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}
Efrat Luttwak, David Nemirovsky, Sean M. Devlin, Alexander P. Boardman, Giulio Cassanello, Magdalena Corona, Danny Luan, Parastoo B. Dahi, Alejandro Luna De Abia, Lorenzo Falchi, Ivan Landego, Richard J. Lin, Jeniffer K. Lue, Lia M. Palomba, Jae H. Park, Sandeep S. Raj, Kai Rejeski, Alfredo Rivas Delgado, Michael Scordo, Ana Alarcon Tomas, Gunjan L. Shah, Miguel-Angel Perales, Gilles Salles, Roni Shouval
Corticosteroids are commonly used to manage cytokine release syndrome (CRS) and immune effector cell–associated neurotoxicity syndrome (ICANS) following chimeric antigen receptor (CAR) T-cell therapy, and yet, their dose-specific impact on outcomes remains uncertain. We retrospectively evaluated 276 adults with large B-cell lymphoma (LBCL) treated with CD19-directed CAR-T therapy (axi-cel, tisa-cel, or liso-cel) between 2016 and 2023 at a single institution. Cumulative corticosteroid dose was defined as the total dose administered within 21 days of infusion. Corticosteroids were administered to 105 patients (38%), initiated at a median of 5 days post-infusion (interquartile range [IQR] 3–7) for CRS (38%), ICANS (15%), or both (47%). Use was more frequent with axi-cel (P < 0.001), although the cumulative dose and duration were similar across products. To assess the impact of corticosteroid exposure, we carried out a 21-day landmark analysis. Corticosteroid exposure, modeled as a time-dependent covariate, was not significantly associated with infection risk, non-relapse mortality, or inferior overall survival (OS) or progression-free survival (PFS). However, in a landmark analysis, patients receiving above-median cumulative corticosteroid doses had a significantly higher risk of infection compared to those receiving below-median corticosteroid doses or no corticosteroids (P = 0.042). In a landmark multivariable analysis, corticosteroid cumulative dose was associated with increased late hematologic toxicity (adjusted hazard ratio [HR] 1.02, 95% CI 1.02–1.03). Finally, in a sensitivity analysis excluding patients with Grade ≥4 CRS/ICANS, corticosteroid cumulative dose remained unassociated with OS or relapse, but was linked to shorter PFS (adjusted HR 1.04, 95% CI 1.01–1.06). These findings support the safe yet judicious use of corticosteroids to manage CAR-T toxicities in LBCL.
皮质类固醇通常用于治疗嵌合抗原受体(CAR) t细胞治疗后的细胞因子释放综合征(CRS)和免疫效应细胞相关神经毒性综合征(ICANS),然而,它们对结果的剂量特异性影响仍不确定。我们回顾性评估了2016年至2023年间在一家机构接受cd19定向CAR-T疗法(轴细胞、组织细胞或liso-细胞)治疗的276名成年大b细胞淋巴瘤(LBCL)患者。累积皮质类固醇剂量定义为输注后21天内给药的总剂量。105例患者(38%)在输注后5天(四分位数范围[IQR] 3-7)开始使用皮质类固醇,用于CRS (38%), ICANS(15%)或两者(47%)。使用axis -cel的频率更高(P < 0.001),尽管不同产品的累积剂量和持续时间相似。为了评估皮质类固醇暴露的影响,我们进行了为期21天的里程碑式分析。皮质类固醇暴露,建模为一个时间依赖的协变量,与感染风险、非复发死亡率、较差的总生存期(OS)或无进展生存期(PFS)没有显著相关。然而,在一项具有里程碑意义的分析中,接受高于中位数累积皮质类固醇剂量的患者与接受低于中位数皮质类固醇剂量或未接受皮质类固醇剂量的患者相比,感染风险明显更高(P = 0.042)。在一项具有里程碑意义的多变量分析中,皮质类固醇累积剂量与晚期血液学毒性增加相关(校正风险比[HR] 1.02, 95% CI 1.02 - 1.03)。最后,在排除≥4级CRS/ICANS患者的敏感性分析中,皮质类固醇累积剂量与OS或复发无关,但与较短的PFS相关(调整后HR 1.04, 95% CI 1.01-1.06)。这些发现支持安全而明智地使用皮质类固醇来控制LBCL的CAR-T毒性。
{"title":"Patterns and safety of glucocorticosteroid use following CD19 CAR-T therapy for large B-cell lymphoma","authors":"Efrat Luttwak, David Nemirovsky, Sean M. Devlin, Alexander P. Boardman, Giulio Cassanello, Magdalena Corona, Danny Luan, Parastoo B. Dahi, Alejandro Luna De Abia, Lorenzo Falchi, Ivan Landego, Richard J. Lin, Jeniffer K. Lue, Lia M. Palomba, Jae H. Park, Sandeep S. Raj, Kai Rejeski, Alfredo Rivas Delgado, Michael Scordo, Ana Alarcon Tomas, Gunjan L. Shah, Miguel-Angel Perales, Gilles Salles, Roni Shouval","doi":"10.1002/hem3.70248","DOIUrl":"https://doi.org/10.1002/hem3.70248","url":null,"abstract":"<p>Corticosteroids are commonly used to manage cytokine release syndrome (CRS) and immune effector cell–associated neurotoxicity syndrome (ICANS) following chimeric antigen receptor (CAR) T-cell therapy, and yet, their dose-specific impact on outcomes remains uncertain. We retrospectively evaluated 276 adults with large B-cell lymphoma (LBCL) treated with CD19-directed CAR-T therapy (axi-cel, tisa-cel, or liso-cel) between 2016 and 2023 at a single institution. Cumulative corticosteroid dose was defined as the total dose administered within 21 days of infusion. Corticosteroids were administered to 105 patients (38%), initiated at a median of 5 days post-infusion (interquartile range [IQR] 3–7) for CRS (38%), ICANS (15%), or both (47%). Use was more frequent with axi-cel (P < 0.001), although the cumulative dose and duration were similar across products. To assess the impact of corticosteroid exposure, we carried out a 21-day landmark analysis. Corticosteroid exposure, modeled as a time-dependent covariate, was not significantly associated with infection risk, non-relapse mortality, or inferior overall survival (OS) or progression-free survival (PFS). However, in a landmark analysis, patients receiving above-median cumulative corticosteroid doses had a significantly higher risk of infection compared to those receiving below-median corticosteroid doses or no corticosteroids (P = 0.042). In a landmark multivariable analysis, corticosteroid cumulative dose was associated with increased late hematologic toxicity (adjusted hazard ratio [HR] 1.02, 95% CI 1.02–1.03). Finally, in a sensitivity analysis excluding patients with Grade ≥4 CRS/ICANS, corticosteroid cumulative dose remained unassociated with OS or relapse, but was linked to shorter PFS (adjusted HR 1.04, 95% CI 1.01–1.06). These findings support the safe yet judicious use of corticosteroids to manage CAR-T toxicities in LBCL.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70248","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626313","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}
Haley Newman, Derek Wong, Jinhua Wu, Jeffrey Schubert, Netta Golenberg, Natali Naveh, Maha Patel, Feng Xu, Sarah Charles, Jiani Chen, Elizabeth H. Denenberg, Elizabeth A. Fanning, Daniel Gallo, Tammy Luke, Morgan Thomas, Kajia Cao, Ada J. S. Chan, Munashe Holloman, Zhiqian Fan, Weixuan Fu, Stephen P. Hunger, Suzanne P. MacFarland, Kathrin M. Bernt, Lea F. Surrey, Minjie Luo, Gerald B. Wertheim, Marilyn M. Li, Sarah K. Tasian, Yiming Zhong
Advancements in the rapidity, cost efficacy, and sensitivity of next-generation sequencing (NGS) have facilitated molecular risk stratification and precision medicine-based treatment for pediatric leukemia. The benefit of uniform cytomolecular analyses for clinical trial risk assignment is clear. However, the clinical impact of comprehensive NGS for pediatric leukemias at an institutional level is not well described. We report the genomic spectrum of one of the largest cohorts of pediatric and adolescent/young adult (AYA) acute leukemias examined to date (n = 1442) via institutional NGS testing from our large tertiary care center. We evaluated the clinical utility of genomic results for informing prognosis and treatment. We identified high utility of the comprehensive DNA-based mutational panel and RNA-fusion panel, which detected leukemia-associated variants in 99% of specimens. We observed 65% of B-cell acute lymphoblastic leukemia cases and 69% of patients with acute myeloid leukemia harbored a prognostic molecular alteration. In total, 325 of 1134 patients (29%) harbored potentially targetable molecular biomarkers, and 23% of cases with follow-up data received precision medicine-based therapy. Paired diagnostic and relapsed leukemia samples aided in differentiation between treatment-related leukemia versus lineage drift/switch. These findings demonstrate the broad utility of comprehensive molecular sequencing for pediatric/AYA leukemia at an institutional level to improve outcomes.
{"title":"Leveraging genomic diagnostics for prognostics and therapeutics in pediatric acute leukemia","authors":"Haley Newman, Derek Wong, Jinhua Wu, Jeffrey Schubert, Netta Golenberg, Natali Naveh, Maha Patel, Feng Xu, Sarah Charles, Jiani Chen, Elizabeth H. Denenberg, Elizabeth A. Fanning, Daniel Gallo, Tammy Luke, Morgan Thomas, Kajia Cao, Ada J. S. Chan, Munashe Holloman, Zhiqian Fan, Weixuan Fu, Stephen P. Hunger, Suzanne P. MacFarland, Kathrin M. Bernt, Lea F. Surrey, Minjie Luo, Gerald B. Wertheim, Marilyn M. Li, Sarah K. Tasian, Yiming Zhong","doi":"10.1002/hem3.70269","DOIUrl":"https://doi.org/10.1002/hem3.70269","url":null,"abstract":"<p>Advancements in the rapidity, cost efficacy, and sensitivity of next-generation sequencing (NGS) have facilitated molecular risk stratification and precision medicine-based treatment for pediatric leukemia. The benefit of uniform cytomolecular analyses for clinical trial risk assignment is clear. However, the clinical impact of comprehensive NGS for pediatric leukemias at an institutional level is not well described. We report the genomic spectrum of one of the largest cohorts of pediatric and adolescent/young adult (AYA) acute leukemias examined to date (<i>n</i> = 1442) via institutional NGS testing from our large tertiary care center. We evaluated the clinical utility of genomic results for informing prognosis and treatment. We identified high utility of the comprehensive DNA-based mutational panel and RNA-fusion panel, which detected leukemia-associated variants in 99% of specimens. We observed 65% of B-cell acute lymphoblastic leukemia cases and 69% of patients with acute myeloid leukemia harbored a prognostic molecular alteration. In total, 325 of 1134 patients (29%) harbored potentially targetable molecular biomarkers, and 23% of cases with follow-up data received precision medicine-based therapy. Paired diagnostic and relapsed leukemia samples aided in differentiation between treatment-related leukemia versus lineage drift/switch. These findings demonstrate the broad utility of comprehensive molecular sequencing for pediatric/AYA leukemia at an institutional level to improve outcomes.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70269","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626496","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}
Romain Stammler, Peter Chen, Orianne Debeaupuis, Lin-Pierre Zhao, Mirabelle Ruyer-Thompson, Eve Zakine, Julien Rossignol, Lauriane Goldwirt, Thibault Comont, Maël Heiblig, Lionel Adès, Marie Sebert, Jean-Sébastien Allain, Julien Campagne, Marie-Anne Couturier, Marina Cumin, Marie-Caroline Dalmas, Guillaume Denis, Cécile Devloo, Adrien De Voeght, Louis Drevon, Pierre Duffau, Sophie Georgin-Lavialle, Delphine Gobert, Olivier Kosmider, Frédéric Rieux-Laucat, Noémie Abisror, Estibaliz Lazaro, Jean-Guillaume Lopez, Alexandre Maria, Wladimir Mauhin, Julie Merindol, Claire Merlot, Tristan Mirault, Laurent Pascal, François Perrin, Emmanuel Raffoux, Ramy Rahme, Damien Roos-Weil, Benjamin Terrier, Benjamin Thoreau, Olivier Fain, Pierre Fenaux, Pierre Hirsch, Vincent Jachiet, Jérôme Hadjadj, Arsène Mékinian, MINHEMON
<p>Myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) are heterogeneous clonal disorders that may be associated with immune-mediated inflammatory disorders (IMIDs), reported in up to 25% of patients.<span><sup>1-4</sup></span> The contribution of clonal hematopoiesis to systemic inflammation is increasingly recognized, exemplified by the recently described VEXAS syndrome.<span><sup>5</sup></span> Among recurrent genetic alterations in these myeloid neoplasms (MNs), isocitrate dehydrogenase (<i>IDH</i>1/2) mutations are present in 2%–12% of MDS and 4%–6% of CMML.<span><sup>6, 7</sup></span> Beyond their leukemogenic role through 2-hydroxyglutarate–mediated epigenetic dysregulation, <i>IDH</i> mutations are enriched in patients with IMIDs (14%–20%), suggesting a contribution to immune dysregulation.<span><sup>8, 9</sup></span> The clinical impact of IDH inhibition on inflammatory manifestations has never been addressed. In this retrospective multicenter study, we characterized <i>IDH</i>-mutated MN with IMIDs and highlighted the striking efficacy of IDH inhibitors on systemic inflammation.</p><p>We analyzed 50 patients with <i>IDH</i>-mutated MN and IMIDs (<i>IDH</i><sup>mut</sup>) and compared them to 61 patients with MN and IMIDs without <i>IDH</i> mutations (<i>IDH</i><sup>wt</sup> group). Clinical, biological, and immunological features, survival, and therapeutic responses were assessed. IMIDs' diagnoses were reviewed by three specialists and defined according to international criteria. Inflammatory responses were classified as complete (iCR) or partial (iPR), based on clinical signs, C-reactive protein (CRP) levels, and corticosteroid dose; overall response (iOR) referred to either iCR or iPR. Methodological details, including definitions of inflammatory manifestations, response criteria, and laboratory methods, are provided in the Supporting Information.</p><p>The <i>IDH</i><sup>mut</sup> group (50% male, median age 73 years [54–85]) included 36 (72%) patients with MDS, mostly with low blasts (69%) or increased blasts (25%), and 14 (28%) with CMML, mostly CMML-1 (93%) (Table 1). Abnormal cytogenetics were present in 10 (20%) patients, most frequently trisomy 8 (<i>n</i> = 4) and complex karyotype (<i>n</i> = 2). <i>IDH1</i> mutations were detected in 54%, <i>IDH2</i> in 44%, and both in 2%. In MDS, 78% were classified as low-risk according to the molecular international prognostic scoring system (IPSS-M) (≤0), while in CMML, 71% were classified as low-risk by chronic myelomonocytic leukemia-specific molecular prognostic scoring system (CPSS-Mol) (≤1). Inflammatory manifestations were dominated by musculoskeletal involvement (78%), mainly polyarthritis, followed by cutaneous manifestations (44%), mostly neutrophilic dermatosis, with 30% of patients presenting both. The most frequent IMIDs diagnoses were seronegative arthritis (34%), polymyalgia rheumatica (PMR, 26%), and giant cell arteritis (GCA, 16%). No significant di
{"title":"Inflammatory disorders in IDH-mutated myeloid neoplasms: Characteristics and response to IDH inhibitors","authors":"Romain Stammler, Peter Chen, Orianne Debeaupuis, Lin-Pierre Zhao, Mirabelle Ruyer-Thompson, Eve Zakine, Julien Rossignol, Lauriane Goldwirt, Thibault Comont, Maël Heiblig, Lionel Adès, Marie Sebert, Jean-Sébastien Allain, Julien Campagne, Marie-Anne Couturier, Marina Cumin, Marie-Caroline Dalmas, Guillaume Denis, Cécile Devloo, Adrien De Voeght, Louis Drevon, Pierre Duffau, Sophie Georgin-Lavialle, Delphine Gobert, Olivier Kosmider, Frédéric Rieux-Laucat, Noémie Abisror, Estibaliz Lazaro, Jean-Guillaume Lopez, Alexandre Maria, Wladimir Mauhin, Julie Merindol, Claire Merlot, Tristan Mirault, Laurent Pascal, François Perrin, Emmanuel Raffoux, Ramy Rahme, Damien Roos-Weil, Benjamin Terrier, Benjamin Thoreau, Olivier Fain, Pierre Fenaux, Pierre Hirsch, Vincent Jachiet, Jérôme Hadjadj, Arsène Mékinian, MINHEMON","doi":"10.1002/hem3.70256","DOIUrl":"https://doi.org/10.1002/hem3.70256","url":null,"abstract":"<p>Myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) are heterogeneous clonal disorders that may be associated with immune-mediated inflammatory disorders (IMIDs), reported in up to 25% of patients.<span><sup>1-4</sup></span> The contribution of clonal hematopoiesis to systemic inflammation is increasingly recognized, exemplified by the recently described VEXAS syndrome.<span><sup>5</sup></span> Among recurrent genetic alterations in these myeloid neoplasms (MNs), isocitrate dehydrogenase (<i>IDH</i>1/2) mutations are present in 2%–12% of MDS and 4%–6% of CMML.<span><sup>6, 7</sup></span> Beyond their leukemogenic role through 2-hydroxyglutarate–mediated epigenetic dysregulation, <i>IDH</i> mutations are enriched in patients with IMIDs (14%–20%), suggesting a contribution to immune dysregulation.<span><sup>8, 9</sup></span> The clinical impact of IDH inhibition on inflammatory manifestations has never been addressed. In this retrospective multicenter study, we characterized <i>IDH</i>-mutated MN with IMIDs and highlighted the striking efficacy of IDH inhibitors on systemic inflammation.</p><p>We analyzed 50 patients with <i>IDH</i>-mutated MN and IMIDs (<i>IDH</i><sup>mut</sup>) and compared them to 61 patients with MN and IMIDs without <i>IDH</i> mutations (<i>IDH</i><sup>wt</sup> group). Clinical, biological, and immunological features, survival, and therapeutic responses were assessed. IMIDs' diagnoses were reviewed by three specialists and defined according to international criteria. Inflammatory responses were classified as complete (iCR) or partial (iPR), based on clinical signs, C-reactive protein (CRP) levels, and corticosteroid dose; overall response (iOR) referred to either iCR or iPR. Methodological details, including definitions of inflammatory manifestations, response criteria, and laboratory methods, are provided in the Supporting Information.</p><p>The <i>IDH</i><sup>mut</sup> group (50% male, median age 73 years [54–85]) included 36 (72%) patients with MDS, mostly with low blasts (69%) or increased blasts (25%), and 14 (28%) with CMML, mostly CMML-1 (93%) (Table 1). Abnormal cytogenetics were present in 10 (20%) patients, most frequently trisomy 8 (<i>n</i> = 4) and complex karyotype (<i>n</i> = 2). <i>IDH1</i> mutations were detected in 54%, <i>IDH2</i> in 44%, and both in 2%. In MDS, 78% were classified as low-risk according to the molecular international prognostic scoring system (IPSS-M) (≤0), while in CMML, 71% were classified as low-risk by chronic myelomonocytic leukemia-specific molecular prognostic scoring system (CPSS-Mol) (≤1). Inflammatory manifestations were dominated by musculoskeletal involvement (78%), mainly polyarthritis, followed by cutaneous manifestations (44%), mostly neutrophilic dermatosis, with 30% of patients presenting both. The most frequent IMIDs diagnoses were seronegative arthritis (34%), polymyalgia rheumatica (PMR, 26%), and giant cell arteritis (GCA, 16%). No significant di","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70256","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626490","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}
Niels W. C. J. van de Donk, Philippe Moreau, Jesus F. San-Miguel, Maria-Victoria Mateos, Meletios A. Dimopoulos, Sonja Zweegman, Francesca Gay, Monika Engelhardt, Roberto Mina, Elena Zamagni, Michel Delforge, Meral Beksac, Andrew Spencer, Fredrik Schjesvold, Christoph Driessen, Martin Kaiser, Aurore Perrot, Ralph Wäsch, Charlotte L. B. M. Korst, Annemiek Broijl, Cyrille Touzeau, Salomon Manier, Roman Hajek, Jelena Bila, Guldane C. Seval, Michael O'Dwyer, Heinz Ludwig, Carlos Fernandez de Larrea, Rakesh Popat, Pellegrino Musto, Paula Rodriguez-Otero, Kwee Yong, Marin Kortüm, Leo Rasche, Evangelos Terpos, Marc S. Raab, Mario Boccadoro, Pieter Sonneveld, Hermann Einsele, the EMN Guidelines Committee
The treatment landscape of heavily pretreated relapsed/refractory MM has changed considerably in recent years with the introduction of novel BCMA- and GPRC5D-directed immunotherapies, including CAR T-cell therapy, bispecific antibodies (BsAbs), and antibody-drug conjugates (ADCs). Treatment selection and sequencing become increasingly complex with the broad range of therapeutic options. In this review, the European Myeloma Network provides recommendations on how to best incorporate these novel therapies into the present treatment landscape using current evidence. The optimal treatment sequence depends on various patient- and tumor-related features, but also reimbursement and availability issues. In addition, mechanisms underlying relapse (e.g., antigen loss, reduced T-cell fitness, or outgrowth of T-cell resistant clones) dictate the efficacy of sequential BCMA- or GPRC5D-directed immunotherapy. BCMA-targeting BsAbs and ADCs should preferably be avoided prior to CAR T-cell therapy, as some studies have shown that these agents negatively influence clinical outcomes after CAR T-cell therapy. Therefore, we recommend the selection of CAR T-cell therapy first, and BsAbs and/or belamaf later in the disease course, if patients are eligible for CAR T-cell therapy and in case CAR T-cell therapy is available within a short time frame. However, bridging therapy with GPRC5D-directed BsAbs (initiation after apheresis) can be considered to significantly reduce tumor burden, because this was shown to improve the efficacy of consecutive BCMA-directed CAR T-cell therapy. Sequential treatment with agents targeting the same antigen, but with different modes of action, is feasible, but several studies have demonstrated that target switch is a more effective strategy. In addition, there is increasing evidence indicating that the efficacy of sequential use of BsAbs can be improved by creating a BsAb-free interval.
{"title":"Sequencing BCMA- and GPRC5D-targeting immunotherapies in multiple myeloma: Practical guidance from the European Myeloma Network","authors":"Niels W. C. J. van de Donk, Philippe Moreau, Jesus F. San-Miguel, Maria-Victoria Mateos, Meletios A. Dimopoulos, Sonja Zweegman, Francesca Gay, Monika Engelhardt, Roberto Mina, Elena Zamagni, Michel Delforge, Meral Beksac, Andrew Spencer, Fredrik Schjesvold, Christoph Driessen, Martin Kaiser, Aurore Perrot, Ralph Wäsch, Charlotte L. B. M. Korst, Annemiek Broijl, Cyrille Touzeau, Salomon Manier, Roman Hajek, Jelena Bila, Guldane C. Seval, Michael O'Dwyer, Heinz Ludwig, Carlos Fernandez de Larrea, Rakesh Popat, Pellegrino Musto, Paula Rodriguez-Otero, Kwee Yong, Marin Kortüm, Leo Rasche, Evangelos Terpos, Marc S. Raab, Mario Boccadoro, Pieter Sonneveld, Hermann Einsele, the EMN Guidelines Committee","doi":"10.1002/hem3.70260","DOIUrl":"https://doi.org/10.1002/hem3.70260","url":null,"abstract":"<p>The treatment landscape of heavily pretreated relapsed/refractory MM has changed considerably in recent years with the introduction of novel BCMA- and GPRC5D-directed immunotherapies, including CAR T-cell therapy, bispecific antibodies (BsAbs), and antibody-drug conjugates (ADCs). Treatment selection and sequencing become increasingly complex with the broad range of therapeutic options. In this review, the European Myeloma Network provides recommendations on how to best incorporate these novel therapies into the present treatment landscape using current evidence. The optimal treatment sequence depends on various patient- and tumor-related features, but also reimbursement and availability issues. In addition, mechanisms underlying relapse (e.g., antigen loss, reduced T-cell fitness, or outgrowth of T-cell resistant clones) dictate the efficacy of sequential BCMA- or GPRC5D-directed immunotherapy. BCMA-targeting BsAbs and ADCs should preferably be avoided prior to CAR T-cell therapy, as some studies have shown that these agents negatively influence clinical outcomes after CAR T-cell therapy. Therefore, we recommend the selection of CAR T-cell therapy first, and BsAbs and/or belamaf later in the disease course, if patients are eligible for CAR T-cell therapy and in case CAR T-cell therapy is available within a short time frame. However, bridging therapy with GPRC5D-directed BsAbs (initiation after apheresis) can be considered to significantly reduce tumor burden, because this was shown to improve the efficacy of consecutive BCMA-directed CAR T-cell therapy. Sequential treatment with agents targeting the same antigen, but with different modes of action, is feasible, but several studies have demonstrated that target switch is a more effective strategy. In addition, there is increasing evidence indicating that the efficacy of sequential use of BsAbs can be improved by creating a BsAb-free interval.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70260","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626489","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}
<p>Chromosomal translocations involving the mixed lineage leukemia (also known as <i>MLL</i> or <i>KMT2A</i>) gene occur in approximately 40% of children with acute myeloid leukemia (AML) under the age of 3. The fusion partners of <i>MLL</i> can vary widely with over 130 different ones identified to date. These fusions result in constitutive activation of MLL and ultimately drive the overexpression of key downstream target genes including <i>MEIS1</i> and <i>HOXA</i> cluster genes that leads to leukemia development. Critically, <i>MLL</i>-rearranged (<i>MLL</i>-r) leukemias all depend on menin for growth and survival that in turn has driven the development of menin inhibitors (e.g., revumenib) currently being evaluated in numerous clinical trials. I refer the reader to two excellent recent reviews on the biology of <i>MLL</i>-r AML and early clinical trial data.<span><sup>1, 2</sup></span></p><p>Another common chromosomal translocation in AML is the one involving the <i>NUP98</i> gene, which has up to 30 distinct fusion partners. These have been shown to drive transcription condensates in the nucleus as well as changes in chromatin architecture that are thought to play an important role in rewiring cells toward leukemia development.<span><sup>3</sup></span> Importantly, NUP98 fusion proteins interact with wild-type MLL (KMT2A) complexes, and this also drives high expression of <i>MEIS1</i> and <i>HOXA</i> cluster genes. Notably, <i>NUP98</i>-rearranged (<i>NUP98</i>-r) AML cases are also very sensitive to menin inhibitors.<span><sup>4</sup></span></p><p>Nevertheless, just like other targeted therapies, resistance mechanisms have now been identified in AML cases treated with menin inhibitors. These include mutations in the drug pocket of menin around amino acid residues M327, G331, T349, and S160 that prevent the binding of menin inhibitors. Alternative mechanisms include epigenetic wiring such as loss of components of the PRC1.1 complex.<span><sup>2</sup></span> Interestingly, a new study showed that chemotherapy could also render <i>MLL</i>-r leukemia more resistant to menin inhibition without prior exposure to menin inhibitors.<span><sup>5</sup></span> Therefore, combination strategies are needed to increase the chances of complete remission and cure.</p><p>Two new studies<span><sup>6, 7</sup></span> have now been published back-to-back in <i>Cancer Discovery</i> addressing this very issue, with both papers looking at the efficacy of combining menin inhibitors with the newly developed orally bioavailable KAT6A/B selective inhibitor PF-9363. Both KAT6A and KAT6B are members of the MYST (MOZ/KAT6A, Ybf2/Sas3, Sas2, and Tip60) family of histone acetyltransferases (HATs) that also includes KAT5, KAT7, and KAT8. These all differentially regulate the acetylation of lysine residues on histone 3, with KAT6A and KAT6B driving H3K23Ac whilst KAT7 is responsible for H3K14Ac for example. Whilst this family is important in regulating chromatin structure and
{"title":"A new KAT on the block rewrites the epigenetic script in menin inhibitor-resistant leukemia","authors":"Yizhou Huang, Charles E. de Bock","doi":"10.1002/hem3.70265","DOIUrl":"https://doi.org/10.1002/hem3.70265","url":null,"abstract":"<p>Chromosomal translocations involving the mixed lineage leukemia (also known as <i>MLL</i> or <i>KMT2A</i>) gene occur in approximately 40% of children with acute myeloid leukemia (AML) under the age of 3. The fusion partners of <i>MLL</i> can vary widely with over 130 different ones identified to date. These fusions result in constitutive activation of MLL and ultimately drive the overexpression of key downstream target genes including <i>MEIS1</i> and <i>HOXA</i> cluster genes that leads to leukemia development. Critically, <i>MLL</i>-rearranged (<i>MLL</i>-r) leukemias all depend on menin for growth and survival that in turn has driven the development of menin inhibitors (e.g., revumenib) currently being evaluated in numerous clinical trials. I refer the reader to two excellent recent reviews on the biology of <i>MLL</i>-r AML and early clinical trial data.<span><sup>1, 2</sup></span></p><p>Another common chromosomal translocation in AML is the one involving the <i>NUP98</i> gene, which has up to 30 distinct fusion partners. These have been shown to drive transcription condensates in the nucleus as well as changes in chromatin architecture that are thought to play an important role in rewiring cells toward leukemia development.<span><sup>3</sup></span> Importantly, NUP98 fusion proteins interact with wild-type MLL (KMT2A) complexes, and this also drives high expression of <i>MEIS1</i> and <i>HOXA</i> cluster genes. Notably, <i>NUP98</i>-rearranged (<i>NUP98</i>-r) AML cases are also very sensitive to menin inhibitors.<span><sup>4</sup></span></p><p>Nevertheless, just like other targeted therapies, resistance mechanisms have now been identified in AML cases treated with menin inhibitors. These include mutations in the drug pocket of menin around amino acid residues M327, G331, T349, and S160 that prevent the binding of menin inhibitors. Alternative mechanisms include epigenetic wiring such as loss of components of the PRC1.1 complex.<span><sup>2</sup></span> Interestingly, a new study showed that chemotherapy could also render <i>MLL</i>-r leukemia more resistant to menin inhibition without prior exposure to menin inhibitors.<span><sup>5</sup></span> Therefore, combination strategies are needed to increase the chances of complete remission and cure.</p><p>Two new studies<span><sup>6, 7</sup></span> have now been published back-to-back in <i>Cancer Discovery</i> addressing this very issue, with both papers looking at the efficacy of combining menin inhibitors with the newly developed orally bioavailable KAT6A/B selective inhibitor PF-9363. Both KAT6A and KAT6B are members of the MYST (MOZ/KAT6A, Ybf2/Sas3, Sas2, and Tip60) family of histone acetyltransferases (HATs) that also includes KAT5, KAT7, and KAT8. These all differentially regulate the acetylation of lysine residues on histone 3, with KAT6A and KAT6B driving H3K23Ac whilst KAT7 is responsible for H3K14Ac for example. Whilst this family is important in regulating chromatin structure and","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70265","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145580893","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}
Peter Johnstone, Peter Allen, Pinky Jimenez-Agrawal, Samir Agrawal, Stephen P. Hibbs
Overdiagnosis describes situations where ‘people are labelled with or treated for a disease that would never cause them harm’.1 We believe the current diagnostic paradigm for chronic lymphocytic leukaemia (CLL) constitutes overdiagnosis, imposing a diagnosis which can cause significant distress to patients without benefit. In this article, we begin with the personal experience of one of the authors (Peter Allen), which powerfully depicts the harms of a CLL label. We then consider the historical context of current CLL diagnostic criteria and question their suitability. Finally, we consider other possible approaches to CLL diagnosis, including intentional non-investigation and de-diagnosis.
Peter Johnstone: Conceptualisation; writing—original draft; writing—review and editing; project administration. Peter Allen: Writing—review and editing; writing—original draft. Pinky Jimenez-Agrawal: Writing—review and editing. Samir Agrawal: Writing—review and editing. Stephen P. Hibbs: Conceptualization; writing—original draft; project administration; writing—review and editing.
Pinky Jimenez-Agrawal reports speaker engagements with AstraZeneca, BeOne (formerly BeiGene) and Eli Lilly. Samir Agrawal reports speaker engagements with AbbVie, BeOne (formerly BeiGene) and AstraZeneca.
The remaining authors (P.J., P.A. and S.P.H.) have no relevant conflicts of interest to declare.
S.P.H. is supported by a HARP doctoral research fellowship, funded by the Wellcome Trust (Grant number 223500/Z/21/Z).
Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.
过度诊断描述的是“人们被贴上了一种永远不会对他们造成伤害的疾病的标签或接受治疗”的情况我们认为,目前慢性淋巴细胞白血病(CLL)的诊断范式构成了过度诊断,强加的诊断可能会给患者带来巨大的痛苦,而没有益处。在本文中,我们从其中一位作者(Peter Allen)的个人经历开始,他有力地描述了CLL标签的危害。然后,我们考虑当前CLL诊断标准的历史背景,并质疑其适用性。最后,我们考虑了CLL诊断的其他可能方法,包括故意不调查和去诊断。彼得·约翰斯通:概念化;原创作品草案;写作——审阅和编辑;项目管理。彼得·艾伦:写作、评论和编辑;原创作品。Pinky Jimenez-Agrawal:写作、评论和编辑。萨米尔·阿格拉瓦尔:写作、评论和编辑。Stephen P. Hibbs:概念化;原创作品草案;项目管理;写作-审查和编辑。Pinky Jimenez-Agrawal报道了阿斯利康、BeOne(原百济神州)和礼来公司的演讲。Samir Agrawal报道了与艾伯维、BeOne(原BeiGene)和阿斯利康的演讲。其余作者(p.j.、P.A.和S.P.H.)没有相关利益冲突需要申报。由惠康信托基金(资助号223500/Z/21/Z)资助的HARP博士研究奖学金支持。数据共享不适用于本文,因为在当前研究期间没有生成或分析数据集。
{"title":"De-diagnosing chronic lymphocytic leukaemia: An ethical and scientific case for changing diagnostic criteria","authors":"Peter Johnstone, Peter Allen, Pinky Jimenez-Agrawal, Samir Agrawal, Stephen P. Hibbs","doi":"10.1002/hem3.70252","DOIUrl":"https://doi.org/10.1002/hem3.70252","url":null,"abstract":"<p>Overdiagnosis describes situations where ‘people are labelled with or treated for a disease that would never cause them harm’.<span><sup>1</sup></span> We believe the current diagnostic paradigm for chronic lymphocytic leukaemia (CLL) constitutes overdiagnosis, imposing a diagnosis which can cause significant distress to patients without benefit. In this article, we begin with the personal experience of one of the authors (Peter Allen), which powerfully depicts the harms of a CLL label. We then consider the historical context of current CLL diagnostic criteria and question their suitability. Finally, we consider other possible approaches to CLL diagnosis, including intentional non-investigation and de-diagnosis.</p><p><b>Peter Johnstone</b>: Conceptualisation; writing—original draft; writing—review and editing; project administration. <b>Peter Allen</b>: Writing—review and editing; writing—original draft. <b>Pinky Jimenez-Agrawal</b>: Writing—review and editing. <b>Samir Agrawal</b>: Writing—review and editing. <b>Stephen P. Hibbs</b>: Conceptualization; writing—original draft; project administration; writing—review and editing.</p><p>Pinky Jimenez-Agrawal reports speaker engagements with AstraZeneca, BeOne (formerly BeiGene) and Eli Lilly. Samir Agrawal reports speaker engagements with AbbVie, BeOne (formerly BeiGene) and AstraZeneca.</p><p>The remaining authors (P.J., P.A. and S.P.H.) have no relevant conflicts of interest to declare.</p><p>S.P.H. is supported by a HARP doctoral research fellowship, funded by the Wellcome Trust (Grant number 223500/Z/21/Z).</p><p>Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70252","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145580894","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}
Mwanasha H. Merrill, Robert A. Redd, Nicholas Lambert, Paolo F. Caimi, Priyanka Pullarkat, Richard C. Godby, David A. Bond, Graham T. Wehmeyer, Jason Romancik, Behzad Amoozgar, Lori Leslie, Loretta J. Nastoupil, Jennifer L. Crombie, Jeremy S. Abramson, Arushi Khurana, Grzegorz S. Nowakowski, Kami Maddocks, Sarah C. Rutherford, Brad Kahl, Michelle Okwali, Michael J. Buege, Connie L. Batlevi, Philippe Armand, Gilles Salles, David A. Qualls
The treatment landscape of relapsed and refractory (R/R) diffuse large B-cell lymphoma (DLBCL) has significantly improved with the development of CD19-directed chimeric antigen receptor T-cell therapy (CAR T).1, 2 Even so, approximately 50%–60% of patients treated with CAR T will ultimately experience disease progression.3
Tafasitamab (tafa) and loncastuximab tesirine (lonca) both target CD19 and are approved for the treatment of R/R DLBCL. Following the approval of CAR T in the second-line setting,4, 5 these agents may be used to treat DLBCL after CAR T failure. Notably, the L-MIND study of tafa with lenalidomide (tafa/len) excluded patients with prior CAR T, while the LOTIS-2 study for lonca enrolled only 13 post–CAR T patients.6, 7
Prior reports, including our own, have shown poor outcomes with CD19-directed therapies following CAR T, though it remains unclear whether this reflects the effects of prior CAR T itself or underlying high-risk disease features.8, 9
We aimed to evaluate the impact of prior CD19-directed therapy on subsequent CD19-based treatment, focusing on the impact of prior CD19 CAR T on tafa/len efficacy. We compared CAR T-exposed versus CAR T-naïve patients receiving tafa/len, and used propensity score matching to address baseline imbalances between CAR-exposed and CAR-naïve cohorts. We also explored CD19 expression patterns and outcomes with other CD19-directed sequences, including lonca and CAR T.
This multicenter, retrospective cohort study included adults (≥18 years) with R/R DLBCL treated with tafa between August 2020 and August 2022 across 11 institutions. Eligible histologies included DLBCL-not otherwise specified (NOS), high-grade B-cell lymphoma (HGBCL), transformed indolent lymphomas, T-cell/histiocyte-rich large B-cell lymphoma, and primary mediastinal B-cell lymphoma. Responses to therapy were evaluated by each site using routine imaging and assessments. CD19 expression was locally assessed by immunohistochemistry or flow cytometry and defined as positive by local center criteria.
We previously reported the overall outcomes of tafa/len.8 Subsequently, additional data were collected on the outcomes of other CD19-directed therapies aside from tafa. This study was approved by the institutional review boards of all participating institutions.
Patients were categorized into CAR T-naïve and CAR T-exposed cohorts based on treatment exposure at the time of tafa/len initiation. Patients who had received prior CD19-directed therapy other than CAR T before tafa/len were excluded. Descriptive statistics were reported by cohort. Fisher's exact and Wilcoxon rank-sum tests were used to assess differences by cohort for categorical and continuous variables, respectively.
The primary outcome was progression-free survival (PFS) from tafa initiation
随着cd19靶向嵌合抗原受体T细胞疗法(CAR - T)的发展,复发和难治性(R/R)弥漫性大b细胞淋巴瘤(DLBCL)的治疗前景显著改善。即便如此,大约50%-60%接受CAR - T治疗的患者最终会经历疾病进展。tafasitamab (tafa)和loncastuximab tesirine (lonca)均靶向CD19,并被批准用于治疗R/R DLBCL。随着CAR - T在二线治疗中的批准,这些药物可能用于治疗CAR - T治疗失败后的DLBCL。值得注意的是,他法联合来那度胺(tafa/len)的L-MIND研究排除了既往CAR - T患者,而lonca的LOTIS-2研究仅纳入了13例CAR - T后患者。6,7先前的报告,包括我们自己的报告,显示CAR - T后cd19定向治疗的预后不佳,尽管尚不清楚这是否反映了先前CAR - T本身的影响或潜在的高风险疾病特征。8,9我们的目的是评估先前的CD19靶向治疗对随后的基于CD19的治疗的影响,重点是先前的CD19 CAR - T对tafa/len疗效的影响。我们比较了CAR- t暴露组和CAR- T-naïve接受tafa/len的患者,并使用倾向评分匹配来解决CAR- t暴露组和CAR-naïve队列之间的基线不平衡。我们还探讨了CD19的表达模式和其他CD19定向序列的结果,包括lonca和CAR t。这项多中心回顾性队列研究包括11个机构在2020年8月至2022年8月期间接受他法治疗的R/R DLBCL成人(≥18岁)。符合条件的组织学包括dlbcl - non - specified (NOS)、高级别b细胞淋巴瘤(HGBCL)、转化惰性淋巴瘤、富含t细胞/组织细胞的大b细胞淋巴瘤和原发性纵隔b细胞淋巴瘤。每个部位使用常规成像和评估来评估对治疗的反应。通过免疫组织化学或流式细胞术局部评估CD19表达,并根据局部中心标准定义为阳性。我们之前报道了tafa/len的总体结果随后,收集了除tafa外其他cd19导向疗法的结果的额外数据。这项研究得到了所有参与机构的机构审查委员会的批准。根据tafa/len启动时的治疗暴露情况,将患者分为CAR - T-naïve和CAR - t暴露队列。在tafa/len之前接受过CAR - T以外的cd19定向治疗的患者被排除在外。描述性统计采用队列报告。采用Fisher精确和Wilcoxon秩和检验分别评估分类变量和连续变量的队列差异。主要终点是从tafa开始到进展或死亡的无进展生存期(PFS)。总生存期(OS)从开始到死亡或最后一次随访。采用Kaplan-Meier法估计生存率,采用log-rank检验和Cox回归模型进行比较,95%置信区间采用Greenwood法估计方差。单变量和多变量模型总结了每个系数的风险比(hr)、95%置信区间和Wald p值。使用似然比检验来评估调整模型中每个变量的总体关联。使用LASSO正则化对PFS的单变量Cox预测因子进行变量选择。使用倾向评分匹配(1:1,最近邻,无替代),将CAR - t暴露患者与CAR - t初始患者的匹配亚队列进行比较,考虑到年龄,东部肿瘤合作组(ECOG)的表现状态,乳酸脱氢酶(LDH)和疾病难治愈度-先前独立与PFS相关的多变量分析因素难治性被定义为在最后一次治疗的6个月内未达到完全缓解(CR)或进展。每组50例患者,中位随访14个月,该研究检测到中位PFS的1.5个月差异(功率85%,α = 0.05)。在R(生存和配对包)中进行分析。共纳入了来自11个中心的174名患者。表1概述了tafa/len启动时的基线患者特征。在41例有CAR - T应答数据的患者中,对既往CAR - T治疗的最佳总缓解(BOR)为73% (30/41),CR率为51%(21/41)。从CAR - T到tafa的中位时间为7.3个月(范围1-55个月,四分位数间距[IQR] 3.6-14.8个月)。在整个队列中,CAR - t暴露患者的完全缓解率(CRR)为19% (95% CI 9%-34%), CAR - T-naïve患者的完全缓解率为20% (95% CI 13-28) (P > 0.99)。CAR - t暴露患者的总缓解率(ORR)为19% (95% CI 9%-34%), CAR - T-naïve患者为36% (95% CI 27%-46%, P = 0.052)。接受CAR - T治疗的患者中位PFS较短(1.7个月vs 2.8个月,P = 0.009; HR: 1.61 [95% CI 1.12, 2]。 30],图1),中位OS为5.0 vs 7.5个月(P = 0.25)。在tafa/len启动后6个月,CAR - T-naïve患者的PFS为28% (95% CI 21-37),而CAR - t暴露患者的PFS为14% (95% CI 7-28)。对既往CAR - T暴露的患者进行单变量分析,以确定与PFS和OS相关的因素(图S1和S2)。从CAR - T输注到tafa/len启动的时间增加与PFS的改善相关(每增加6个月,HR 0.78 [95% CI 0.62-0.94])。CAR - T术后给予tafa/len治疗12个月和≥12个月时,中位PFS为1.4个月(95% CI 1.0 - 2.5)和4.3个月(95% CI 2.0-NR) (P = 0.0059)(图1C,D)。小于CAR - T的CR与较差的PFS (HR 2.86 [95% CI 1.43-5.87])、既往CAR - T的神经毒性(HR 2.44 [95% CI 1.04-5.16])、启动CAR - T时的国际预后指数(IPI)(评分4-5比0-2,HR 3.38 [95% CI 1.36-9.88])和LDH升高(HR 2.62 [95% CI 1.27-6.03])相关。接下来,我们进行了多变量分析,在单变量分析中评估与PFS显著相关的因素。从CAR - T到tafa/len的时间与PFS显著相关(每增加6个月,HR 0.77 [95% Cl 0.61-0.98];似然比检验,P = 0.02)。既往CAR - T神经毒性与更差的PFS相关,具有临界意义(HR 2.51 [95% Cl 1.01-6.23];似然比检验,P = 0.059)。IPI 3与1-2与较差的PFS相关(HR 3.56 [95% CI 1.08-11.79]),尽管总体IPI与PFS没有独立相关(似然比检验,P = 0.13)(图S3)。我们使用倾向匹配方法建立了一个由50名CAR - T-naïve患者组成的队列,这些患者与50名CAR - t暴露患者具有相似的特征,使用与tafa/len治疗后PFS相关的基线特征(表S1)。CAR暴露组的ORR为19% (9-34),CAR T-naïve组的ORR为39% (25-54)(P = 0.065)。CAR - t暴露的CRR为19% (9-34),CAR - T-naïve暴露的CRR为20%(10-34)。配对队列的中位随访时间为14个月。CAR - t暴露患者的中位PFS为1.7个月,而CA
{"title":"Outcomes of tafasitamab and lenalidomide in large B-cell lymphoma based on prior CD19-directed CAR T exposure","authors":"Mwanasha H. Merrill, Robert A. Redd, Nicholas Lambert, Paolo F. Caimi, Priyanka Pullarkat, Richard C. Godby, David A. Bond, Graham T. Wehmeyer, Jason Romancik, Behzad Amoozgar, Lori Leslie, Loretta J. Nastoupil, Jennifer L. Crombie, Jeremy S. Abramson, Arushi Khurana, Grzegorz S. Nowakowski, Kami Maddocks, Sarah C. Rutherford, Brad Kahl, Michelle Okwali, Michael J. Buege, Connie L. Batlevi, Philippe Armand, Gilles Salles, David A. Qualls","doi":"10.1002/hem3.70253","DOIUrl":"10.1002/hem3.70253","url":null,"abstract":"<p>The treatment landscape of relapsed and refractory (R/R) diffuse large B-cell lymphoma (DLBCL) has significantly improved with the development of CD19-directed chimeric antigen receptor T-cell therapy (CAR T).<span><sup>1, 2</sup></span> Even so, approximately 50%–60% of patients treated with CAR T will ultimately experience disease progression.<span><sup>3</sup></span></p><p>Tafasitamab (tafa) and loncastuximab tesirine (lonca) both target CD19 and are approved for the treatment of R/R DLBCL. Following the approval of CAR T in the second-line setting,<span><sup>4, 5</sup></span> these agents may be used to treat DLBCL after CAR T failure. Notably, the L-MIND study of tafa with lenalidomide (tafa/len) excluded patients with prior CAR T, while the LOTIS-2 study for lonca enrolled only 13 post–CAR T patients.<span><sup>6, 7</sup></span></p><p>Prior reports, including our own, have shown poor outcomes with CD19-directed therapies following CAR T, though it remains unclear whether this reflects the effects of prior CAR T itself or underlying high-risk disease features.<span><sup>8, 9</sup></span></p><p>We aimed to evaluate the impact of prior CD19-directed therapy on subsequent CD19-based treatment, focusing on the impact of prior CD19 CAR T on tafa/len efficacy. We compared CAR T-exposed versus CAR T-naïve patients receiving tafa/len, and used propensity score matching to address baseline imbalances between CAR-exposed and CAR-naïve cohorts. We also explored CD19 expression patterns and outcomes with other CD19-directed sequences, including lonca and CAR T.</p><p>This multicenter, retrospective cohort study included adults (≥18 years) with R/R DLBCL treated with tafa between August 2020 and August 2022 across 11 institutions. Eligible histologies included DLBCL-not otherwise specified (NOS), high-grade B-cell lymphoma (HGBCL), transformed indolent lymphomas, T-cell/histiocyte-rich large B-cell lymphoma, and primary mediastinal B-cell lymphoma. Responses to therapy were evaluated by each site using routine imaging and assessments. CD19 expression was locally assessed by immunohistochemistry or flow cytometry and defined as positive by local center criteria.</p><p>We previously reported the overall outcomes of tafa/len.<span><sup>8</sup></span> Subsequently, additional data were collected on the outcomes of other CD19-directed therapies aside from tafa. This study was approved by the institutional review boards of all participating institutions.</p><p>Patients were categorized into CAR T-naïve and CAR T-exposed cohorts based on treatment exposure at the time of tafa/len initiation. Patients who had received prior CD19-directed therapy other than CAR T before tafa/len were excluded. Descriptive statistics were reported by cohort. Fisher's exact and Wilcoxon rank-sum tests were used to assess differences by cohort for categorical and continuous variables, respectively.</p><p>The primary outcome was progression-free survival (PFS) from tafa initiation","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12626236/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145556764","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}
In a recent Editorial in HemaSphere, an important aspect of advanced therapy medicinal products (ATMPs) produced and used in therapy under the hospital exemption (HE) status is raised by Domanovic et al.1 The European Blood Alliance, together with scientific societies and professional organizations, strongly supports maintaining and strengthening the HE scheme provided that specific criteria are approved by Regulatory Authorities, European and National.
As the authors mark, a working system for monitoring outcomes in patients treated with ATMP/HE is lacking in the EU with public reporting, despite the clinical importance of this therapeutic modality. However, we also note that although important scientific and technological advancements were experienced since the establishment of ARMP Regulation in the European Union,2 the scientific community in general is not yet in a position to evaluate the whole results of chimeric antigen receptor T (CAR-T) cell therapies as applied with the commercial products available in the EU. This is mainly due to the lack of any active system gathering cumulative data regarding the short- and long-term outcomes of ATMPs. The data individually kept by each medical center or by marketed CAR-T cell suppliers cannot provide a global insight to this mode of treatment. Besides several parameters involved in evaluating the results (population group applied, disease, age, sex, management history, comorbidities, etc.) before a final argument on the efficacy of this treatment is achieved, the variability of commercial products has also to be accounted for. Different particular products produced by different protocols may also turn to have different therapeutic effects or untoward effects. Therefore, treatment trials with particular commercial products may be non-comparable as far as the outcome is concerned. It is only a cumulative analysis that can permit such a conclusion.
It seems that it is now the proper time for establishing a monitoring system for the outcomes in cases treated with ATMPs/CAR-T cell approved centrally by commercial products or via the HE mode in Europe. The authorization of these products was in principle a “conditional” one; the regulatory authorities and scientists must insist on consistent quality, safety, and efficacy standards.
Kostas Konstantopoulos: Conceptualization; writing—original draft; writing—review and editing.
The author declares no conflicts of interest.
This research received no funding.
The data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions.
{"title":"Monitoring outcomes in patients treated by ATMPs","authors":"Kostas Konstantopoulos","doi":"10.1002/hem3.70257","DOIUrl":"https://doi.org/10.1002/hem3.70257","url":null,"abstract":"<p>In a recent Editorial in <i>HemaSphere</i>, an important aspect of advanced therapy medicinal products (ATMPs) produced and used in therapy under the hospital exemption (HE) status is raised by Domanovic et al.<span><sup>1</sup></span> The European Blood Alliance, together with scientific societies and professional organizations, strongly supports maintaining and strengthening the HE scheme provided that specific criteria are approved by Regulatory Authorities, European and National.</p><p>As the authors mark, a working system for monitoring outcomes in patients treated with ATMP/HE is lacking in the EU with public reporting, despite the clinical importance of this therapeutic modality. However, we also note that although important scientific and technological advancements were experienced since the establishment of ARMP Regulation in the European Union,<span><sup>2</sup></span> the scientific community in general is not yet in a position to evaluate the whole results of chimeric antigen receptor T (CAR-T) cell therapies as applied with the commercial products available in the EU. This is mainly due to the lack of any active system gathering cumulative data regarding the short- and long-term outcomes of ATMPs. The data individually kept by each medical center or by marketed CAR-T cell suppliers cannot provide a global insight to this mode of treatment. Besides several parameters involved in evaluating the results (population group applied, disease, age, sex, management history, comorbidities, etc.) before a final argument on the efficacy of this treatment is achieved, the variability of commercial products has also to be accounted for. Different particular products produced by different protocols may also turn to have different therapeutic effects or untoward effects. Therefore, treatment trials with particular commercial products may be non-comparable as far as the outcome is concerned. It is only a cumulative analysis that can permit such a conclusion.</p><p>It seems that it is now the proper time for establishing a monitoring system for the outcomes in cases treated with ATMPs/CAR-T cell approved centrally by commercial products or via the HE mode in Europe. The authorization of these products was in principle a “conditional” one; the regulatory authorities and scientists must insist on consistent quality, safety, and efficacy standards.</p><p><b>Kostas Konstantopoulos</b>: Conceptualization; writing—original draft; writing—review and editing.</p><p>The author declares no conflicts of interest.</p><p>This research received no funding.</p><p>The data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70257","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145530069","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}
Gerotziafas G, Fotiou D, Nijhof I, et al. Prevention and treatment of venous thromboembolism in patients with multiple myeloma: clinical practice guidelines on behalf of the European Myeloma Network. HemaSphere. 2025;9(8):e70177. doi:10.1002/hem3.70177
In the author listing of the manuscript, the name of an author was incorrectly listed as Alessandra Laroca. The correct name is Alessandra Larocca.
The original publication has been corrected. We apologize for this error.
{"title":"Correction to “Prevention and treatment of venous thromboembolism in patients with multiple myeloma: Clinical practice guidelines on behalf of the European Myeloma Network”","authors":"","doi":"10.1002/hem3.70250","DOIUrl":"https://doi.org/10.1002/hem3.70250","url":null,"abstract":"<p>Gerotziafas G, Fotiou D, Nijhof I, et al. Prevention and treatment of venous thromboembolism in patients with multiple myeloma: clinical practice guidelines on behalf of the European Myeloma Network. <i>HemaSphere</i>. 2025;9(8):e70177. doi:10.1002/hem3.70177</p><p>In the author listing of the manuscript, the name of an author was incorrectly listed as Alessandra Laroca. The correct name is Alessandra Larocca.</p><p>The original publication has been corrected. We apologize for this error.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 11","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70250","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145530070","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}