Alessandra Iurlo, Daniele Cattaneo, Umberto Gianelli, Francesco Passamonti
Atypical chronic myeloid leukemia (aCML) is a rare form of myelodysplastic (MDS)/myeloproliferative neoplasm (MPN) overlap disorder characterized by neutrophilic leukocytosis with circulating immature myeloid cells (IMC), frequent hepatosplenomegaly, and poor prognosis with high rates of leukemic transformation. In the 2022 World Health Organization (WHO) classification, aCML was therefore renamed to “MDS/MPN with neutrophilia.” Diagnostic criteria for aCML include leukocytosis ≥ 13 × 109/L, circulating IMC ≥ 10%, dysgranulopoiesis, and at least one cytopenia for MDS-thresholds (per International Consensus Classification [ICC]). Bone marrow is hypercellular due to granulocytic proliferation, associated with dysplasia in granulocytes ± other cell lines. Mutations can be used to support the diagnosis in both classifications, that is, SETBP1 and ASXL1 or SETBP1 and/or ETNK1. Absence of monocytosis, basophilia, or eosinophilia, <20% blasts, and exclusion of other MPN, MDS/MPN, and tyrosine kinase fusions are mandatory. Cytogenetic abnormalities are identified in roughly 20%–40% of aCML patients, along with a complex genomic context with high rates of recurrent somatic mutations in ASXL1, SETBP1, SRSF2, TET2, EZH2, and, less frequently, in NRAS/KRAS, CBL, CSF3R, JAK2, and ETNK1. Unfortunately, effective risk stratification systems for identifying prognostic subgroups of aCML patients are lacking, resulting in the absence of a standard of care for its management. The most used agents include hydroxyurea, interferon, hypomethylating agents, and JAK inhibitors, although none of them are disease-modifying. Allogeneic hematopoietic stem cell transplant remains the only potentially curative approach and should be considered in all eligible patients. Actionable mutations (CSF3R, NRAS/KRAS, and KIT) have also been identified, supporting the development of new agents targeting the involved pathways.
{"title":"Atypical chronic myeloid leukemia: From diagnosis to molecular features and therapeutic options","authors":"Alessandra Iurlo, Daniele Cattaneo, Umberto Gianelli, Francesco Passamonti","doi":"10.1002/hem3.70270","DOIUrl":"10.1002/hem3.70270","url":null,"abstract":"<p>Atypical chronic myeloid leukemia (aCML) is a rare form of myelodysplastic (MDS)/myeloproliferative neoplasm (MPN) overlap disorder characterized by neutrophilic leukocytosis with circulating immature myeloid cells (IMC), frequent hepatosplenomegaly, and poor prognosis with high rates of leukemic transformation. In the 2022 World Health Organization (WHO) classification, aCML was therefore renamed to “MDS/MPN with neutrophilia.” Diagnostic criteria for aCML include leukocytosis ≥ 13 × 10<sup>9</sup>/L, circulating IMC ≥ 10%, dysgranulopoiesis, and at least one cytopenia for MDS-thresholds (per International Consensus Classification [ICC]). Bone marrow is hypercellular due to granulocytic proliferation, associated with dysplasia in granulocytes ± other cell lines. Mutations can be used to support the diagnosis in both classifications, that is, <i>SETBP1</i> and <i>ASXL1</i> or <i>SETBP1</i> and/or <i>ETNK1</i>. Absence of monocytosis, basophilia, or eosinophilia, <20% blasts, and exclusion of other MPN, MDS/MPN, and tyrosine kinase fusions are mandatory. Cytogenetic abnormalities are identified in roughly 20%–40% of aCML patients, along with a complex genomic context with high rates of recurrent somatic mutations in <i>ASXL1</i>, <i>SETBP1</i>, <i>SRSF2</i>, <i>TET2</i>, <i>EZH2</i>, and, less frequently, in <i>NRAS/KRAS</i>, <i>CBL</i>, <i>CSF3R</i>, <i>JAK2</i>, and <i>ETNK1</i>. Unfortunately, effective risk stratification systems for identifying prognostic subgroups of aCML patients are lacking, resulting in the absence of a standard of care for its management. The most used agents include hydroxyurea, interferon, hypomethylating agents, and JAK inhibitors, although none of them are disease-modifying. Allogeneic hematopoietic stem cell transplant remains the only potentially curative approach and should be considered in all eligible patients. Actionable mutations (<i>CSF3R</i>, <i>NRAS/KRAS</i>, and <i>KIT</i>) have also been identified, supporting the development of new agents targeting the involved pathways.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12687300/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145722510","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lucca L. M. Derks, Konradin F. Müskens, Markus J. van Roosmalen, Aniek O. de Graaf, Nina Epskamp, Laurianne Trabut, Rico Hagelaar, Joop H. Jansen, Caroline A. Lindemans, Maaike G. J. M. van Bergen, Ruben van Boxtel, Mirjam E. Belderbos
Clonal hematopoiesis (CH), a prevalent and premalignant state in the elderly, has been detected in young individuals under selective pressures such as hematopoietic cell transplantation (HCT). However, the origin of CH and mutational processes underlying CH driver mutations in young blood systems remain unclear. Here, we used genome-wide somatic mutation profiles to retrospectively trace the origin of DNMT3A-mutant CH in three individuals, 14–41 years after childhood HCT. Both the rate and spectrum of somatic mutations in individuals with posttransplant CH were consistent with normal age-associated mutagenesis. Phylogenetic analysis revealed that DNMT3A-mutant HSPCs were present in the donor before 6.8 years of age, including during fetal development, despite being undetectable with a limit of detection of variant allele frequency of 0.001 at the time of transplantation. These findings were validated by comparing the observed mutations to expected age-dependent mutational signatures. Our results reveal that undetectable DNMT3A-mutant clones in young donors can expand into significant CH clones within decades upon transplantation. The rapid expansion of these clones in this context indicates that specific environmental pressures, rather than solely mutation acquisition, drive the development of CH.
{"title":"Posttransplantation clonal dynamics of hematopoietic stem cells carrying prenatal and early-life DNMT3A mutations","authors":"Lucca L. M. Derks, Konradin F. Müskens, Markus J. van Roosmalen, Aniek O. de Graaf, Nina Epskamp, Laurianne Trabut, Rico Hagelaar, Joop H. Jansen, Caroline A. Lindemans, Maaike G. J. M. van Bergen, Ruben van Boxtel, Mirjam E. Belderbos","doi":"10.1002/hem3.70262","DOIUrl":"10.1002/hem3.70262","url":null,"abstract":"<p>Clonal hematopoiesis (CH), a prevalent and premalignant state in the elderly, has been detected in young individuals under selective pressures such as hematopoietic cell transplantation (HCT). However, the origin of CH and mutational processes underlying CH driver mutations in young blood systems remain unclear. Here, we used genome-wide somatic mutation profiles to retrospectively trace the origin of <i>DNMT3A</i>-mutant CH in three individuals, 14–41 years after childhood HCT. Both the rate and spectrum of somatic mutations in individuals with posttransplant CH were consistent with normal age-associated mutagenesis. Phylogenetic analysis revealed that <i>DNMT3A</i>-mutant HSPCs were present in the donor before 6.8 years of age, including during fetal development, despite being undetectable with a limit of detection of variant allele frequency of 0.001 at the time of transplantation. These findings were validated by comparing the observed mutations to expected age-dependent mutational signatures. Our results reveal that undetectable <i>DNMT3A</i>-mutant clones in young donors can expand into significant CH clones within decades upon transplantation. The rapid expansion of these clones in this context indicates that specific environmental pressures, rather than solely mutation acquisition, drive the development of CH.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12686829/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721721","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kazimierz Groen, Febe Smits, Kazem Nasserinejad, Mark-David Levin, Josien C. Regelink, Gert-Jan Timmers, Esther G. M. de Waal, Matthijs Westerman, Gerjo A. Velders, Koen de Heer, Rineke B. L. Leys, Roel J. W. van Kampen, Claudia A. M. Stege, Maarten R. Seefat, Inger S. Nijhof, Ellen van der Spek, Saskia K. Klein, Niels W. C. J. van de Donk, Paula F. Ypma, Sonja Zweegman
<p>Non-transplant eligible patients with newly diagnosed multiple myeloma (NTE-NDMM) who are categorized as frail according to the International Myeloma Working Group Frailty Index (IMWG-FI) show pronounced heterogeneity in clinical characteristics, leading to differences in frailty scores—ranging from 2 to 5—based on the type and number of geriatric impairments and comorbidities. As a result, clinical outcomes may differ markedly within this subgroup.<span><sup>1</sup></span> Indeed, a post hoc analysis of the HOVON-123 trial, in which patients were treated with nine cycles of dose-adjusted melphalan, prednisone, and bortezomib, revealed that the overall survival (OS) of frail patients with an IMWG-FI of 2 had an OS comparable to intermediate-fit patients and superior to frail patients with scores of 3–5. In addition, treatment discontinuation after 9 months was considerably higher in patients with higher frailty scores.<span><sup>2</sup></span> Accordingly, in the IFM 2017-03 trial, in which daratumumab–lenalidomide was compared with lenalidomide–dexamethasone, progression free survival (PFS) was significantly prolonged in patients with a frailty score of 2 compared to those with scores of 4–5.<span><sup>3</sup></span> Notably, a similar frailty score may capture distinct aging phenotypes: for example a score of 2 may solely result from advanced age or may reflect significant geriatric impairments in a younger individual. To address this heterogeneity, frail patients may be more rationally stratified based on the underlying drivers of frailty, yielding three frail subgroups: frail-by-age (>80 years, no comorbidities or impairments in [I]ADL), frail-by-impairments (≤80 years, with comorbidities and/or impairments in [I]ADL), and ultra-frail (>80 years, with comorbidities and/or impairments in [I]ADL). The prognostic impact of these three distinct frailty subgroups on OS has been variably reported. In the HOVON-143 trial, in which patients were treated with nine cycles of ixazomib, daratumumab, and dexamethasone (IDd) followed by a maintenance phase of IDd for a maximum of 2 years, patients classified frail-by-age-alone demonstrated superior OS, although the difference did not reach statistical significance, likely due to limited sample size.<span><sup>4, 5</sup></span> In contrast, a retrospective analysis of the original IMWG-FI patient cohort reported comparable OS between patients classified frail-by-age-alone and all other frail patients.<span><sup>6</sup></span> To resolve this inconsistency, we examined the impact of frailty subtype on OS in a larger group size by pooling data of two prospective HOVON trials (HOVON-123 and HOVON-143) in NTE-NDMM patients. The studies were conducted in accordance with the Declaration of Helsinki and were approved by the institutional review board and ethics committees before study initiation. All patients provided written informed consent.</p><p>The subsets of frail NTE-NDMM patients from the HOVON-12
{"title":"Uncovering the ultra-frail: A distinct subgroup in non-transplant eligible newly diagnosed patients with multiple myeloma, with an inferior clinical outcome","authors":"Kazimierz Groen, Febe Smits, Kazem Nasserinejad, Mark-David Levin, Josien C. Regelink, Gert-Jan Timmers, Esther G. M. de Waal, Matthijs Westerman, Gerjo A. Velders, Koen de Heer, Rineke B. L. Leys, Roel J. W. van Kampen, Claudia A. M. Stege, Maarten R. Seefat, Inger S. Nijhof, Ellen van der Spek, Saskia K. Klein, Niels W. C. J. van de Donk, Paula F. Ypma, Sonja Zweegman","doi":"10.1002/hem3.70268","DOIUrl":"https://doi.org/10.1002/hem3.70268","url":null,"abstract":"<p>Non-transplant eligible patients with newly diagnosed multiple myeloma (NTE-NDMM) who are categorized as frail according to the International Myeloma Working Group Frailty Index (IMWG-FI) show pronounced heterogeneity in clinical characteristics, leading to differences in frailty scores—ranging from 2 to 5—based on the type and number of geriatric impairments and comorbidities. As a result, clinical outcomes may differ markedly within this subgroup.<span><sup>1</sup></span> Indeed, a post hoc analysis of the HOVON-123 trial, in which patients were treated with nine cycles of dose-adjusted melphalan, prednisone, and bortezomib, revealed that the overall survival (OS) of frail patients with an IMWG-FI of 2 had an OS comparable to intermediate-fit patients and superior to frail patients with scores of 3–5. In addition, treatment discontinuation after 9 months was considerably higher in patients with higher frailty scores.<span><sup>2</sup></span> Accordingly, in the IFM 2017-03 trial, in which daratumumab–lenalidomide was compared with lenalidomide–dexamethasone, progression free survival (PFS) was significantly prolonged in patients with a frailty score of 2 compared to those with scores of 4–5.<span><sup>3</sup></span> Notably, a similar frailty score may capture distinct aging phenotypes: for example a score of 2 may solely result from advanced age or may reflect significant geriatric impairments in a younger individual. To address this heterogeneity, frail patients may be more rationally stratified based on the underlying drivers of frailty, yielding three frail subgroups: frail-by-age (>80 years, no comorbidities or impairments in [I]ADL), frail-by-impairments (≤80 years, with comorbidities and/or impairments in [I]ADL), and ultra-frail (>80 years, with comorbidities and/or impairments in [I]ADL). The prognostic impact of these three distinct frailty subgroups on OS has been variably reported. In the HOVON-143 trial, in which patients were treated with nine cycles of ixazomib, daratumumab, and dexamethasone (IDd) followed by a maintenance phase of IDd for a maximum of 2 years, patients classified frail-by-age-alone demonstrated superior OS, although the difference did not reach statistical significance, likely due to limited sample size.<span><sup>4, 5</sup></span> In contrast, a retrospective analysis of the original IMWG-FI patient cohort reported comparable OS between patients classified frail-by-age-alone and all other frail patients.<span><sup>6</sup></span> To resolve this inconsistency, we examined the impact of frailty subtype on OS in a larger group size by pooling data of two prospective HOVON trials (HOVON-123 and HOVON-143) in NTE-NDMM patients. The studies were conducted in accordance with the Declaration of Helsinki and were approved by the institutional review board and ethics committees before study initiation. All patients provided written informed consent.</p><p>The subsets of frail NTE-NDMM patients from the HOVON-12","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70268","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695522","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lukas Scheller, Xiang Zhou, Henry Loeffler-Wirth, Markus Kreuz, Sofie-Katrin Kadel, Sven Schimanski, Hannah Schulze, Anna Ruckdeschel, Florian Eisele, Verena Konetzki, Maria Jornet Culubret, Maximilian Merz, Julia Mersi, Johannes Waldschmidt, Sophia Danhof, Ulrike Köhl, K. Martin Kortüm, Leo Rasche, Hermann Einsele, Johannes Düll, Max S. Topp, Michael Hudecek, Kristin Reiche, Miriam Alb
Hematotoxicity and infections are the main drivers of non-relapse mortality after chimeric antigen receptor (CAR)-T therapy. Consequently, reliable predictive biomarkers are highly needed to improve risk assessment and optimize patient management. In this study, we applied the immune-related adverse outcome pathway concept to delineate key events and risk factors of CAR-T-associated hematotoxicity. To identify predictive biomarkers, we performed flow cytometry and multiplex assays before and early after CAR-T infusion on 78 patients (ide-cel n = 31; axi-cel n = 24; and cilta-cel n = 23) undergoing CAR-T therapy. Severe hematotoxicity was linked to endothelial dysfunction, as evidenced by reduced levels of ANG1, soluble selectins, and increased soluble VCAM-1 (sVCAM-1) early after CAR-T infusion. Increased sVCAM-1, reflecting endothelial dysfunction, elevated soluble IL-2R (sIL-2R), indicating a proinflammatory state, and high tumor burden before lymphodepletion were key risk factors for CAR-T-associated hematotoxicity. Patients with elevated sVCAM-1 and sIL-2R at baseline (pre-lymphodepletion) exhibited significantly reduced overall survival (OS) (sVCAM-1; P = 0.0009), prolonged Grade 4 neutropenia (sVCAM-1; 12.1 vs. 6.0 days; P = 0.0016), more aplastic neutrophil recovery (5% vs. 30%; P = 0.007), and more severe infections (22.4% vs. 55%; P = 0.011). Baseline sIL-2R and sVCAM-1 demonstrated robust predictive value for prolonged neutropenia, severe infections, and mortality independently of key clinical variables such as the underlying disease and CAR-T product. Integration of these markers improves existing models and can help to refine risk assessment and guide individualized patient management in CAR-T therapy.
血液毒性和感染是嵌合抗原受体(CAR)-T治疗后非复发死亡率的主要驱动因素。因此,非常需要可靠的预测性生物标志物来改善风险评估和优化患者管理。在这项研究中,我们应用免疫相关不良结果通路概念来描述car - t相关血液毒性的关键事件和危险因素。为了确定预测性生物标志物,我们对78名接受CAR-T治疗的患者(idel - cell n = 31, axial - cell n = 24, cilta- cell n = 23)在CAR-T输注前后进行了流式细胞术和多重检测。CAR-T输注后早期ANG1、可溶性选择素水平降低和可溶性VCAM-1 (sVCAM-1)升高证明了严重的血液毒性与内皮功能障碍有关。反映内皮功能障碍的sVCAM-1升高、表明促炎状态的可溶性IL-2R (sIL-2R)升高以及淋巴细胞清除前的高肿瘤负荷是car - t相关血液毒性的关键危险因素。基线(淋巴细胞耗损前)sVCAM-1和sIL-2R升高的患者表现出明显降低的总生存期(OS) (sVCAM-1, P = 0.0009),延长的4级中性粒细胞减少(sVCAM-1, 12.1天对6.0天,P = 0.0016),更多的再生中性粒细胞恢复(5%对30%,P = 0.007),更严重的感染(22.4%对55%,P = 0.011)。基线sIL-2R和sVCAM-1对长期中性粒细胞减少症、严重感染和死亡率具有强大的预测价值,与潜在疾病和CAR-T产品等关键临床变量无关。这些标志物的整合改进了现有的模型,可以帮助改进CAR-T治疗中的风险评估和指导个体化患者管理。
{"title":"Endothelial dysfunction and proinflammatory state determine severe hematotoxicity and inferior outcome of CAR-T therapy","authors":"Lukas Scheller, Xiang Zhou, Henry Loeffler-Wirth, Markus Kreuz, Sofie-Katrin Kadel, Sven Schimanski, Hannah Schulze, Anna Ruckdeschel, Florian Eisele, Verena Konetzki, Maria Jornet Culubret, Maximilian Merz, Julia Mersi, Johannes Waldschmidt, Sophia Danhof, Ulrike Köhl, K. Martin Kortüm, Leo Rasche, Hermann Einsele, Johannes Düll, Max S. Topp, Michael Hudecek, Kristin Reiche, Miriam Alb","doi":"10.1002/hem3.70267","DOIUrl":"10.1002/hem3.70267","url":null,"abstract":"<p>Hematotoxicity and infections are the main drivers of non-relapse mortality after chimeric antigen receptor (CAR)-T therapy. Consequently, reliable predictive biomarkers are highly needed to improve risk assessment and optimize patient management. In this study, we applied the immune-related adverse outcome pathway concept to delineate key events and risk factors of CAR-T-associated hematotoxicity. To identify predictive biomarkers, we performed flow cytometry and multiplex assays before and early after CAR-T infusion on 78 patients (ide-cel <i>n</i> = 31; axi-cel <i>n</i> = 24; and cilta-cel <i>n</i> = 23) undergoing CAR-T therapy. Severe hematotoxicity was linked to endothelial dysfunction, as evidenced by reduced levels of ANG1, soluble selectins, and increased soluble VCAM-1 (sVCAM-1) early after CAR-T infusion. Increased sVCAM-1, reflecting endothelial dysfunction, elevated soluble IL-2R (sIL-2R), indicating a proinflammatory state, and high tumor burden before lymphodepletion were key risk factors for CAR-T-associated hematotoxicity. Patients with elevated sVCAM-1 and sIL-2R at baseline (pre-lymphodepletion) exhibited significantly reduced overall survival (OS) (sVCAM-1; P = 0.0009), prolonged Grade 4 neutropenia (sVCAM-1; 12.1 vs. 6.0 days; P = 0.0016), more aplastic neutrophil recovery (5% vs. 30%; P = 0.007), and more severe infections (22.4% vs. 55%; P = 0.011). Baseline sIL-2R and sVCAM-1 demonstrated robust predictive value for prolonged neutropenia, severe infections, and mortality independently of key clinical variables such as the underlying disease and CAR-T product. Integration of these markers improves existing models and can help to refine risk assessment and guide individualized patient management in CAR-T therapy.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12684805/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145714175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Janin Dingfelder, Michael Aigner, Jana Lindacher, Pascal Lukas, Cindy Flamann, Gina Nusser, Katharina Zimmermann, Axel Schambach, Frederik Graw, Simon Völkl, Heiko Bruns, Manuela Krumbholz, Martina Haibach, Jochen Wilke, Andreas Mackensen, Gloria Lutzny-Geier
Despite advances in targeted therapies, treatment of chronic lymphocytic leukemia (CLL) remains challenging, highlighting the urgent need for effective new therapeutic strategies. Although chimeric antigen receptor (CAR) T-cell therapy dramatically improved outcomes in acute lymphoblastic leukemia (ALL), its efficacy in CLL is limited. We hypothesize that this disparity results from pronounced CAR T-cell exhaustion and the immunosuppressive tumor microenvironment (TME) in CLL. We utilized an autologous 3D TME co-culture model to investigate the functionality of CAR T cells derived from CLL and ALL patients within physiologically relevant conditions. Our results revealed increased exhaustion levels and diminished cytotoxicity of CAR T cells from CLL patients compared to those from ALL patients. Importantly, combining CAR T-cell treatment with interleukin-10 (IL-10) or CXCR4 blockade effectively improved cytotoxicity against CLL cells, even in stromal-protected regions within the 3D model. These findings offer insights into CAR T-cell dysfunction in CLL and support novel TME-targeted combination strategies to improve clinical outcomes.
{"title":"Functional differences between CLL- and ALL-derived CAR T cells in a 3D tumor microenvironment highlight CXCR4 and IL-10 as potential modulatory targets","authors":"Janin Dingfelder, Michael Aigner, Jana Lindacher, Pascal Lukas, Cindy Flamann, Gina Nusser, Katharina Zimmermann, Axel Schambach, Frederik Graw, Simon Völkl, Heiko Bruns, Manuela Krumbholz, Martina Haibach, Jochen Wilke, Andreas Mackensen, Gloria Lutzny-Geier","doi":"10.1002/hem3.70279","DOIUrl":"10.1002/hem3.70279","url":null,"abstract":"<p>Despite advances in targeted therapies, treatment of chronic lymphocytic leukemia (CLL) remains challenging, highlighting the urgent need for effective new therapeutic strategies. Although chimeric antigen receptor (CAR) T-cell therapy dramatically improved outcomes in acute lymphoblastic leukemia (ALL), its efficacy in CLL is limited. We hypothesize that this disparity results from pronounced CAR T-cell exhaustion and the immunosuppressive tumor microenvironment (TME) in CLL. We utilized an autologous 3D TME co-culture model to investigate the functionality of CAR T cells derived from CLL and ALL patients within physiologically relevant conditions. Our results revealed increased exhaustion levels and diminished cytotoxicity of CAR T cells from CLL patients compared to those from ALL patients. Importantly, combining CAR T-cell treatment with interleukin-10 (IL-10) or CXCR4 blockade effectively improved cytotoxicity against CLL cells, even in stromal-protected regions within the 3D model. These findings offer insights into CAR T-cell dysfunction in CLL and support novel TME-targeted combination strategies to improve clinical outcomes.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145714136","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jean Galtier, Pierre Sesques, Vivien Dupont, Emmanuel Bachy, Roberta Di Blasi, Catherine Thieblemont, Gabriel Brisou, Thomas Gastinne, Guillaume Cartron, François-Xavier Gros, Franck Morschhauser, Axel André, Jacques-Olivier Bay, Magalie Joris, Aline Tanguy-Schmidt, Audrey Demailly, Steven Le Gouill, Roch Houot, Krimo Bouabdallah, Vincent Camus
<p>Primary mediastinal B-cell lymphoma (PMBL) is a rare subset of aggressive lymphoma with excellent outcome after dose-dense frontline immunochemotherapy but a historically dismal prognosis at relapse.<span><sup>1-4</sup></span> Leveraging data from the nation-wide prospective DESCAR-T registry, in which all patients who received commercial CAR-T cells in France as a treatment for a hematologic malignancy were included, we recently showed that relapsed or refractory (R/R) PMBL receiving anti-CD19 CAR-T cell infusion exhibited a high complete response (CR) rate and satisfactory survivals.<span><sup>5</sup></span> This was particularly meaningful for the 62 patients treated with axicabtagene-ciloleucel (axi-cel), for whom 2 years progression-free survival (PFS) and overall survival (OS) rate reached 70.4% and 86.9%, respectively, and compared favorably with more than 1000 other patients with R/R diffuse large B-cell lymphoma (DLBCL) receiving the same treatment. These data, along with other evidence,<span><sup>6-8</sup></span> strongly support anti-CD19 CAR-T cells as current standard-of-care for R/R PMBL. However, PMBL patients experiencing CAR-T cells therapy failure still represent an unmet clinical need and their outcomes have not been described so far. Albeit a previous work from DESCAR-T registry did describe the very poor OS of patients with DLBCL progressing after CAR-T cell therapy,<span><sup>9</sup></span> recent changes in the treatment paradigm of R/R PMBL could translate into unexpected outcomes even in very advanced lines and their clinical course need to be specifically studied. Notably, anti-PD1 checkpoint inhibitors (CPI) demonstrated promising efficacy in heavily pre-treated PMBL, and association with brentuximab-vedotin (Bv) may even increase response rate and survivals.<span><sup>10-13</sup></span></p><p>To address this question, we analyzed the clinical course of patients experiencing treatment failure in the CARTHYM study, whose procedures have been previously described.<span><sup>5</sup></span> Briefly, it leveraged the data of all adults patients with a diagnosis of PMBL confirmed by an expert hematopathologist from the LYMPHOPATH network<span><sup>14</sup></span> who received an anti-CD19 CAR-T cell infusion in France within the prospective DESCAR-T registry (NCT04328298), between January 2018 and February 2024. All patients provided written informed consent. The study was conducted in accordance with the Declaration of Helsinki and approved by the local ethics committees of each participating center. The CARTHYM study included 82 patients with R/R PMBL treated across 21 centers: 62 received axi-cel, 14 received tisagenlecleucel (tisa-cel), and 6 received lisocabtagene maraleucel (liso-cel). After a median follow-up of 21 months, 27 progression events were observed. These 27 patients, treated across 13 different centers, constitute the study population analyzed in the present report.</p><p>Characteristics of the 27 patient
{"title":"Outcomes and treatment patterns of patients with primary mediastinal B-cell lymphoma after CAR-T cell therapy failure: A DESCAR-T analysis","authors":"Jean Galtier, Pierre Sesques, Vivien Dupont, Emmanuel Bachy, Roberta Di Blasi, Catherine Thieblemont, Gabriel Brisou, Thomas Gastinne, Guillaume Cartron, François-Xavier Gros, Franck Morschhauser, Axel André, Jacques-Olivier Bay, Magalie Joris, Aline Tanguy-Schmidt, Audrey Demailly, Steven Le Gouill, Roch Houot, Krimo Bouabdallah, Vincent Camus","doi":"10.1002/hem3.70263","DOIUrl":"10.1002/hem3.70263","url":null,"abstract":"<p>Primary mediastinal B-cell lymphoma (PMBL) is a rare subset of aggressive lymphoma with excellent outcome after dose-dense frontline immunochemotherapy but a historically dismal prognosis at relapse.<span><sup>1-4</sup></span> Leveraging data from the nation-wide prospective DESCAR-T registry, in which all patients who received commercial CAR-T cells in France as a treatment for a hematologic malignancy were included, we recently showed that relapsed or refractory (R/R) PMBL receiving anti-CD19 CAR-T cell infusion exhibited a high complete response (CR) rate and satisfactory survivals.<span><sup>5</sup></span> This was particularly meaningful for the 62 patients treated with axicabtagene-ciloleucel (axi-cel), for whom 2 years progression-free survival (PFS) and overall survival (OS) rate reached 70.4% and 86.9%, respectively, and compared favorably with more than 1000 other patients with R/R diffuse large B-cell lymphoma (DLBCL) receiving the same treatment. These data, along with other evidence,<span><sup>6-8</sup></span> strongly support anti-CD19 CAR-T cells as current standard-of-care for R/R PMBL. However, PMBL patients experiencing CAR-T cells therapy failure still represent an unmet clinical need and their outcomes have not been described so far. Albeit a previous work from DESCAR-T registry did describe the very poor OS of patients with DLBCL progressing after CAR-T cell therapy,<span><sup>9</sup></span> recent changes in the treatment paradigm of R/R PMBL could translate into unexpected outcomes even in very advanced lines and their clinical course need to be specifically studied. Notably, anti-PD1 checkpoint inhibitors (CPI) demonstrated promising efficacy in heavily pre-treated PMBL, and association with brentuximab-vedotin (Bv) may even increase response rate and survivals.<span><sup>10-13</sup></span></p><p>To address this question, we analyzed the clinical course of patients experiencing treatment failure in the CARTHYM study, whose procedures have been previously described.<span><sup>5</sup></span> Briefly, it leveraged the data of all adults patients with a diagnosis of PMBL confirmed by an expert hematopathologist from the LYMPHOPATH network<span><sup>14</sup></span> who received an anti-CD19 CAR-T cell infusion in France within the prospective DESCAR-T registry (NCT04328298), between January 2018 and February 2024. All patients provided written informed consent. The study was conducted in accordance with the Declaration of Helsinki and approved by the local ethics committees of each participating center. The CARTHYM study included 82 patients with R/R PMBL treated across 21 centers: 62 received axi-cel, 14 received tisagenlecleucel (tisa-cel), and 6 received lisocabtagene maraleucel (liso-cel). After a median follow-up of 21 months, 27 progression events were observed. These 27 patients, treated across 13 different centers, constitute the study population analyzed in the present report.</p><p>Characteristics of the 27 patient","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683939/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145714115","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Francesca Maria Quaglia, Annalisa Arcari, Lucia Morello, Luigi Marcheselli, Alessandra Tucci, Chiara Pagani, Valentina Bozzoli, Greta Scapinello, Francesco Piazza, Vittorio Ruggero Zilioli, Cristina Muzi, Benedetta Puccini, Benedetta Sordi, Maria Chiara Tisi, Nicolò Rampi, Alessia Castellino, Sara Veronica Usai, Jari Paternoster, Emanuele Cencini, Simone Ferrero, Chiara Consoli, Marco Basso, Elisa Lucchini, Elsa Pennese, Maria Elena Nizzoli, Michele Spina, Luca Pezzullo, Maria Stella De Candia, Rita Tavarozzi, Mauro Krampera, Carlo Visco
<p>Mantle cell lymphoma (MCL) is an aggressive and incurable disease.<span><sup>1, 2</sup></span> Most patients are over 65 years old and exhibit heterogeneous fitness status alongside multiple comorbidities. Until now, conventional chemo-immunotherapy (CIT) followed by rituximab maintenance has represented the standard approach for elderly patients.<span><sup>3</sup></span> Approximately 40% of MCL patients experience disease relapse or progression (POD) within 24 months from diagnosis and have been defined as early-POD.<span><sup>4</sup></span> The MANTLE-FIRST study has divided patients' outcome depending on time to first relapse (early- vs. late-POD) after intensive upfront therapy including high-dose cytarabine.<span><sup>5, 6</sup></span> However, the predictive significance of early-POD in elderly MCL patients following less intensive frontline treatment is not well established. Recent studies have addressed this topic, but only as pooled analysis from clinical trials or retrospective series including also young patients.<span><sup>7-9</sup></span></p><p>In our retrospective real-life study, we evaluated the prognostic significance of early-POD specifically among relapsed/refractory (r/r) MCL patients ≥ 65 years old. We evaluated progression-free survival and overall survival, estimated from the start of salvage therapy (PFS-2, OS-2) in the whole cohort and according to different second-line treatments. We also evaluated PFS-2 and OS-2 in relation to a simplified geriatric assessment (sGA) tool, uniformly assessed in all participating Centers (Table S1).<span><sup>10</sup></span> Patient selection/statistics/ethics are reported in the Supporting Information.</p><p>We included 231 patients ≥65 years old with r/r disease after rituximab-based non-intensive induction regimens, who were treated at relapse with at least one cycle of systemic therapy (Bruton's tyrosine kinase inhibitor [BTKi] vs. CIT vs. other) between 2007 and 2021 in 21 centers belonging to the Fondazione Italiana Linfomi (FIL) (Table S2). Overall, 121 patients (53%) were defined as early-POD. Patients with older age at diagnosis, male sex, Eastern Cooperative Oncology Group performance status (ECOG PS) > 1, high lactate dehydrogenase and mantle cell lymphoma international prognostic index (MIPI), Ki67 > 30%, and nonclassical histology were more represented in the early-POD subgroup (Table S3). Frontline treatments included rituximab–bendamustine (RB = 102 patients, 45%), rituximab, bendamustine, and cytarabine (R-BAC = 67 patients, 29%), and rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP/R-CHOP-like = 60 patients, 26%). Patients treated with RB were older compared to R-BAC and R-CHOP, with a worse fitness profile. We showed more interruptions/dose reductions during R-BAC, with a higher incidence of hematological adverse events (AEs) (Table S4). Table S5 describes the impact of patients' characteristics/type of first-line treatment on time
套细胞淋巴瘤(MCL)是一种侵袭性且无法治愈的疾病。1,2大多数患者年龄超过65岁,表现出不同的健康状况,并伴有多种合并症。到目前为止,传统的化疗免疫治疗(CIT)加美罗华维持是老年患者的标准治疗方法大约40%的MCL患者在诊断后24个月内出现疾病复发或进展(POD),并被定义为早期PODMANTLE-FIRST研究根据患者在接受包括高剂量阿糖胞苷在内的强化前期治疗后首次复发的时间(早期与晚期pod)对结果进行了划分。5,6然而,早期pod在低强度一线治疗的老年MCL患者中的预测意义尚不明确。最近的研究已经解决了这个问题,但只是作为临床试验或回顾性系列的汇总分析,其中也包括年轻患者。在我们的回顾性现实研究中,我们评估了早期pod在≥65岁的复发/难治性(r/r) MCL患者中的预后意义。我们评估了无进展生存期和总生存期,从整个队列的挽救治疗开始(PFS-2, OS-2)估计,并根据不同的二线治疗。我们还评估了PFS-2和OS-2与简化老年评估(sGA)工具的关系,在所有参与中心进行统一评估(表S1) 10患者选择/统计/伦理在辅助信息中报告。我们纳入了231例≥65岁的r/r疾病患者,他们接受了基于利妥昔单抗的非强化诱导方案,这些患者在2007年至2021年间复发时接受了至少一个周期的全身治疗(布鲁顿酪氨酸激酶抑制剂[BTKi]与CIT与其他),这些患者属于意大利林福米基金会(FIL)的21个中心(表S2)。总体而言,121例(53%)患者被定义为早期pod。诊断时年龄较大、性别为男性、Eastern Cooperative Oncology Group performance status (ECOG PS) >; 1、高乳酸脱氢酶和套细胞淋巴瘤国际预后指数(MIPI)、Ki67 >; 30%、非经典组织学在早期pod亚组中更有代表性(表S3)。一线治疗包括利妥昔单抗-苯达莫司汀(RB = 102例,45%),利妥昔单抗-苯达莫司汀-阿糖胞苷(R-BAC = 67例,29%),利妥昔单抗-环磷酰胺-阿霉素-长春新碱-泼尼松(R-CHOP/R-CHOP样= 60例,26%)。与R-BAC和R-CHOP相比,接受RB治疗的患者年龄更大,健康状况更差。我们发现在R-BAC期间有更多的中断/剂量减少,血液不良事件(ae)的发生率更高(表S4)。表S5描述了患者特征/一线治疗类型对POD时间的影响,突出了年龄、ECOG PS >; 1和Ki67 >; 30%的相关性。然后我们关注不同二线药物的安全性和有效性。患者复发时的特征(n = 223)见表1。总体而言,124名患者(56%)接受了BTKi治疗(122名依鲁替尼,1名扎鲁替尼和1名阿卡拉布替尼)。BTKi治疗的患者比补救性CIT治疗的患者年龄大(中位年龄77比75,p值= 0.026,40%≥80岁),超过50%的患者不健康/虚弱。其余临床特征分布均匀。BTKi血液学毒性较小,但血液学外ae略高。心脏毒性在BTKi亚组中更为常见(4例心房颤动,2例心力衰竭,1例急性心肌缺血)。在90例停止使用BTKi的患者中,61例(68%)的原因是进展,9例(10%)的原因是不耐受,20例(22%)的原因是其他原因(n = 16未知,n = 3继发性肿瘤,n = 1异基因移植桥)。PFS-2和OS-2见图S1。中位随访4.6年,中位PFS-2为1.0年(95% CI 0.76-1.3),中位OS-2为2.2年(95% CI 1.6-2.6)。死亡原因为106例进展(69%,n = 1例中枢神经系统),12例感染(8%),6例继发性癌症(4%),4例心力衰竭(3%),1例肾衰竭,23例未知(15%)。不同二线处理的PFS-2和OS-2如图1A、B所示。接受BTKi治疗的患者中位PFS-2为1.39年,3年PFS-2 (3y-PFS-2)为24%,而接受CIT治疗的患者中位PFS-2为0.98年,3y-PFS-2为11%(风险比[HR] 1.43, 95% CI 1.02-2.00, p值= 0.037)。通过MIPI和早期pod等混杂因素调整二线治疗的Cox回归结果如表S6所示,证实了BTKi对PFS-2的独立有利影响。在OS-2方面,BTKi没有显示出显著优势(OS-2中位数为2.10年,3年OS-2 [3y-OS-2]为36%,而CIT的OS-2中位数为3.16年,3y-OS-2为53%;HR 0.82, 95% CI 0.57-1.20, p值= 0.314)。 然后,我们评估到POD的时间对生存结果的作用(图1C,D)。早期pod患者的中位PFS-2为0.66年,晚期pod患者的中位PFS-2为1.47年(HR 1.49, 95% CI 1.11-2.01, p值= 0.008)。早期pod的中位OS-2为1.44年,晚期pod的中位OS-2为3.17年(HR 1.55, 95% CI 1.12-2.15, p值= 0.009)。众所周知的预后因素如ECOG PS、MIPI和中枢神经系统受累证实了它们在复发中的作用(表S7和S8)。不适合/虚弱的患者表现出明显更差的结果,中位PFS-2为0.76年(95% CI 0.41-1.16),而适合患者的中位PFS-2为1.16年(95% CI 0.92-1.95)。3y-PFS-2在不适合/虚弱患者中为11% (95% CI 5-20),而在适合患者中为32% (95% CI 22-42) (p值= 0.007)。3y-OS-2为26% (95% CI 17-36),而拟合病例为47% (95% CI 35-57) (p值= 0.003)(图1E,F)。最后,我们考虑了sGA与POD时间之间的相互作用,以3y-PFS-2和3y-OS-2评估不同亚组的结果。晚期复发的Fit患者(Fit- late亚组)在PFS-2和OS-2方面预后最好(分别为34%和62%)。在不健康/虚弱的患者中,无论POD时间如何,3y-PFS-2和OS-2非常相似(表S9)。在多变量分析中,二线治疗CIT(与BTKi相比)、不健康/虚弱状态(pod早期和晚期)、高MIPI和复发时的中枢神经系统疾病与更差的PFS-2相关(表S10)。就OS-2而言,适合的早期pod患者和不适合/虚弱的患者(包括早期和晚期pod),以及高MIPI和CNS复发,证实了它们的独立预后意义。在多变量分析中,BTKi治疗与PFS-2改善相关,但与OS-2无关。图S2显示了通过多重Cox回归预测的PFS-2和OS-2,校正了sGA-POD、MIPI和复发时中枢神经系统疾病。144例接受二线治疗的r/r患者仅获得OS数据。中位OS-3(第二次复发后的总生存期)为6.4个月(95% CI 4.4-10.4)。35例患者接受BTKi作为三线治疗,仅有6例患者在既往BTKi治疗后再次接受治疗,而23例和9例患者曾接受CIT或其他方案治疗。如图S3所示,在不同既往治疗后接受BTKi作为三线治疗的患者预后较差(中位OS-3为20个月)。在我们的研究中,嵌合抗原受体T细胞(CAR-T细胞)和匹托鲁替尼在意大利还不能用于r/r m
{"title":"The role of time to first progression and fitness status in elderly patients with mantle cell lymphoma: Results from the ELDERLY MANTLE-FIRST study","authors":"Francesca Maria Quaglia, Annalisa Arcari, Lucia Morello, Luigi Marcheselli, Alessandra Tucci, Chiara Pagani, Valentina Bozzoli, Greta Scapinello, Francesco Piazza, Vittorio Ruggero Zilioli, Cristina Muzi, Benedetta Puccini, Benedetta Sordi, Maria Chiara Tisi, Nicolò Rampi, Alessia Castellino, Sara Veronica Usai, Jari Paternoster, Emanuele Cencini, Simone Ferrero, Chiara Consoli, Marco Basso, Elisa Lucchini, Elsa Pennese, Maria Elena Nizzoli, Michele Spina, Luca Pezzullo, Maria Stella De Candia, Rita Tavarozzi, Mauro Krampera, Carlo Visco","doi":"10.1002/hem3.70254","DOIUrl":"https://doi.org/10.1002/hem3.70254","url":null,"abstract":"<p>Mantle cell lymphoma (MCL) is an aggressive and incurable disease.<span><sup>1, 2</sup></span> Most patients are over 65 years old and exhibit heterogeneous fitness status alongside multiple comorbidities. Until now, conventional chemo-immunotherapy (CIT) followed by rituximab maintenance has represented the standard approach for elderly patients.<span><sup>3</sup></span> Approximately 40% of MCL patients experience disease relapse or progression (POD) within 24 months from diagnosis and have been defined as early-POD.<span><sup>4</sup></span> The MANTLE-FIRST study has divided patients' outcome depending on time to first relapse (early- vs. late-POD) after intensive upfront therapy including high-dose cytarabine.<span><sup>5, 6</sup></span> However, the predictive significance of early-POD in elderly MCL patients following less intensive frontline treatment is not well established. Recent studies have addressed this topic, but only as pooled analysis from clinical trials or retrospective series including also young patients.<span><sup>7-9</sup></span></p><p>In our retrospective real-life study, we evaluated the prognostic significance of early-POD specifically among relapsed/refractory (r/r) MCL patients ≥ 65 years old. We evaluated progression-free survival and overall survival, estimated from the start of salvage therapy (PFS-2, OS-2) in the whole cohort and according to different second-line treatments. We also evaluated PFS-2 and OS-2 in relation to a simplified geriatric assessment (sGA) tool, uniformly assessed in all participating Centers (Table S1).<span><sup>10</sup></span> Patient selection/statistics/ethics are reported in the Supporting Information.</p><p>We included 231 patients ≥65 years old with r/r disease after rituximab-based non-intensive induction regimens, who were treated at relapse with at least one cycle of systemic therapy (Bruton's tyrosine kinase inhibitor [BTKi] vs. CIT vs. other) between 2007 and 2021 in 21 centers belonging to the Fondazione Italiana Linfomi (FIL) (Table S2). Overall, 121 patients (53%) were defined as early-POD. Patients with older age at diagnosis, male sex, Eastern Cooperative Oncology Group performance status (ECOG PS) > 1, high lactate dehydrogenase and mantle cell lymphoma international prognostic index (MIPI), Ki67 > 30%, and nonclassical histology were more represented in the early-POD subgroup (Table S3). Frontline treatments included rituximab–bendamustine (RB = 102 patients, 45%), rituximab, bendamustine, and cytarabine (R-BAC = 67 patients, 29%), and rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP/R-CHOP-like = 60 patients, 26%). Patients treated with RB were older compared to R-BAC and R-CHOP, with a worse fitness profile. We showed more interruptions/dose reductions during R-BAC, with a higher incidence of hematological adverse events (AEs) (Table S4). Table S5 describes the impact of patients' characteristics/type of first-line treatment on time ","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70254","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sotiroula Chatzimatthaiou, Fedele Bonifazi, Anna C. Gimbert, Raffaella Colombatti, Francesco Cremonesi, Andreas Glenthøj, Elisabetta Mezzalira, Coralea Stephanou, Jan Traeger-Synodinos, Darko Antic, Burak Durmaz, Eleni Gavriilaki, Baba Inusa, Annalisa Landi, Mariangela Pellegrini, Francesca Basile, Stephanie Muth, Holger Cario, Eleni Katsantoni, Dilem Ruhluel, Carsten Werner Lederer, María del Mar Mañú-Pereira, Petros Kountouris
<p>Recent decades have witnessed remarkable advances in hematology research,<span><sup>1</sup></span> and yet, for many affected populations, particularly those with sickle cell disease (SCD) and Thalassemia, more so in low-resource settings, clinical outcomes remain suboptimal.<span><sup>2</sup></span> Despite being among the most common inherited disorders globally, with an estimated 330,000 conceptions annually,<span><sup>3</sup></span> patients with hemoglobinopathies encounter fragmented care systems and unequal access to diagnosis and treatment, and there is limited coordination in research and clinical practices across countries.<span><sup>4</sup></span></p><p>Although hemoglobinopathies have gained recognition as a global public health concern,<span><sup>3</sup></span> there are still four persistent challenges. First, there are only a limited number of harmonized, multicentric studies that capture the genetic and clinical variability of these disorders to enable personalized approaches in clinical management and treatment.<span><sup>5</sup></span> Second, care standards for hemoglobinopathies remain inconsistent, with substantial gaps between and within countries.<span><sup>6</sup></span> Third, there is limited access to structured training and clinical education, particularly in countries with underdeveloped health systems.<span><sup>7</sup></span> Finally, a fragmented research landscape has impeded the development of innovative, cross-border strategies that could guide policy and practice.<span><sup>8</sup></span> These limitations are compounded by poor coordination globally, restricting the sharing of knowledge and the development of common standards.</p><p>The HELIOS Action (Haemoglobinopathies in European Liaison of Medicine and Science, www.heliosaction.eu) was launched in 2023 to address these persistent gaps, through networking and training activities. Funded by the European Cooperation in Science and Technology (COST), HELIOS is a collaborative open network designed to strengthen research infrastructure, standardize care practices, and promote inclusive participation in hemoglobinopathy-related science and policy. HELIOS thus provides a coordinated and inclusive platform and facilitates concerted action to tackle major challenges in hemoglobinopathies. Specifically, HELIOS fosters multicentric collaboration and genetic mapping to address the lack of harmonized research, develops regionally adapted clinical protocols to reduce inconsistent standards, expands structured training programs to close education gaps, and strengthens international data sharing and policy engagement to overcome fragmentation. To date, the Action involves over 245 members from 36 countries (Figure 1), and continues to grow, with participants spanning Europe, Africa, Asia, North America, and Australia, reflecting the global relevance of these disorders.</p><p>A key strength of the HELIOS Action lies in its inclusive and collaborative structure. As a glo
{"title":"HELIOS Action: Advancing research, education, and equity in hemoglobinopathies across Europe and beyond","authors":"Sotiroula Chatzimatthaiou, Fedele Bonifazi, Anna C. Gimbert, Raffaella Colombatti, Francesco Cremonesi, Andreas Glenthøj, Elisabetta Mezzalira, Coralea Stephanou, Jan Traeger-Synodinos, Darko Antic, Burak Durmaz, Eleni Gavriilaki, Baba Inusa, Annalisa Landi, Mariangela Pellegrini, Francesca Basile, Stephanie Muth, Holger Cario, Eleni Katsantoni, Dilem Ruhluel, Carsten Werner Lederer, María del Mar Mañú-Pereira, Petros Kountouris","doi":"10.1002/hem3.70258","DOIUrl":"https://doi.org/10.1002/hem3.70258","url":null,"abstract":"<p>Recent decades have witnessed remarkable advances in hematology research,<span><sup>1</sup></span> and yet, for many affected populations, particularly those with sickle cell disease (SCD) and Thalassemia, more so in low-resource settings, clinical outcomes remain suboptimal.<span><sup>2</sup></span> Despite being among the most common inherited disorders globally, with an estimated 330,000 conceptions annually,<span><sup>3</sup></span> patients with hemoglobinopathies encounter fragmented care systems and unequal access to diagnosis and treatment, and there is limited coordination in research and clinical practices across countries.<span><sup>4</sup></span></p><p>Although hemoglobinopathies have gained recognition as a global public health concern,<span><sup>3</sup></span> there are still four persistent challenges. First, there are only a limited number of harmonized, multicentric studies that capture the genetic and clinical variability of these disorders to enable personalized approaches in clinical management and treatment.<span><sup>5</sup></span> Second, care standards for hemoglobinopathies remain inconsistent, with substantial gaps between and within countries.<span><sup>6</sup></span> Third, there is limited access to structured training and clinical education, particularly in countries with underdeveloped health systems.<span><sup>7</sup></span> Finally, a fragmented research landscape has impeded the development of innovative, cross-border strategies that could guide policy and practice.<span><sup>8</sup></span> These limitations are compounded by poor coordination globally, restricting the sharing of knowledge and the development of common standards.</p><p>The HELIOS Action (Haemoglobinopathies in European Liaison of Medicine and Science, www.heliosaction.eu) was launched in 2023 to address these persistent gaps, through networking and training activities. Funded by the European Cooperation in Science and Technology (COST), HELIOS is a collaborative open network designed to strengthen research infrastructure, standardize care practices, and promote inclusive participation in hemoglobinopathy-related science and policy. HELIOS thus provides a coordinated and inclusive platform and facilitates concerted action to tackle major challenges in hemoglobinopathies. Specifically, HELIOS fosters multicentric collaboration and genetic mapping to address the lack of harmonized research, develops regionally adapted clinical protocols to reduce inconsistent standards, expands structured training programs to close education gaps, and strengthens international data sharing and policy engagement to overcome fragmentation. To date, the Action involves over 245 members from 36 countries (Figure 1), and continues to grow, with participants spanning Europe, Africa, Asia, North America, and Australia, reflecting the global relevance of these disorders.</p><p>A key strength of the HELIOS Action lies in its inclusive and collaborative structure. As a glo","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70258","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lydia O. Okoibhole, Funmi Dasaolu, Zainab Garba-Sani, Stephen P. Hibbs
<p>Recent advances in gene therapy and expanded access for haematopoietic stem cell transplantation (HSCT) have made conversations about a <i>cure</i> more pertinent for individuals living with sickle cell disease (SCD). These advances, including the approval of exa-cel gene therapy in the United Kingdom,<span><sup>1</sup></span> have amplified the language of cure within policy, media, and clinical discussions. However, as this language becomes more visible, questions emerge about what <i>cure</i> means for individuals living with SCD, for healthcare systems, and for communities historically underserved and marginalized within research and clinical care.</p><p>The term <i>cure</i>, derived from the Latin “curare,” meaning “to care for” or “to heal,” suggests the complete elimination of symptoms caused by disease and a restoration to a previous state of health,<span><sup>2</sup></span> but in the context of a lifelong disease like SCD, the meaning is more complex.</p><p>In this article, we begin with the personal reflections of one author (F.D.) living with SCD as she grapples with the possibility and uncertainty of cellular therapies (Box 1). Next, we survey how the language of cure is used in public discussions about SCD cellular therapies. We share the account of one author (Z.G.-S.) whose experience befits the word <i>cure</i>, before exploring complexities that challenge the use of curative language. Finally, we highlight important considerations for health professionals and health systems relating to cellular therapies for SCD.</p><p>The last decade has seen a significant increase in discussion about curative therapies in SCD. In 2023, when the US Food and Drug Administration (FDA) approved the gene editing therapy exa-cel, it was described as “the first CRISPR cure for a genetic disease.”<span><sup>3, 4</sup></span> Similarly, in 2025, the UK National Institute for Health and Care Excellence (NICE) described exa-cel's approval as a “potential cure for some people with severe sickle cell disease.”<span><sup>5</sup></span> This approval followed a long and complex journey of regulatory review and appeal,<span><sup>6</sup></span> which finally resulted in the National Health Service (NHS) funding this one-time therapy,<span><sup>5</sup></span> described by NHS England as a source of hope.<span><sup>1</sup></span></p><p>Additionally, conversation continues to build around allogeneic HSCT in SCD, as developments in conditioning regimens increase accessibility for a much wider group of people, including older recipients, those without a fully matched donor and those with additional comorbidities.<span><sup>7</sup></span> In a patient guide addressing SCD and HSCT, UK charities Anthony Nolan and the Sickle Cell Society described the treatment as a cure, while also defining the benefits and risks associated.<span><sup>8</sup></span> However, medical regulators, healthcare professionals, and media reports rarely explicitly define cure, and the pote
基因治疗的最新进展和造血干细胞移植(HSCT)的广泛应用使得关于镰状细胞病(SCD)患者治疗方法的讨论更加切合实际。这些进步,包括英国批准的exa- cell基因疗法1,扩大了在政策、媒体和临床讨论中的治愈语言。然而,随着这种语言变得越来越明显,关于治疗对SCD患者、医疗保健系统以及历史上在研究和临床护理中服务不足和边缘化的社区意味着什么的问题出现了。“治愈”一词源于拉丁语“curare”,意思是“照顾”或“治愈”,暗示着完全消除由疾病引起的症状,恢复到以前的健康状态,但在像SCD这样的终身疾病的背景下,其含义更为复杂。在本文中,我们从一位患有SCD的作者(F.D.)的个人反思开始,她努力应对细胞治疗的可能性和不确定性(方框1)。接下来,我们调查了治愈的语言是如何在关于SCD细胞治疗的公开讨论中使用的。在探索挑战治疗语言使用的复杂性之前,我们分享一位作者(zg.s.)的经历,他的经历适合“治疗”这个词。最后,我们强调了与SCD细胞治疗相关的卫生专业人员和卫生系统的重要考虑。在过去的十年中,关于SCD治疗方法的讨论显著增加。2023年,当美国食品和药物管理局(FDA)批准基因编辑疗法exa-cel时,它被描述为“首个CRISPR治疗遗传病的方法”。同样,在2025年,英国国家健康与护理卓越研究所(NICE)将exa-cel的批准描述为“一些患有严重镰状细胞病的人的潜在治疗方法”。这一批准经历了漫长而复杂的监管审查和上诉过程,最终导致英国国家医疗服务体系(NHS)资助了这种一次性疗法,英国国家医疗服务体系(NHS England)将其描述为希望之源。此外,随着治疗方案的发展,更广泛的人群(包括老年受者、没有完全匹配供体的人以及有其他合并症的人)可以获得同种异体造血干细胞移植,围绕SCD的对话继续建立在一份关于SCD和HSCT的患者指南中,英国慈善机构Anthony Nolan和镰状细胞协会将这种治疗描述为一种治愈方法,同时也定义了相关的益处和风险然而,医疗监管机构、医疗保健专业人员和媒体报道很少明确定义治疗,误解的可能性很大。每一种定义都有不同的关注点:经济的、生物医学的或整体的,这就提出了一个问题:在SCD中,治愈到底意味着什么?细胞疗法具有变革性。对于那些因疼痛危机、每隔几周输血或器官并发症进行性加重而多次入院的患者来说,成功的治疗意味着摆脱这些负担。细胞疗法已被证明能显著减少甚至消除血管闭塞危象,提高血红蛋白水平,并防止进一步的器官损伤病人描述说,他们能够带着一种新的稳定和独立的感觉回到教育、工作和家庭生活中去一些人所经历的这种转变反映在“治愈”这个词中。此外,治愈的语言可以提高患者群体的可见度和希望,他们长期遭受研究投资不足,缺乏治疗方法和其他新治疗方案的退出。下面,其中一位作者(z.g.s.)分享了为什么cure准确地描述了她在HSCT后的生活(方框2)。细胞疗法不能治愈所有的身体症状。例如,许多人患有SCD的并发症,如肾病、肝病或认知障碍,细胞治疗可能会稳定这些并发症,但不能逆转此外,包括化疗、免疫治疗和放疗在内的调理方案可能会造成新的身体损伤,如不孕症、继发性癌症和其他长期并发症。如果接受同种异体造血干细胞移植的个体发生慢性移植物抗宿主病,他们将一种严重的慢性疾病换成另一种此外,可能还会出现额外的危害,例如报道的SCD基因治疗后与髓系肿瘤相关的潜在驱动突变的增加这些临床局限性反映在生活经验中。治疗语言和生活现实之间的紧张关系在Genesis Jones的故事中得到了体现,她在美国接受了SCD的HSCT,尽管她的移植手术成功植入,琼斯随后被诊断出患有继发性癌症。 她继续与慢性疼痛和以前由SCD引起的器官损伤症状作斗争。此外,一旦她不再被归类为患有SCD,她就无法获得某些形式的医疗保健。琼斯报告说,她在治疗后与自己的身份和心理健康作斗争,感觉“处于中间”,不再是SCD社区的真正一部分由于人们生来就患有SCD,它从出生起就深刻地塑造了身份,影响了个人叙事、社会互动和社区动态尽管SCD的治疗通常以治愈为框架,但它需要患者协商一种新的身份,这种身份是由他们的病情遗留问题和治疗可能带来的转变形成的。现有的与家庭成员、朋友和照顾者的关系可能会中断,需要进行重大调整。15,16正如琼斯的经历所表明的,一旦一个人被认为“治愈了”,这可能会限制他们获得的支持。例如,一个人在治疗前可能有资格获得残疾经济支持,但在治疗后就不再有资格了。然而,SCD的长期并发症,包括慢性疼痛,可能仍然深刻地影响着他们的日常生活,这突显了生物医学治疗可能无法完全符合生活现实或持续的支持需求。在细胞治疗SCD的背景下,治愈是最合适的术语吗?对于一些接受这些治疗的人来说,包括其中一位作者,答案是肯定的——治愈是一个有用的、必要的词,它既反映了这种治疗的严重性,也为更广泛的社区带来了急需的希望和关注。然而,对其他人来说,治愈语言可能会产生错误的期望,破坏幸存者、他们的扩展社区和临床医生的经历,并关闭重要的更广泛的对话。至关重要的是,围绕SCD细胞疗法的讨论是在历史不平等、健康不平等和不信任的背景下进行的。黑人社区——受到性别歧视的影响尤为严重——在研究和医疗保健方面面临着系统性的种族主义、忽视和怀疑。17,18这种情况决定了如何解释和接受治疗语言,这就需要谨慎和透明的沟通。解决这一问题对于任何关于治疗语言的讨论都是至关重要的,这不仅是为了确保公平获得新兴疗法,也是为了重建研究人员、临床医生和社区之间的信任和问责关系。如果不认识到并积极解决这些系统性的不公正,治愈治疗的承诺就有可能再现历史上塑造了SCD患者经历的同样的不平等。最后,由于缺乏深思熟虑的实施,治疗的语言可能会混淆——甚至加剧——许多患有SCD的人所面临的深层次的未满足需求。由于年龄、合并症、费用或卫生保健系统基础设施和专业知识,绝大多数SCD患者仍然无法获得这些治疗。在低收入和中等收入国家尤其如此,因为大多数SCD患者居住在这些国家。随着细胞疗法的发展,对可获得的治疗、管理和遗传咨询的投资仍然至关重要。总之,尽管“治愈”这个词面临挑战,但没有简单、完美的替代方案。“变革疗法”或“激进疗法”这些术语可能不太容易引起不切实际的期望。但也许最重要的目标是让人们看到SCD患者生活的复杂性、韧性和尊严,无论患者是否接受细胞治疗。考虑这些治疗的个人需要透明和仔细沟通的信息来做出明智的选择。卫生系统需要考虑如何从整体上支持接受细胞疗法的患者,从决策到生存。除了术语之外,重要的是要为诚实、细致的对话创造空间,讨论动机、期望、需求和治疗后的生活。Lydia O. Okoibhole:概念化;写作——审阅和编辑;原创作品。冯米·达索鲁:写作、审稿、编辑;原创作品。Zainab Garba-Sani:写作、评论和编辑;原创作品。Stephen P. Hibbs:概念化;原创作品草案;写作-审查和编辑。作者报告没有利益冲突。由惠康信托基金支持(授权号220540/Z/20/A给惠康桑格研究所和惠康连接科学)。F.D.由NIHR博士前临床学术奖学金支持,奖励号NIHR304921。S.P.H.由惠康信托基金资助的HARP博士研究奖学金(资助号223500/Z/21/Z)支持。数据共享不适用于
{"title":"Cellular therapies for sickle cell disease: Should we be calling them a cure?","authors":"Lydia O. Okoibhole, Funmi Dasaolu, Zainab Garba-Sani, Stephen P. Hibbs","doi":"10.1002/hem3.70283","DOIUrl":"https://doi.org/10.1002/hem3.70283","url":null,"abstract":"<p>Recent advances in gene therapy and expanded access for haematopoietic stem cell transplantation (HSCT) have made conversations about a <i>cure</i> more pertinent for individuals living with sickle cell disease (SCD). These advances, including the approval of exa-cel gene therapy in the United Kingdom,<span><sup>1</sup></span> have amplified the language of cure within policy, media, and clinical discussions. However, as this language becomes more visible, questions emerge about what <i>cure</i> means for individuals living with SCD, for healthcare systems, and for communities historically underserved and marginalized within research and clinical care.</p><p>The term <i>cure</i>, derived from the Latin “curare,” meaning “to care for” or “to heal,” suggests the complete elimination of symptoms caused by disease and a restoration to a previous state of health,<span><sup>2</sup></span> but in the context of a lifelong disease like SCD, the meaning is more complex.</p><p>In this article, we begin with the personal reflections of one author (F.D.) living with SCD as she grapples with the possibility and uncertainty of cellular therapies (Box 1). Next, we survey how the language of cure is used in public discussions about SCD cellular therapies. We share the account of one author (Z.G.-S.) whose experience befits the word <i>cure</i>, before exploring complexities that challenge the use of curative language. Finally, we highlight important considerations for health professionals and health systems relating to cellular therapies for SCD.</p><p>The last decade has seen a significant increase in discussion about curative therapies in SCD. In 2023, when the US Food and Drug Administration (FDA) approved the gene editing therapy exa-cel, it was described as “the first CRISPR cure for a genetic disease.”<span><sup>3, 4</sup></span> Similarly, in 2025, the UK National Institute for Health and Care Excellence (NICE) described exa-cel's approval as a “potential cure for some people with severe sickle cell disease.”<span><sup>5</sup></span> This approval followed a long and complex journey of regulatory review and appeal,<span><sup>6</sup></span> which finally resulted in the National Health Service (NHS) funding this one-time therapy,<span><sup>5</sup></span> described by NHS England as a source of hope.<span><sup>1</sup></span></p><p>Additionally, conversation continues to build around allogeneic HSCT in SCD, as developments in conditioning regimens increase accessibility for a much wider group of people, including older recipients, those without a fully matched donor and those with additional comorbidities.<span><sup>7</sup></span> In a patient guide addressing SCD and HSCT, UK charities Anthony Nolan and the Sickle Cell Society described the treatment as a cure, while also defining the benefits and risks associated.<span><sup>8</sup></span> However, medical regulators, healthcare professionals, and media reports rarely explicitly define cure, and the pote","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70283","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695298","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ivan Sergeev, Josef T. Prchal, Seyed Mehdi Nouraie, Binal N. Shah, Xu Zhang, Adelina Sergueeva, Galina Miasnikova, Tomas Ganz, Victor R. Gordeuk
<p>Hepcidin, the master regulator of iron metabolism, is a hepatic peptide hormone that inhibits the absorption of iron by enterocytes and the release of iron from macrophages through interaction with ferroportin in the cellular membrane.<span><sup>1</sup></span> Hepcidin binds to ferroportin, occluding it and causing it to move to the interior of cells, thereby preventing the release of cellular iron to plasma, and also causing a conformational change that leads to ferroportin ubiquitination and lysosomal degradation.<span><sup>2</sup></span> Hepcidin is upregulated in response to (1) higher intracellular iron stores and plasma iron concentrations as reflected by serum ferritin and elevated circulating transferrin-bound iron<span><sup>3</sup></span> via a bone morphogenetic protein receptor complex<span><sup>4</sup></span> and (2) inflammation through interleukin-6 and a JAK-STAT pathway.<span><sup>5</sup></span> Hepcidin is downregulated by increased erythropoiesis via erythroferrone secreted by erythroblasts, which suppresses bone morphogenetic protein receptor complex signaling.<span><sup>6</sup></span> Hypoxia downregulates hepcidin mostly indirectly, via erythropoietin and erythroferrone.<span><sup>7</sup></span> Upregulation of hypoxia sensing as seen in Chuvash erythrocytosis also leads to increased erythropoietin and erythroferrone and decreased hepcidin.<span><sup>8-10</sup></span> The relationship of hepcidin to transferrin concentration in a model that includes ferritin, erythropoietin, and upregulated hypoxia sensing has not previously been reported.</p><p>Hypoxia-inducible factors (HIFs) are dimers of a common HIF-β subunit and one of several HIF-α subunits that are regulated posttranslationally; HIF-1 and HIF-2 are best studied.<span><sup>11</sup></span> Prolyl hydroxylases (PHDs, Fe-dependent enzymes) are the principal negative regulators of HIF-α subunits. In the presence of O<sub>2</sub> and α-ketoglutarate, HIF-1α and HIF-2α subunits are hydroxylated by PHDs, facilitating binding to von Hippel–Lindau (VHL) protein, which leads to their ubiquitination and rapid proteosomal degradation.<span><sup>12, 13</sup></span> In hypoxia and iron deficiency, degradation of HIF-α decreases, leading to increased HIF-1 and HIF-2 heterodimers that augment transcription of hypoxia-inducible genes.<span><sup>11</sup></span></p><p>Homozygous germline loss-of-function <i>VHL</i><sup>R200W</sup> causes congenital Chuvash erythrocytosis with thrombosis as the major cause of morbidity and mortality. <i>VHL</i><sup>R200W</sup> homozygosity leads to decreased ubiquitination of HIF-1α and HIF-2α,<span><sup>8</sup></span> which is necessary for their degradation. In turn, HIFs upregulate several genes that influence oxygen homeostasis, erythropoiesis, and iron metabolism.<span><sup>8, 14, 15</sup></span> Erythropoietin, upregulated by hypoxia and iron deficiency via HIF-2,<span><sup>16</sup></span> is increased in <i>VHL</i><sup>R200W</sup> homozygotes ev
{"title":"VHL, transferrin, and erythropoietin in the regulation of hepcidin","authors":"Ivan Sergeev, Josef T. Prchal, Seyed Mehdi Nouraie, Binal N. Shah, Xu Zhang, Adelina Sergueeva, Galina Miasnikova, Tomas Ganz, Victor R. Gordeuk","doi":"10.1002/hem3.70271","DOIUrl":"https://doi.org/10.1002/hem3.70271","url":null,"abstract":"<p>Hepcidin, the master regulator of iron metabolism, is a hepatic peptide hormone that inhibits the absorption of iron by enterocytes and the release of iron from macrophages through interaction with ferroportin in the cellular membrane.<span><sup>1</sup></span> Hepcidin binds to ferroportin, occluding it and causing it to move to the interior of cells, thereby preventing the release of cellular iron to plasma, and also causing a conformational change that leads to ferroportin ubiquitination and lysosomal degradation.<span><sup>2</sup></span> Hepcidin is upregulated in response to (1) higher intracellular iron stores and plasma iron concentrations as reflected by serum ferritin and elevated circulating transferrin-bound iron<span><sup>3</sup></span> via a bone morphogenetic protein receptor complex<span><sup>4</sup></span> and (2) inflammation through interleukin-6 and a JAK-STAT pathway.<span><sup>5</sup></span> Hepcidin is downregulated by increased erythropoiesis via erythroferrone secreted by erythroblasts, which suppresses bone morphogenetic protein receptor complex signaling.<span><sup>6</sup></span> Hypoxia downregulates hepcidin mostly indirectly, via erythropoietin and erythroferrone.<span><sup>7</sup></span> Upregulation of hypoxia sensing as seen in Chuvash erythrocytosis also leads to increased erythropoietin and erythroferrone and decreased hepcidin.<span><sup>8-10</sup></span> The relationship of hepcidin to transferrin concentration in a model that includes ferritin, erythropoietin, and upregulated hypoxia sensing has not previously been reported.</p><p>Hypoxia-inducible factors (HIFs) are dimers of a common HIF-β subunit and one of several HIF-α subunits that are regulated posttranslationally; HIF-1 and HIF-2 are best studied.<span><sup>11</sup></span> Prolyl hydroxylases (PHDs, Fe-dependent enzymes) are the principal negative regulators of HIF-α subunits. In the presence of O<sub>2</sub> and α-ketoglutarate, HIF-1α and HIF-2α subunits are hydroxylated by PHDs, facilitating binding to von Hippel–Lindau (VHL) protein, which leads to their ubiquitination and rapid proteosomal degradation.<span><sup>12, 13</sup></span> In hypoxia and iron deficiency, degradation of HIF-α decreases, leading to increased HIF-1 and HIF-2 heterodimers that augment transcription of hypoxia-inducible genes.<span><sup>11</sup></span></p><p>Homozygous germline loss-of-function <i>VHL</i><sup>R200W</sup> causes congenital Chuvash erythrocytosis with thrombosis as the major cause of morbidity and mortality. <i>VHL</i><sup>R200W</sup> homozygosity leads to decreased ubiquitination of HIF-1α and HIF-2α,<span><sup>8</sup></span> which is necessary for their degradation. In turn, HIFs upregulate several genes that influence oxygen homeostasis, erythropoiesis, and iron metabolism.<span><sup>8, 14, 15</sup></span> Erythropoietin, upregulated by hypoxia and iron deficiency via HIF-2,<span><sup>16</sup></span> is increased in <i>VHL</i><sup>R200W</sup> homozygotes ev","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 12","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70271","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}