of KIT.AIMSWe report results from the randomized, open-label, phase 3 trial of intensive chemotherapy with or without the multi-kinase inhibitor dasatinib in adult patients with CBF-AML.METHODSPatients received "3+7" induction therapy, followed by 4 cycles of high-dose cytarabine; in the investigational arm, patients received dasatinib 100 mg QD on days 8-21 in induction, and on days 6-28 in consolidation cycles, followed by 12-month single-agent dasatinib 100 mg QD. Primary endpoint was event-free survival (EFS). Secondary endpoints included overall survival, relapse-free survival, and cumulative incidence of relapse.RESULTS202 patients were randomly assigned to the standard arm (n=102) and to the dasatinib arm (n=100). Median age was 49 years (range, 18, 77); 94 patients had t(8;21), 108 had inv(16)/t(16;16); 58 (28.7%) patients had a KIT co-mutation. There was no statistically significant difference in EFS (HR 0.92, 95%-CI 0.63, 1.33; p=0.66) or secondary endpoints between treatment arms. There was also no significant difference in EFS in subgroup analyses according to age, CBF-AML type, and KIT mutation status. The incidence of serious adverse events was higher in the investigational arm (64%) than in the standard arm (36%).CONCLUSIONIn patients with CBF-AML, the addition of dasatinib to intensive chemotherapy failed to improve survival outcomes. The addition of dasatinib was associated with an increase in toxicity. This trial was registered at www.CLINICALTRIALSgov as NCT02013648.
{"title":"Phase 3 study of intensive chemotherapy with or without dasatinib in core-binding factor acute myeloid leukemia.","authors":"Hartmut Döhner,Daniela Späth,Maral Saadati,Walter Fiedler,Katharina S Götze,Elisabeth Koller,Jörg Westermann,Wichard Vogel,Michael Heuser,Michael Lübbert,Hans-Joachim Tischler,Ulrich Germing,Lino Lars Teichmann,Lars Fransecky,Albert Wölfler,David Nachbaur,Bernd Hertenstein,Roland Schroers,Uwe M Martens,Stephanie von Harsdorf,Markus P Radsak,Gregor Aschauer,Stefanie Weißhaar,Andrea Corbacioglu,Anika Schrade,Verena I Gaidzik,Felicitas R Thol,Peter Paschka,Lars Bullinger,Axel Benner,Konstanze Döhner,Arnold Ganser","doi":"10.1182/blood.2025030722","DOIUrl":"https://doi.org/10.1182/blood.2025030722","url":null,"abstract":"BACKGROUNDCore-binding factor acute myeloid leukemia (CBF-AML) is associated with KIT mutations and deregulated expression of KIT.AIMSWe report results from the randomized, open-label, phase 3 trial of intensive chemotherapy with or without the multi-kinase inhibitor dasatinib in adult patients with CBF-AML.METHODSPatients received \"3+7\" induction therapy, followed by 4 cycles of high-dose cytarabine; in the investigational arm, patients received dasatinib 100 mg QD on days 8-21 in induction, and on days 6-28 in consolidation cycles, followed by 12-month single-agent dasatinib 100 mg QD. Primary endpoint was event-free survival (EFS). Secondary endpoints included overall survival, relapse-free survival, and cumulative incidence of relapse.RESULTS202 patients were randomly assigned to the standard arm (n=102) and to the dasatinib arm (n=100). Median age was 49 years (range, 18, 77); 94 patients had t(8;21), 108 had inv(16)/t(16;16); 58 (28.7%) patients had a KIT co-mutation. There was no statistically significant difference in EFS (HR 0.92, 95%-CI 0.63, 1.33; p=0.66) or secondary endpoints between treatment arms. There was also no significant difference in EFS in subgroup analyses according to age, CBF-AML type, and KIT mutation status. The incidence of serious adverse events was higher in the investigational arm (64%) than in the standard arm (36%).CONCLUSIONIn patients with CBF-AML, the addition of dasatinib to intensive chemotherapy failed to improve survival outcomes. The addition of dasatinib was associated with an increase in toxicity. This trial was registered at www.CLINICALTRIALSgov as NCT02013648.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"18 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903568","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-05DOI: 10.1182/blood.2025031409
Alberto Carturan,Mathew G Angelos,Puneeth Guruprasad,Ruchi P Patel,Raymone Pajarillo,Andrew Evan Lee,David Espie,Yunlin Zhang,Yi-Hao Chiang,Wei Xie,Jesse L Rodriguez,Jaryse Harris,Pooja Devi,Olabisi I Afolayan-Oloye,Jason Xu,Jonathan H Sussman,Omar Elghawy,Austin G Yang,Adam Barsouk,Jong Hyun Cho,Carolyn E Shaw,Ekta Brijesh Singh,Ositadimma Ugwuanyi,Luca Paruzzo,Federico Stella,Shan Liu,Siena Nason,Antonio Imparato,Antonia Rotolo,Jean Lemoine,David M Barrett,Avery D Posey,Alain H Rook,Vinodh Pillai,Adam Bagg,Stefano Aldo Pileri,Dongfang Liu,Kai Tan,Stephen J Schuster,David Trent Teachey,Patrizia Porazzi,Marco Ruella
Patients with T-cell lymphomas and leukemias have overall poor outcomes due to the lack of targeted and effective treatments, particularly in the relapsed and refractory settings. Development of chimeric antigen receptor (CAR) T-cells against T-cell neoplasms is limited by a lack of discriminating T-cell antigens that allow for effective anti-tumor responses while preventing CAR T-cell fratricide. We hypothesized that targeting CD2, a pan-T-cell antigen, using anti-CD2 CAR T-cells engineered without CD2 expression (CART2), would support CAR T-cell manufacturability and preclinical efficacy. Optimized CD2-knockout CART2, generated using CRISPR-Cas9, eradicated primary patient-derived CD2+ hematological neoplasms in vitro and in vivo, secreted effector cytokines, and exhibited adequate proliferative capacity. Nevertheless, CD2 has a key costimulatory function, and its deletion could lead to CAR T-cell dysfunction. Therefore, we tested the role of the CD2:CD58 axis in CAR T-cells, using the anti-CD19 CART models. We demonstrate that CD2 loss attenuates CART19 efficacy by reducing avidity for tumor antigen, co-stimulation, and ultimately in vivo activity. Analogously, we show that tumor CD58 loss reduces CART19 efficacy. To overcome this issue, we developed a novel PD-1:CD2 switch receptor that rescues intracellular CD2 signaling, particularly when PD-L1 is engaged, resulting in improved in vivo outcomes. Collectively, we studied the role of CD2 both as a target for CAR T cell therapy and as a critical costimulatory protein, whose signaling can be rescued using the PD-1:CD2 switch receptor. This receptor can be incorporated into CAR T-cells and provides an effective strategy to overcome CD2-signaling deficiencies.
{"title":"Harnessing the CD2 axis to broaden and enhance the efficacy of CAR T-cell therapies.","authors":"Alberto Carturan,Mathew G Angelos,Puneeth Guruprasad,Ruchi P Patel,Raymone Pajarillo,Andrew Evan Lee,David Espie,Yunlin Zhang,Yi-Hao Chiang,Wei Xie,Jesse L Rodriguez,Jaryse Harris,Pooja Devi,Olabisi I Afolayan-Oloye,Jason Xu,Jonathan H Sussman,Omar Elghawy,Austin G Yang,Adam Barsouk,Jong Hyun Cho,Carolyn E Shaw,Ekta Brijesh Singh,Ositadimma Ugwuanyi,Luca Paruzzo,Federico Stella,Shan Liu,Siena Nason,Antonio Imparato,Antonia Rotolo,Jean Lemoine,David M Barrett,Avery D Posey,Alain H Rook,Vinodh Pillai,Adam Bagg,Stefano Aldo Pileri,Dongfang Liu,Kai Tan,Stephen J Schuster,David Trent Teachey,Patrizia Porazzi,Marco Ruella","doi":"10.1182/blood.2025031409","DOIUrl":"https://doi.org/10.1182/blood.2025031409","url":null,"abstract":"Patients with T-cell lymphomas and leukemias have overall poor outcomes due to the lack of targeted and effective treatments, particularly in the relapsed and refractory settings. Development of chimeric antigen receptor (CAR) T-cells against T-cell neoplasms is limited by a lack of discriminating T-cell antigens that allow for effective anti-tumor responses while preventing CAR T-cell fratricide. We hypothesized that targeting CD2, a pan-T-cell antigen, using anti-CD2 CAR T-cells engineered without CD2 expression (CART2), would support CAR T-cell manufacturability and preclinical efficacy. Optimized CD2-knockout CART2, generated using CRISPR-Cas9, eradicated primary patient-derived CD2+ hematological neoplasms in vitro and in vivo, secreted effector cytokines, and exhibited adequate proliferative capacity. Nevertheless, CD2 has a key costimulatory function, and its deletion could lead to CAR T-cell dysfunction. Therefore, we tested the role of the CD2:CD58 axis in CAR T-cells, using the anti-CD19 CART models. We demonstrate that CD2 loss attenuates CART19 efficacy by reducing avidity for tumor antigen, co-stimulation, and ultimately in vivo activity. Analogously, we show that tumor CD58 loss reduces CART19 efficacy. To overcome this issue, we developed a novel PD-1:CD2 switch receptor that rescues intracellular CD2 signaling, particularly when PD-L1 is engaged, resulting in improved in vivo outcomes. Collectively, we studied the role of CD2 both as a target for CAR T cell therapy and as a critical costimulatory protein, whose signaling can be rescued using the PD-1:CD2 switch receptor. This receptor can be incorporated into CAR T-cells and provides an effective strategy to overcome CD2-signaling deficiencies.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"41 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903573","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-05DOI: 10.1182/blood.2025030489
Laura Wiegand,Patricia Silva,Daniel Noerenberg,Friederike Christen,Klara Kopp,Benjamin Nick Locher,Pelle Löwe,Marlon Tilgner,Robert Altwasser,Vanessa Storzer,Catarina M Stein,Franziska Briest,Christopher Maximilian Arends,Mareike Frick,Jana Ihlow,Anna Dolnik,Naveed Ishaque,Ulrich Keller,Il-Kang Na,Livius Penter,Lars Bullinger,Raphael Hablesreiter,Frederik Damm
Gene-specific expansion patterns were evident among the most frequent CH lesions, with DNMT3A-mutant clones exhibiting impaired hematopoietic differentiation and TET2-mutant clones showing multi-lineage propagation. Notably, identical CH clones were detected in 41% of corresponding lymphomas, displaying distinct clonal dynamics: tumor-promoting CH (expansion in B-NHL; 10/16 clones; mainly TP53) and tumor-infiltrating CH (no expansion; mainly DNMT3A). Moreover, we identified lymphoma-associated mutations in flow-sorted hematopoietic progenitors from patients with indolent but not aggressive B-NHL and observed a stepwise accumulation of mutations along the lymphoid differentiation path. Single-cell genotyping confirmed the presence of mutated progenitors in 3 follicular, 2 mantle cell and 2 marginal zone lymphoma patients, providing direct evidence of a pre-neoplastic state in disease pathogenesis. Our findings offer novel insight into the cellular origin of nodal B-NHLs and highlight a previously underappreciated role for early clonal events involving the stem/progenitor cell compartment.
{"title":"Clonal Hematopoiesis and Lymphoma-Associated Mutations in Hematopoietic Progenitors in B-Cell Non-Hodgkin Lymphoma.","authors":"Laura Wiegand,Patricia Silva,Daniel Noerenberg,Friederike Christen,Klara Kopp,Benjamin Nick Locher,Pelle Löwe,Marlon Tilgner,Robert Altwasser,Vanessa Storzer,Catarina M Stein,Franziska Briest,Christopher Maximilian Arends,Mareike Frick,Jana Ihlow,Anna Dolnik,Naveed Ishaque,Ulrich Keller,Il-Kang Na,Livius Penter,Lars Bullinger,Raphael Hablesreiter,Frederik Damm","doi":"10.1182/blood.2025030489","DOIUrl":"https://doi.org/10.1182/blood.2025030489","url":null,"abstract":"Gene-specific expansion patterns were evident among the most frequent CH lesions, with DNMT3A-mutant clones exhibiting impaired hematopoietic differentiation and TET2-mutant clones showing multi-lineage propagation. Notably, identical CH clones were detected in 41% of corresponding lymphomas, displaying distinct clonal dynamics: tumor-promoting CH (expansion in B-NHL; 10/16 clones; mainly TP53) and tumor-infiltrating CH (no expansion; mainly DNMT3A). Moreover, we identified lymphoma-associated mutations in flow-sorted hematopoietic progenitors from patients with indolent but not aggressive B-NHL and observed a stepwise accumulation of mutations along the lymphoid differentiation path. Single-cell genotyping confirmed the presence of mutated progenitors in 3 follicular, 2 mantle cell and 2 marginal zone lymphoma patients, providing direct evidence of a pre-neoplastic state in disease pathogenesis. Our findings offer novel insight into the cellular origin of nodal B-NHLs and highlight a previously underappreciated role for early clonal events involving the stem/progenitor cell compartment.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"21 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1182/blood.2025028762
Bethany Verkamp, Michael B Jordan, Carl E Allen
Pediatric hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory syndrome, characterized by heightened T-cell activation and overwhelming IFN-g production. Rather than a binary "primary" versus "secondary" framework, it is best conceptualized as a threshold phenomenon that occurs when combined genetic and environmental factors exceed a critical tipping point, leading to a recognizable clinical pattern of abnormal immunopathology. In addition to classic genetic cytotoxic defects, an increasing number of genetic lesions continue to be recognized related to HLH. Environmental triggers frequently include malignancy, infection, and rheumatologic disorders and identifying underlying factors driving HLH is a crucial component of evaluation. The revised HLH-2004 criteria are an important tool in promptly recognizing HLH, but clinical criteria may be fulfilled by other inflammatory disorders. Genetic and functional immune testing are complementary tools for diagnosis and establishing recurrence risk. Dexamethasone and etoposide remain cornerstones of initial therapy but are best utilized in an individualized, response-based manner with close monitoring of inflammatory markers. Newer, more targeted agents continue to emerge and are often critical additions for achieving disease control. Most patients with inherited cytotoxic defects and other select causes require hematopoietic cell transplant (HCT) for cure which should occur as soon as feasible once adequate, not perfect, HLH control aims to minimize risk of reactivation, infection, and toxicity. This review aims to highlight key advancements in defining, diagnosing, and treating HLH based on a growing understanding of HLH pathophysiology. We also provide perspectives on practical clinical approaches that may be useful for diagnosing and treating pediatric HLH.
{"title":"Pediatric hemophagocytic lymphohistiocytosis: current conceptualization, diagnosis, and treatment.","authors":"Bethany Verkamp, Michael B Jordan, Carl E Allen","doi":"10.1182/blood.2025028762","DOIUrl":"https://doi.org/10.1182/blood.2025028762","url":null,"abstract":"<p><p>Pediatric hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory syndrome, characterized by heightened T-cell activation and overwhelming IFN-g production. Rather than a binary \"primary\" versus \"secondary\" framework, it is best conceptualized as a threshold phenomenon that occurs when combined genetic and environmental factors exceed a critical tipping point, leading to a recognizable clinical pattern of abnormal immunopathology. In addition to classic genetic cytotoxic defects, an increasing number of genetic lesions continue to be recognized related to HLH. Environmental triggers frequently include malignancy, infection, and rheumatologic disorders and identifying underlying factors driving HLH is a crucial component of evaluation. The revised HLH-2004 criteria are an important tool in promptly recognizing HLH, but clinical criteria may be fulfilled by other inflammatory disorders. Genetic and functional immune testing are complementary tools for diagnosis and establishing recurrence risk. Dexamethasone and etoposide remain cornerstones of initial therapy but are best utilized in an individualized, response-based manner with close monitoring of inflammatory markers. Newer, more targeted agents continue to emerge and are often critical additions for achieving disease control. Most patients with inherited cytotoxic defects and other select causes require hematopoietic cell transplant (HCT) for cure which should occur as soon as feasible once adequate, not perfect, HLH control aims to minimize risk of reactivation, infection, and toxicity. This review aims to highlight key advancements in defining, diagnosing, and treating HLH based on a growing understanding of HLH pathophysiology. We also provide perspectives on practical clinical approaches that may be useful for diagnosing and treating pediatric HLH.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":23.1,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1182/blood.2025031060
Yunfeng Liu, Sarah Shayo, Shan Su, Weili Bao, Hui Zhong, Irina Murakhovskaya, Cheryl A Lobo, Xiuli An, Deepa G Manwani, Patricia A Shi, Karina Yazdanbakhsh
Sickle cell nephropathy (SCN) is a major clinical complication in sickle cell disease (SCD), yet its underlying mechanisms remain incompletely defined. Hemolysis, a hallmark of SCD, has been implicated in SCN pathogenesis, but the downstream inflammatory pathways are not fully understood. We previously demonstrated that hemolysis triggers type I interferon (IFN-I) responses, leading to upregulation of the chemokine CCL2 and recruitment of classical monocytes that differentiate into monocyte-derived macrophages (MoMϕ) within livers in SCD. In this study, we show that IFN-I and CCL2 levels are elevated in the plasma of SCD patients with abnormal urine albumin-creatinine ratio (uACR) and in the kidneys of Townes SCD mouse model. Using IFN-I receptor (Ifnar1)-/- and CCL2 receptor (Ccr2)-/- mouse models of SCD, we demonstrate that loss of IFN-I or CCL2 signaling reduces MoMϕ accumulation, renal inflammation, and renal injury. Mechanistically, we identify that hemin-induced IFN-I production occurs via the TLR3/TRIF signaling axis, independent of MyD88, MAVS, or STING. These findings uncover a previously unrecognized heme-TLR3/TRIF-IFN-I-CCL2 pathway that contributes to renal pathology in SCD and suggest that targeting this axis may offer therapeutic benefit.
{"title":"Heme-Induced Activation of the TLR3/TRIF-IFN-I-CCL2 Pathway Contributes to Kidney Injury in Sickle Cell Disease.","authors":"Yunfeng Liu, Sarah Shayo, Shan Su, Weili Bao, Hui Zhong, Irina Murakhovskaya, Cheryl A Lobo, Xiuli An, Deepa G Manwani, Patricia A Shi, Karina Yazdanbakhsh","doi":"10.1182/blood.2025031060","DOIUrl":"https://doi.org/10.1182/blood.2025031060","url":null,"abstract":"<p><p>Sickle cell nephropathy (SCN) is a major clinical complication in sickle cell disease (SCD), yet its underlying mechanisms remain incompletely defined. Hemolysis, a hallmark of SCD, has been implicated in SCN pathogenesis, but the downstream inflammatory pathways are not fully understood. We previously demonstrated that hemolysis triggers type I interferon (IFN-I) responses, leading to upregulation of the chemokine CCL2 and recruitment of classical monocytes that differentiate into monocyte-derived macrophages (MoMϕ) within livers in SCD. In this study, we show that IFN-I and CCL2 levels are elevated in the plasma of SCD patients with abnormal urine albumin-creatinine ratio (uACR) and in the kidneys of Townes SCD mouse model. Using IFN-I receptor (Ifnar1)-/- and CCL2 receptor (Ccr2)-/- mouse models of SCD, we demonstrate that loss of IFN-I or CCL2 signaling reduces MoMϕ accumulation, renal inflammation, and renal injury. Mechanistically, we identify that hemin-induced IFN-I production occurs via the TLR3/TRIF signaling axis, independent of MyD88, MAVS, or STING. These findings uncover a previously unrecognized heme-TLR3/TRIF-IFN-I-CCL2 pathway that contributes to renal pathology in SCD and suggest that targeting this axis may offer therapeutic benefit.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":23.1,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1182/blood.2024027897
Chloe Al Thompson-Peach, Daniel Thomas, Mara Dottore, Kelly Lim, Frank Stomski, Romana Panagopoulos, Johannes Foßelteder, Andreas Reinisch, Vashe Chandrakanthan, Sandra Jenkner, Luke Bland, Sharon Paton, Stan Gronthos, Timothy Hercus, Hannah Wardill, Winnie L Kan, Maha Kamel, Paul A Moretti, Stuart M Pitson, Kate Burbury, David Morrall Ross, Pramod Nair, Angel F Lopez, Denis Tvorogov
Somatic frameshift mutations in the gene encoding calreticulin (CALR) give rise to myelofibrosis and are classified as Type 1 (del52) or Type 2 (ins5) according to the degree of wildtype sequence retained adjacent to the neopeptide, with each type conferring different clinical outcomes. Targeting strategies specific for Type 1 vs Type 2 mutations would have enormous clinical utility in the treatment and prevention of myelofibrosis as responses to tyrosine kinase inhibitors are not durable nor mutation-specific. Here we show that dual targeting of Type 1 (del52) mutant CALR with two monoclonal antibodies directed against distinct epitopes in CALR have significant advantages compared to single agent treatment in the eradication of primary megakaryocyte progenitors in vitro and in a humanized ossicle microenvironment leading to improved survival in xenograft models. Dual targeting was superior in blocking constitutive STAT5 and ERK phosphorylation induced by del52 and prevented accumulation of JAK2 phosphorylation, overcoming ruxolitinib resistance. In contrast, Type 2 mutations showed increased CALR dimerization and were partially resistant to antibody targeting but could be impacted by ruxolitinib triple combination. Together, our data demonstrate an ultra-precision medicine approach tailored to either Type 1 OR Type 2 mutation classes will be required for maximal efficacy and complete blockade of JAK/STAT signalling, with far-reaching implications for patient management.
{"title":"Ultraprecision Therapy for Type 1 vs Type 2 CALR+ MPN by Dual Epitope Targeting that Restores Ruxolitinib Sensitivity.","authors":"Chloe Al Thompson-Peach, Daniel Thomas, Mara Dottore, Kelly Lim, Frank Stomski, Romana Panagopoulos, Johannes Foßelteder, Andreas Reinisch, Vashe Chandrakanthan, Sandra Jenkner, Luke Bland, Sharon Paton, Stan Gronthos, Timothy Hercus, Hannah Wardill, Winnie L Kan, Maha Kamel, Paul A Moretti, Stuart M Pitson, Kate Burbury, David Morrall Ross, Pramod Nair, Angel F Lopez, Denis Tvorogov","doi":"10.1182/blood.2024027897","DOIUrl":"https://doi.org/10.1182/blood.2024027897","url":null,"abstract":"<p><p>Somatic frameshift mutations in the gene encoding calreticulin (CALR) give rise to myelofibrosis and are classified as Type 1 (del52) or Type 2 (ins5) according to the degree of wildtype sequence retained adjacent to the neopeptide, with each type conferring different clinical outcomes. Targeting strategies specific for Type 1 vs Type 2 mutations would have enormous clinical utility in the treatment and prevention of myelofibrosis as responses to tyrosine kinase inhibitors are not durable nor mutation-specific. Here we show that dual targeting of Type 1 (del52) mutant CALR with two monoclonal antibodies directed against distinct epitopes in CALR have significant advantages compared to single agent treatment in the eradication of primary megakaryocyte progenitors in vitro and in a humanized ossicle microenvironment leading to improved survival in xenograft models. Dual targeting was superior in blocking constitutive STAT5 and ERK phosphorylation induced by del52 and prevented accumulation of JAK2 phosphorylation, overcoming ruxolitinib resistance. In contrast, Type 2 mutations showed increased CALR dimerization and were partially resistant to antibody targeting but could be impacted by ruxolitinib triple combination. Together, our data demonstrate an ultra-precision medicine approach tailored to either Type 1 OR Type 2 mutation classes will be required for maximal efficacy and complete blockade of JAK/STAT signalling, with far-reaching implications for patient management.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":23.1,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892125","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Heatstroke, a severe hyperthermic condition, is characterized by a core body temperature exceeding 40℃ and multiple organ dysfunction syndrome (MODS) with an extremely high mortality rate. Despite advances in identifying heatstroke-induced cell death pathways, the molecular cascades that bridge heat-induced cell death to MODS and mortality are not yet fully characterized. Our findings demonstrate that Z-DNA binding protein 1 (ZBP1)-triggered disseminated intravascular coagulation critically drives MODS and fatal outcomes in heatstroke. Heat stress activates ZBP1-dependent necroptosis, promoting tissue factor (TF) release and phosphatidylserine (PS) externalization. Genetic knockout of ZBP1 or its downstream necroptotic effectors, reduction of global TF expression, suppression of PS exposure, or pharmacological inhibition of the coagulation cascade attenuates heat stress-induced coagulation activation, organ injury, and death. Comparable results are obtained in heat-stressed mice with conditional knockout of ZBP1 in hematopoietic or myeloid lineages. Overall, our study reveals the critical role of ZBP1-mediated necroptosis in bridging heat stress and coagulation dysfunction.
{"title":"Heat stress activates the coagulation cascade through Z-DNA-binding protein 1-dependent necroptosis.","authors":"Fanglin Li, Jinxiu Li, Jian Shi, Xinyu Yang, Ting Zhang, Hao Liu, Fengjie Wang, Xiao Yan, Qiuli He, Long Wu, Yong Zou, Yiting Tang, Erhua Wang, Yanjun Zhong","doi":"10.1182/blood.2025030764","DOIUrl":"https://doi.org/10.1182/blood.2025030764","url":null,"abstract":"<p><p>Heatstroke, a severe hyperthermic condition, is characterized by a core body temperature exceeding 40℃ and multiple organ dysfunction syndrome (MODS) with an extremely high mortality rate. Despite advances in identifying heatstroke-induced cell death pathways, the molecular cascades that bridge heat-induced cell death to MODS and mortality are not yet fully characterized. Our findings demonstrate that Z-DNA binding protein 1 (ZBP1)-triggered disseminated intravascular coagulation critically drives MODS and fatal outcomes in heatstroke. Heat stress activates ZBP1-dependent necroptosis, promoting tissue factor (TF) release and phosphatidylserine (PS) externalization. Genetic knockout of ZBP1 or its downstream necroptotic effectors, reduction of global TF expression, suppression of PS exposure, or pharmacological inhibition of the coagulation cascade attenuates heat stress-induced coagulation activation, organ injury, and death. Comparable results are obtained in heat-stressed mice with conditional knockout of ZBP1 in hematopoietic or myeloid lineages. Overall, our study reveals the critical role of ZBP1-mediated necroptosis in bridging heat stress and coagulation dysfunction.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":23.1,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1182/blood.2025029996
Marcella Kaddoura, Julia Erin Wiedmeier-Nutor, Vikas A Gupta, Tomas Jelinek, Bachisio Ziccheddu, Suganti Shivaram, Hongwei Tang, Rebecca W Owens, Tereza Ševčíková, Rodrigo Fonseca, Michael A Durante, Benjamin T Diamond, Logan Zhao, Yuan Xiao Zhu, Chang-Xin Shi, Shannon M Matulis, Constantine S Mitsiades, Carl Ola Landgren, Saad Z Usmani, Roman Hajek, Marta Chesi, P Leif Bergsagel, Esteban Braggio, Lawrence H Boise, Rafael Fonseca, Shaji K Kumar, Francesco Maura, Linda B Baughn
Multiple myeloma (MM) with t(11;14)(CCND1;IGH) remains the only subset sensitive to the BCL2 inhibitor venetoclax. Not all t(11;14)(CCND1;IGH) patients respond to treatment and some progress early after initial response. To investigate this, we interrogated 44 whole genome and exome sequencing data from 34 patients with t(11;14) MM treated with venetoclax. The presence of mutations in the RAS pathway was strongly associated with shortened progression-free survival (PFS) and was validated in an independent cohort of 21 MM patients. Presence of 1q gain was also associated with shorter PFS in patients without RAS mutations. In 10 patients with paired, pre- and post-venetoclax treatment samples, post-venetoclax progression was recurrently driven by the selection of genomic events in BCL2/MCL1 and RAS pathways and of high-risk features (e.g., loss of TP53 and CDKN2C). Overall, our study shows that comprehensive genomic profiling can identify most mechanisms underlying resistance to BCL2 inhibition in t(11;14)(CCND1;IGH) MM.
{"title":"Genomic mechanisms of resistance to venetoclax in multiple myeloma with t(11;14)(CCND1;IGH).","authors":"Marcella Kaddoura, Julia Erin Wiedmeier-Nutor, Vikas A Gupta, Tomas Jelinek, Bachisio Ziccheddu, Suganti Shivaram, Hongwei Tang, Rebecca W Owens, Tereza Ševčíková, Rodrigo Fonseca, Michael A Durante, Benjamin T Diamond, Logan Zhao, Yuan Xiao Zhu, Chang-Xin Shi, Shannon M Matulis, Constantine S Mitsiades, Carl Ola Landgren, Saad Z Usmani, Roman Hajek, Marta Chesi, P Leif Bergsagel, Esteban Braggio, Lawrence H Boise, Rafael Fonseca, Shaji K Kumar, Francesco Maura, Linda B Baughn","doi":"10.1182/blood.2025029996","DOIUrl":"https://doi.org/10.1182/blood.2025029996","url":null,"abstract":"<p><p>Multiple myeloma (MM) with t(11;14)(CCND1;IGH) remains the only subset sensitive to the BCL2 inhibitor venetoclax. Not all t(11;14)(CCND1;IGH) patients respond to treatment and some progress early after initial response. To investigate this, we interrogated 44 whole genome and exome sequencing data from 34 patients with t(11;14) MM treated with venetoclax. The presence of mutations in the RAS pathway was strongly associated with shortened progression-free survival (PFS) and was validated in an independent cohort of 21 MM patients. Presence of 1q gain was also associated with shorter PFS in patients without RAS mutations. In 10 patients with paired, pre- and post-venetoclax treatment samples, post-venetoclax progression was recurrently driven by the selection of genomic events in BCL2/MCL1 and RAS pathways and of high-risk features (e.g., loss of TP53 and CDKN2C). Overall, our study shows that comprehensive genomic profiling can identify most mechanisms underlying resistance to BCL2 inhibition in t(11;14)(CCND1;IGH) MM.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":23.1,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892048","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}