Myrthe J. van Dijk, Titine J. J. Ruiter, Sigrid van der Veen, Minke A. E. Rab, Brigitte A. van Oirschot, Jennifer Bos, Cleo Derichs, Anita W. Rijneveld, Marjon H. Cnossen, Erfan Nur, Bart J. Biemond, Marije Bartels, Roger E. G. Schutgens, Wouter W. van Solinge, Judith J. M. Jans, Eduard J. van Beers, Richard van Wijk
Mitapivat is an investigational, oral, small-molecule allosteric activator of pyruvate kinase (PK). PK is a regulatory glycolytic enzyme that is key in providing the red blood cell (RBC) with sufficient amounts of adenosine triphosphate (ATP). In sickle cell disease (SCD), decreased 2,3-DPG levels increase the oxygen affinity of hemoglobin, thereby preventing deoxygenation and polymerization of sickle hemoglobin. The PK activator mitapivat has been shown to decrease levels of 2,3-DPG and increase levels of ATP in RBCs in patients with SCD. In this phase 2, investigator-initiated, open-label study (https://www.clinicaltrialsregister.eu/ NL8517; EudraCT 2019-003438-18), untargeted metabolomics was used to explore the overall metabolic effects of 8-week treatment with mitapivat in the dose-finding period. In total, 1773 unique metabolites were identified in dried blood spots of whole blood from ten patients with SCD and 42 healthy controls (HCs). The metabolic phenotype of patients with SCD revealed alterations in 139/1773 (7.8%) metabolites at baseline when compared to HCs (false discovery rate-adjusted p < 0.05), including increases of (derivatives of) polyamines, purines, and acyl carnitines. Eight-week treatment with mitapivat in nine patients with SCD altered 85/1773 (4.8%) of the total metabolites and 18/139 (12.9%) of the previously identified altered metabolites in SCD (unadjusted p < 0.05). Effects were observed on a broad spectrum of metabolites and were not limited to glycolytic intermediates. Our results show the relevance of metabolic profiling in SCD, not only to unravel potential pathophysiological pathways and biomarkers in multisystem diseases but also to determine the effect of treatment.
Mitapivat 是一种正在研究的丙酮酸激酶(PK)口服小分子异位激活剂。PK是一种调节性糖酵解酶,是为红细胞(RBC)提供足量三磷酸腺苷(ATP)的关键。在镰状细胞病(SCD)中,2,3-DPG 水平的降低会增加血红蛋白的氧亲和力,从而阻止镰状血红蛋白的脱氧和聚合。PK 激活剂米他匹伐已被证明能降低 SCD 患者红细胞中的 2,3-DPG 含量并提高 ATP 含量。在这项由研究者发起的开放标签 2 期研究(https://www.clinicaltrialsregister.eu/ NL8517; EudraCT 2019-003438-18)中,采用了非靶向代谢组学来探索米他匹伐在剂量探索期进行 8 周治疗的总体代谢效应。在10名SCD患者和42名健康对照组(HCs)的全血干血斑中,共鉴定出1773种独特的代谢物。与健康对照组相比,SCD 患者的代谢表型显示基线时有 139/1773 个(7.8%)代谢物发生了改变(经假发现率调整的 p p
{"title":"Metabolic blood profile and response to treatment with the pyruvate kinase activator mitapivat in patients with sickle cell disease","authors":"Myrthe J. van Dijk, Titine J. J. Ruiter, Sigrid van der Veen, Minke A. E. Rab, Brigitte A. van Oirschot, Jennifer Bos, Cleo Derichs, Anita W. Rijneveld, Marjon H. Cnossen, Erfan Nur, Bart J. Biemond, Marije Bartels, Roger E. G. Schutgens, Wouter W. van Solinge, Judith J. M. Jans, Eduard J. van Beers, Richard van Wijk","doi":"10.1002/hem3.109","DOIUrl":"10.1002/hem3.109","url":null,"abstract":"<p>Mitapivat is an investigational, oral, small-molecule allosteric activator of pyruvate kinase (PK). PK is a regulatory glycolytic enzyme that is key in providing the red blood cell (RBC) with sufficient amounts of adenosine triphosphate (ATP). In sickle cell disease (SCD), decreased 2,3-DPG levels increase the oxygen affinity of hemoglobin, thereby preventing deoxygenation and polymerization of sickle hemoglobin. The PK activator mitapivat has been shown to decrease levels of 2,3-DPG and increase levels of ATP in RBCs in patients with SCD. In this phase 2, investigator-initiated, open-label study (https://www.clinicaltrialsregister.eu/ NL8517; EudraCT 2019-003438-18), untargeted metabolomics was used to explore the overall metabolic effects of 8-week treatment with mitapivat in the dose-finding period. In total, 1773 unique metabolites were identified in dried blood spots of whole blood from ten patients with SCD and 42 healthy controls (HCs). The metabolic phenotype of patients with SCD revealed alterations in 139/1773 (7.8%) metabolites at baseline when compared to HCs (false discovery rate-adjusted <i>p</i> < 0.05), including increases of (derivatives of) polyamines, purines, and acyl carnitines. Eight-week treatment with mitapivat in nine patients with SCD altered 85/1773 (4.8%) of the total metabolites and 18/139 (12.9%) of the previously identified altered metabolites in SCD (unadjusted <i>p</i> < 0.05). Effects were observed on a broad spectrum of metabolites and were not limited to glycolytic intermediates. Our results show the relevance of metabolic profiling in SCD, not only to unravel potential pathophysiological pathways and biomarkers in multisystem diseases but also to determine the effect of treatment.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11196954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450365","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}
Alexandre Fagnan, Zakia Aid, Marie Baille, Aneta Drakul, Elie Robert, Cécile K. Lopez, Cécile Thirant, Yann Lecluse, Julie Rivière, Cathy Ignacimouttou, Silvia Salmoiraghi, Eduardo Anguita, Audrey Naimo, Christophe Marzac, Françoise Pflumio, Sébastien Malinge, Christian Wichmann, Yun Huang, Camille Lobry, Julie Chaumeil, Eric Soler, Jean-Pierre Bourquin, Claus Nerlov, Olivier A. Bernard, Juerg Schwaller, Thomas Mercher
Transcriptional cofactors of the ETO family are recurrent fusion partners in acute leukemia. We characterized the ETO2 regulome by integrating transcriptomic and chromatin binding analyses in human erythroleukemia xenografts and controlled ETO2 depletion models. We demonstrate that beyond its well-established repressive activity, ETO2 directly activates transcription of MYB, among other genes. The ETO2-activated signature is associated with a poorer prognosis in erythroleukemia but also in other acute myeloid and lymphoid leukemia subtypes. Mechanistically, ETO2 colocalizes with EP300 and MYB at enhancers supporting the existence of an ETO2/MYB feedforward transcription activation loop (e.g., on MYB itself). Both small-molecule and PROTAC-mediated inhibition of EP300 acetyltransferases strongly reduced ETO2 protein, chromatin binding, and ETO2-activated transcripts. Taken together, our data show that ETO2 positively enforces a leukemia maintenance program that is mediated in part by the MYB transcription factor and that relies on acetyltransferase cofactors to stabilize ETO2 scaffolding activity.
{"title":"The ETO2 transcriptional cofactor maintains acute leukemia by driving a MYB/EP300-dependent stemness program","authors":"Alexandre Fagnan, Zakia Aid, Marie Baille, Aneta Drakul, Elie Robert, Cécile K. Lopez, Cécile Thirant, Yann Lecluse, Julie Rivière, Cathy Ignacimouttou, Silvia Salmoiraghi, Eduardo Anguita, Audrey Naimo, Christophe Marzac, Françoise Pflumio, Sébastien Malinge, Christian Wichmann, Yun Huang, Camille Lobry, Julie Chaumeil, Eric Soler, Jean-Pierre Bourquin, Claus Nerlov, Olivier A. Bernard, Juerg Schwaller, Thomas Mercher","doi":"10.1002/hem3.90","DOIUrl":"https://doi.org/10.1002/hem3.90","url":null,"abstract":"<p>Transcriptional cofactors of the ETO family are recurrent fusion partners in acute leukemia. We characterized the ETO2 regulome by integrating transcriptomic and chromatin binding analyses in human erythroleukemia xenografts and controlled ETO2 depletion models. We demonstrate that beyond its well-established repressive activity, ETO2 directly activates transcription of MYB, among other genes. The ETO2-activated signature is associated with a poorer prognosis in erythroleukemia but also in other acute myeloid and lymphoid leukemia subtypes. Mechanistically, ETO2 colocalizes with EP300 and MYB at enhancers supporting the existence of an ETO2/MYB feedforward transcription activation loop (e.g., on MYB itself). Both small-molecule and PROTAC-mediated inhibition of EP300 acetyltransferases strongly reduced ETO2 protein, chromatin binding, and ETO2-activated transcripts. Taken together, our data show that ETO2 positively enforces a leukemia maintenance program that is mediated in part by the MYB transcription factor and that relies on acetyltransferase cofactors to stabilize ETO2 scaffolding activity.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.90","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141430255","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}
Hosuk Ryou, Korsuk Sirinukunwattana, Ruby Wood, Alan Aberdeen, Jens Rittscher, Olga K. Weinberg, Robert Hasserjian, Olga Pozdnyakova, Frank Peale, Brian Higgins, Pontus Lundberg, Kerstin Trunzer, Claire N. Harrison, Daniel Royston
<p>Accurate assessment of bone marrow fibrosis is central to the diagnosis and assessment of patients with myeloproliferative neoplasms (MPNs).<span><sup>1-3</sup></span> However, European consensus criteria for fibrosis are subjective, only semiquantitative, and cannot fully capture sample fibrosis heterogeneity.<span><sup>4-6</sup></span> In response, we have recently demonstrated the potential of machine learning to improve the detection and quantitation of marrow fibrosis in MPN using routinely prepared bone marrow trephine (BMT) samples.<span><sup>7</sup></span> Such approaches can support accurate MPN classification/risk stratification and provide quantitative analysis of fibrosis heterogeneity, with the potential to support clinical trial teams in the evaluation of current and novel antifibrotic therapies.<span><sup>6</sup></span> Here, we report evidence of such utility in the context of stage 2 of a phase II study of zinpentraxin alfa in patients diagnosed with primary or secondary myelofibrosis (MF) [ClinicalTrials.gov identifier: NCT01981850]. The primary trial endpoint was bone marrow response (≥1 grade reduction from baseline fibrosis at any timepoint). Secondary endpoints included effects on disease-related anemia, thrombocytopenia, and constitutional symptoms.</p><p>Zinpentraxin alfa (ZPN; previously PRM-151) is a recombinant form of human pentraxin-2 (PTX2; also known as serum amyloid P component or SAP), a circulating endogenous regulator of the inflammatory response to tissue damage and a natural inhibitor of fibrosis.<span><sup>8-10</sup></span> In the open-label stage 1 of this phase 2 study, ZPN showed evidence of clinical activity and tolerable safety as monotherapy or in combination with ruxolitinib in patients with primary MF, post-polycythemia vera (PV) MF, or post-essential thrombocythemia (ET) MF.<span><sup>11</sup></span> A subsequent randomized dose-ranging study (stage 2) evaluated the efficacy and safety of three different doses of ZPN as monotherapy in patients with IPSS intermediate-1, intermediate-2, and high-risk primary MF, post-PV MF, or post-ET MF who were anemic or thrombocytopenic and ineligible for, intolerant of, or had an inadequate prior response to ruxolitinib.<span><sup>12</sup></span> Patients were randomized to receive 0.3, 3.0, or 10.0 mg/kg ZPN on Days 1, 3, and 5 of cycle 1 and every 4 weeks thereafter for up to nine cycles. Reticulin-stained BMTs from three timepoints (screening, cycle 4 [C4D1], and cycle 9 [C9D29]) were analyzed for a subset of patients enrolled in the stage 2 study for whom digital scanned images were available at all three timepoints (50/97) (Figure 1A,B). Prior manual assessment of marrow fibrosis had been performed as part of a blinded, independent central review by three expert hematopathologists. Quantitative assessment of fibrosis using Continuous Indexing of Fibrosis (CIF) was performed by automated analyses as previously described.<span><sup>7</sup></span> Briefly, CIF
{"title":"Quantitative analysis of bone marrow fibrosis highlights heterogeneity in myelofibrosis and augments histological assessment: An Insight from a phase II clinical study of zinpentraxin alfa","authors":"Hosuk Ryou, Korsuk Sirinukunwattana, Ruby Wood, Alan Aberdeen, Jens Rittscher, Olga K. Weinberg, Robert Hasserjian, Olga Pozdnyakova, Frank Peale, Brian Higgins, Pontus Lundberg, Kerstin Trunzer, Claire N. Harrison, Daniel Royston","doi":"10.1002/hem3.105","DOIUrl":"https://doi.org/10.1002/hem3.105","url":null,"abstract":"<p>Accurate assessment of bone marrow fibrosis is central to the diagnosis and assessment of patients with myeloproliferative neoplasms (MPNs).<span><sup>1-3</sup></span> However, European consensus criteria for fibrosis are subjective, only semiquantitative, and cannot fully capture sample fibrosis heterogeneity.<span><sup>4-6</sup></span> In response, we have recently demonstrated the potential of machine learning to improve the detection and quantitation of marrow fibrosis in MPN using routinely prepared bone marrow trephine (BMT) samples.<span><sup>7</sup></span> Such approaches can support accurate MPN classification/risk stratification and provide quantitative analysis of fibrosis heterogeneity, with the potential to support clinical trial teams in the evaluation of current and novel antifibrotic therapies.<span><sup>6</sup></span> Here, we report evidence of such utility in the context of stage 2 of a phase II study of zinpentraxin alfa in patients diagnosed with primary or secondary myelofibrosis (MF) [ClinicalTrials.gov identifier: NCT01981850]. The primary trial endpoint was bone marrow response (≥1 grade reduction from baseline fibrosis at any timepoint). Secondary endpoints included effects on disease-related anemia, thrombocytopenia, and constitutional symptoms.</p><p>Zinpentraxin alfa (ZPN; previously PRM-151) is a recombinant form of human pentraxin-2 (PTX2; also known as serum amyloid P component or SAP), a circulating endogenous regulator of the inflammatory response to tissue damage and a natural inhibitor of fibrosis.<span><sup>8-10</sup></span> In the open-label stage 1 of this phase 2 study, ZPN showed evidence of clinical activity and tolerable safety as monotherapy or in combination with ruxolitinib in patients with primary MF, post-polycythemia vera (PV) MF, or post-essential thrombocythemia (ET) MF.<span><sup>11</sup></span> A subsequent randomized dose-ranging study (stage 2) evaluated the efficacy and safety of three different doses of ZPN as monotherapy in patients with IPSS intermediate-1, intermediate-2, and high-risk primary MF, post-PV MF, or post-ET MF who were anemic or thrombocytopenic and ineligible for, intolerant of, or had an inadequate prior response to ruxolitinib.<span><sup>12</sup></span> Patients were randomized to receive 0.3, 3.0, or 10.0 mg/kg ZPN on Days 1, 3, and 5 of cycle 1 and every 4 weeks thereafter for up to nine cycles. Reticulin-stained BMTs from three timepoints (screening, cycle 4 [C4D1], and cycle 9 [C9D29]) were analyzed for a subset of patients enrolled in the stage 2 study for whom digital scanned images were available at all three timepoints (50/97) (Figure 1A,B). Prior manual assessment of marrow fibrosis had been performed as part of a blinded, independent central review by three expert hematopathologists. Quantitative assessment of fibrosis using Continuous Indexing of Fibrosis (CIF) was performed by automated analyses as previously described.<span><sup>7</sup></span> Briefly, CIF","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.105","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141326447","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}
Maeve A. O'Reilly, William Wilson, David Burns, Andrea Kuhnl, Frances Seymour, Ben Uttenthal, Caroline Besley, Rajesh Alajangi, Thomas Creasey, Shankara Paneesha, Johnathon Elliot, Carlos Gonzalez Arias, Sunil Iyengar, Matthew R. Wilson, Alison Delaney, Lourdes Rubio, Jonathan Lambert, Khalil Begg, Stephen Boyle, Kathleen P. L. Cheok, Graham P. Collins, Claire Roddie, Rod Johnson, Robin Sanderson
Brexucabtagene autoleucel (brexu-cel) is an autologous CD19 CAR T-cell product, approved for relapsed/refractory (r/r) mantle cell lymphoma (MCL). In ZUMA-2, brexu-cel demonstrated impressive responses in patients failing ≥2 lines, including a bruton's tyrosine kinase inhibitor, with an overall and complete response rate of 93% and 67%, respectively. Here, we report our real-world intention-to-treat (ITT) outcomes for brexu-cel in consecutive, prospectively approved patients, from 12 institutions in the United Kingdom between February 2021 and June 2023, with a focus on feasibility, efficacy, and tolerability. Of 119 approved, 104 underwent leukapheresis and 83 received a brexu-cel infusion. Progressive disease (PD) and/or manufacturing (MF) were the most common reasons for failure to reach harvest and/or infusion. For infused patients, best overall and complete response rates were 87% and 81%, respectively. At a median follow-up of 13.3 months, median progression-free survival (PFS) for infused patients was 21 months (10.1–NA) with a 6- and 12-month PFS of 82% (95% confidence interval [CI], 71–89) and 62% (95% CI, 49–73), respectively. ≥Grade 3 cytokine release syndrome and neurotoxicity occurred in 12% and 22%, respectively. On multivariate analysis, inferior PFS was associated with male sex, bulky disease, ECOG PS > 1 and previous MF. Cumulative incidence of non-relapse mortality (NRM) was 6%, 15%, and 25% at 6, 12, and 24 months, respectively, and mostly attributable to infection. Outcomes for infused patients in the UK are comparable to ZUMA-2 and other real-world reports. However, ITT analysis highlights a significant dropout due to PD and/or MF. NRM events warrant further attention.
Brexucabtagene autoleucel(brexu-cel)是一种自体 CD19 CAR T 细胞产品,已获准用于治疗复发/难治(r/r)套细胞淋巴瘤(MCL)。在ZUMA-2研究中,brexu-cel对≥2种疗法(包括一种布氏酪氨酸激酶抑制剂)无效的患者产生了令人印象深刻的反应,总反应率和完全反应率分别为93%和67%。在此,我们报告了2021年2月至2023年6月期间,英国12家机构对连续、前瞻性获批患者进行brexu-cel治疗的真实世界意向治疗(ITT)结果,重点关注可行性、疗效和耐受性。在119名获批患者中,104人接受了白细胞清除术,83人接受了brexu-cel输注。疾病进展(PD)和/或制造(MF)是未能达到收获和/或输注的最常见原因。输注患者的最佳总体反应率和完全反应率分别为87%和81%。中位随访时间为13.3个月,输注患者的中位无进展生存期(PFS)为21个月(10.1-NA),6个月和12个月的PFS分别为82%(95%置信区间[CI],71-89)和62%(95% CI,49-73)。多变量分析显示,PFS较差与男性、大块疾病、ECOG PS > 1和既往MF有关。在6、12和24个月时,非复发死亡率(NRM)的累积发生率分别为6%、15%和25%,主要归因于感染。英国输液患者的治疗结果与 ZUMA-2 和其他真实世界的报告相当。然而,ITT分析显示,由于PD和/或MF,有大量患者退出治疗。NRM事件值得进一步关注。
{"title":"Brexucabtagene autoleucel for relapsed or refractory mantle cell lymphoma in the United Kingdom: A real-world intention-to-treat analysis","authors":"Maeve A. O'Reilly, William Wilson, David Burns, Andrea Kuhnl, Frances Seymour, Ben Uttenthal, Caroline Besley, Rajesh Alajangi, Thomas Creasey, Shankara Paneesha, Johnathon Elliot, Carlos Gonzalez Arias, Sunil Iyengar, Matthew R. Wilson, Alison Delaney, Lourdes Rubio, Jonathan Lambert, Khalil Begg, Stephen Boyle, Kathleen P. L. Cheok, Graham P. Collins, Claire Roddie, Rod Johnson, Robin Sanderson","doi":"10.1002/hem3.87","DOIUrl":"https://doi.org/10.1002/hem3.87","url":null,"abstract":"<p>Brexucabtagene autoleucel (brexu-cel) is an autologous CD19 CAR T-cell product, approved for relapsed/refractory (r/r) mantle cell lymphoma (MCL). In ZUMA-2, brexu-cel demonstrated impressive responses in patients failing ≥2 lines, including a bruton's tyrosine kinase inhibitor, with an overall and complete response rate of 93% and 67%, respectively. Here, we report our real-world intention-to-treat (ITT) outcomes for brexu-cel in consecutive, prospectively approved patients, from 12 institutions in the United Kingdom between February 2021 and June 2023, with a focus on feasibility, efficacy, and tolerability. Of 119 approved, 104 underwent leukapheresis and 83 received a brexu-cel infusion. Progressive disease (PD) and/or manufacturing (MF) were the most common reasons for failure to reach harvest and/or infusion. For infused patients, best overall and complete response rates were 87% and 81%, respectively. At a median follow-up of 13.3 months, median progression-free survival (PFS) for infused patients was 21 months (10.1–NA) with a 6- and 12-month PFS of 82% (95% confidence interval [CI], 71–89) and 62% (95% CI, 49–73), respectively. ≥Grade 3 cytokine release syndrome and neurotoxicity occurred in 12% and 22%, respectively. On multivariate analysis, inferior PFS was associated with male sex, bulky disease, ECOG PS > 1 and previous MF. Cumulative incidence of non-relapse mortality (NRM) was 6%, 15%, and 25% at 6, 12, and 24 months, respectively, and mostly attributable to infection. Outcomes for infused patients in the UK are comparable to ZUMA-2 and other real-world reports. However, ITT analysis highlights a significant dropout due to PD and/or MF. NRM events warrant further attention.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.87","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141315481","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}
Bela Patel, Amy A. Kirkwood, Clare J. Rowntree, Krisztina Z. Alapi, Emilio Barretta, Laura Clifton-Hadley, Tom Creasey, SooWah Lee, David I. Marks, Anthony V. Moorman, Nicholas Morley, Pip Patrick, Zaynab Rana, Anita Rijneveld, John A. Snowden, Adele K. Fielding
<p>Poor outcome for older patients with ALL has multiple attributions, including a higher incidence of high-risk genetic features,<span><sup>1</sup></span> and comorbidities as well as treatment intolerance.<span><sup>2, 3</sup></span> The phase 2 clinical trial UKALL60+ (NCT01616238) was a collaboration between the UK National Cancer Research Institute Adult ALL Group and the Haemato-Oncology Foundation for Adults in the Netherlands (HOVON) to study treatment choices, quality of life (QoL) and outcomes in older patients with ALL. UKALL60+ offered four treatment “pathways”: pathway A for <i>BCR::ABL1</i>+ ALL and pathways B, C, and D offering three choices of intensity for <i>BCR::ABL1</i> negative ALL (Intensive, Intensive-plus and Non-Intensive, respectively), to be selected by investigator and patients. A registration-only choice (Pathway E) was also available. Details of treatment regimens are given in Figure S1. There were no exclusions for any comorbidities. The primary endpoint was complete remission (CR) after a 2-phase induction. Secondary endpoints included event-free survival (EFS) and overall survival (OS), the predictive value of MRD (Ig/TCR quantification, EuroMRD criteria),<span><sup>4</sup></span> patient-reported outcomes, and the relationship between the baseline characteristics (Charlson index. ECOG, Karnofsky and Chemotherapy Risk Assessment Scale for High-Age Patients [CRASH] scores) and treatment option chosen.</p><p>Between January 2013 and November 2018, 121 eligible patients, median age 69 (interquartile range [IQR]: 65–73, range: 55–83), of whom 107 had B-ALL and 14 T-ALL, were recruited at 34 sites (Table S1). Baseline characteristics are shown in Table 1 alongside the characteristics of the 65 patients aged over 60 years that were recruited to the contemporaneous UKALL14 trial, age 25–65 years. A consort diagram is shown in Figure S2.</p><p>Fifty-one of 81 (63%) patients with <i>BCR::ABL1</i> negative disease were allocated to pathway B, 11% (9/81) to pathway C, and the remaining 26% (21/81) to pathway D. At a median follow-up: 65.9 months (IQR: 38.1–80.9), CR rate after two phases of induction, was achieved by 92% (70% confidence interval [CI]: 82.1–97.2) on pathway A, 70.6% (70% CI: 62.6–77.6) on pathway B, 55.6% (70% CI: 33.6–75.9) on pathway C and 47.6% (70% CI: 34.5%–61%) of those on pathway D. No participant achieved CR on study later than end of induction. Molecular remission occurred in 5/25 (20%; A), 13/51 (25.5%; B), 2/9 (22.2%; C), and 1/21 (4.8%; D) with data available. Only 26/121 (21.5%) patients achieved molecular remission at any point. The relationship between MRD and outcome at the three study timepoints is given in Table S2.</p><p>Ninety-six deaths were reported; 32 patients died without achieving CR (22/32, primary cause, ALL). Fifty-six patients died after relapse and eight died in CR (four from infection, three from second malignancies [small cell lung cancer, AML, and CMML] and one unknown). Surv
只有 21/106 例(19.8%)患者完成了所有治疗方案。)只有 21/106 例(19.8%)患者完成了所有方案治疗。在第1和第2次诱导期间,终止治疗的比例最高,分别为27/106(25.5%)和11/106(10.4%),主要是由于难治性/复发性疾病(19/38;50%)。复发/难治性 ALL 也是所有治疗组在其他时间点终止治疗的主要原因(33/47;70.2%)。106例患者中只有5例(4.7%)因毒性明显停止治疗。路径 D 的参与者明显比路径 B 的参与者年长(中位数为 73 岁 [IQR: 70-78] vs. 67 岁 [IQR: 62-70],p = 0.0001),且合并症较多;路径 D 中查尔斯顿指数为 7 或以上的患者有 9/21 人(45%),而路径 B 中仅有 8/51 人(16.7%)。在比较基线 QoL 指标和合并症时,路径 D 组患者更虚弱的情况也很明显,身体功能明显低于路径 B 组;中位数为 60.0(IQR:53.3-80)对 86.7(IQR:66.7-100),p = 0.014(表 S6 和 S7)。没有查尔斯顿指数为 7 分或以上的患者被分配到路径 C(p = 0.013)。与年龄相关的主要合并症在整个研究队列中很常见,包括心脏病 27/121 例(22.3%)、糖尿病 17/121 例(14.0%)、高血压 39/121 例(32.2%)和其他癌症 22/121 例(18.1%),其中 8 例曾患乳腺癌,7 例曾患血液恶性肿瘤。与路径A和路径D相比,接受路径B和路径C的患者住院治疗的时间更长,在诱导期间的影响最为明显(p = 0.0001),路径B和路径C参与者的住院时间分别为62.1%(46.3-96.7)和75.8%(68.8-83.0),而路径A和路径D参与者的住院时间分别为22.8%(IQR:9.8-55.6)和31.1%(IQR:14.5-51.5)。QoL按途径进行了比较--总结见补充结果。没有迹象表明强化程度最低的路径 D 能提供更好的 QoL,某些量表的得分在数值上低于路径 A-C(图 S4A-D)。与基线相比,QoL 的任何下降一般都出现在诱导阶段的末期,而在巩固 1 和维持 1 阶段末期,FACT 分数有所改善(表 S8)。我们将UKALL60+队列的EFS和OS与65名60-65岁患者的EFS和OS进行了比较,这些患者是在招募时间重叠的UKALL14成人ALL试验中接受治疗的。不出所料,UKALL14 的患者 ECOG 评分较低(40.0% 对 60.9% ECOG 0,P = 0.0068)。基线合并症较少(67.7% vs. 85.0%,p = 0.0059),尤其是心脏疾病(6.2% vs. 22.5% p = 0.0046)。虽然 CR 率更高;UKALL14 55/63 (87.3%) vs. UKALL60+ 82/118 (69.2%),如表 1 所示,UKALL14 3 年的 EFS 和 OS 率分别为 20.5% (11.5-31.3) 和 25.2% (15.2-36.5),UKALL60+ 为 18.8% (12.2-26.5) 和 23.2% (15.9-31.4)(图 1C、D)。然而,事件的类型有所不同,UKALL14中只有18/55(32.7%)人复发,9/55人在病情未缓解时死亡,26/55(47.2%)人在病情缓解时死亡,而UKALL60+中分别为56/95(58.9%)、32/95(33.7%)和7/95(7.4%)。在 UKALL14 60-65 岁的队列中,有 28/65 人接受了异体 SCT,结果有 3/28 人(46%)在缓解期死亡。5 在接受路径 B 治疗的患者中,高风险/极高风险遗传学患者与标准风险遗传学患者的预后无差异(EFS HR:1.35 [0.68-2.我们的生存数据与中位年龄相似的 GMALL 队列6 的数据基本一致,后者 3 年的 OS 为 32%。然而,UKALL60+人群的总体健康状况较差;与GMALL队列相比,27.5%的人的Charlson指数为>7,只有11%的人的指数为>3,与GMALL的合并症排除标准相符。与 GMALL 14% 的早期死亡率相比,我们观察到与治疗相关的死亡率很低。UKALL60+的治疗计划成功地将伤害降到了最低,但这并没有改善OS.出乎意料的是,根据UKALL14数据接受治疗的60-65岁患者的3年EFS和OS分别为20.5%和25.2%,与UKALL60+招募者的情况并无不同,生存曲线完全重叠(图1)。这些结果与 EBMT 一项针对 418 名 55 岁以上接受异体干细胞移植患者的研究中报告的结果相似;5 年 LFS 为 34%,但非复发死亡率为 51%。
{"title":"Results from UKALL60+, a phase 2 study in older patients with untreated acute lymphoblastic leukemia","authors":"Bela Patel, Amy A. Kirkwood, Clare J. Rowntree, Krisztina Z. Alapi, Emilio Barretta, Laura Clifton-Hadley, Tom Creasey, SooWah Lee, David I. Marks, Anthony V. Moorman, Nicholas Morley, Pip Patrick, Zaynab Rana, Anita Rijneveld, John A. Snowden, Adele K. Fielding","doi":"10.1002/hem3.88","DOIUrl":"https://doi.org/10.1002/hem3.88","url":null,"abstract":"<p>Poor outcome for older patients with ALL has multiple attributions, including a higher incidence of high-risk genetic features,<span><sup>1</sup></span> and comorbidities as well as treatment intolerance.<span><sup>2, 3</sup></span> The phase 2 clinical trial UKALL60+ (NCT01616238) was a collaboration between the UK National Cancer Research Institute Adult ALL Group and the Haemato-Oncology Foundation for Adults in the Netherlands (HOVON) to study treatment choices, quality of life (QoL) and outcomes in older patients with ALL. UKALL60+ offered four treatment “pathways”: pathway A for <i>BCR::ABL1</i>+ ALL and pathways B, C, and D offering three choices of intensity for <i>BCR::ABL1</i> negative ALL (Intensive, Intensive-plus and Non-Intensive, respectively), to be selected by investigator and patients. A registration-only choice (Pathway E) was also available. Details of treatment regimens are given in Figure S1. There were no exclusions for any comorbidities. The primary endpoint was complete remission (CR) after a 2-phase induction. Secondary endpoints included event-free survival (EFS) and overall survival (OS), the predictive value of MRD (Ig/TCR quantification, EuroMRD criteria),<span><sup>4</sup></span> patient-reported outcomes, and the relationship between the baseline characteristics (Charlson index. ECOG, Karnofsky and Chemotherapy Risk Assessment Scale for High-Age Patients [CRASH] scores) and treatment option chosen.</p><p>Between January 2013 and November 2018, 121 eligible patients, median age 69 (interquartile range [IQR]: 65–73, range: 55–83), of whom 107 had B-ALL and 14 T-ALL, were recruited at 34 sites (Table S1). Baseline characteristics are shown in Table 1 alongside the characteristics of the 65 patients aged over 60 years that were recruited to the contemporaneous UKALL14 trial, age 25–65 years. A consort diagram is shown in Figure S2.</p><p>Fifty-one of 81 (63%) patients with <i>BCR::ABL1</i> negative disease were allocated to pathway B, 11% (9/81) to pathway C, and the remaining 26% (21/81) to pathway D. At a median follow-up: 65.9 months (IQR: 38.1–80.9), CR rate after two phases of induction, was achieved by 92% (70% confidence interval [CI]: 82.1–97.2) on pathway A, 70.6% (70% CI: 62.6–77.6) on pathway B, 55.6% (70% CI: 33.6–75.9) on pathway C and 47.6% (70% CI: 34.5%–61%) of those on pathway D. No participant achieved CR on study later than end of induction. Molecular remission occurred in 5/25 (20%; A), 13/51 (25.5%; B), 2/9 (22.2%; C), and 1/21 (4.8%; D) with data available. Only 26/121 (21.5%) patients achieved molecular remission at any point. The relationship between MRD and outcome at the three study timepoints is given in Table S2.</p><p>Ninety-six deaths were reported; 32 patients died without achieving CR (22/32, primary cause, ALL). Fifty-six patients died after relapse and eight died in CR (four from infection, three from second malignancies [small cell lung cancer, AML, and CMML] and one unknown). Surv","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.88","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141264659","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}
Andreas J. M. Ferreri, Gerald Illerhaus, Jeanette K. Doorduijn, Dorothee P. Auer, Jacoline E. C. Bromberg, Teresa Calimeri, Kate Cwynarski, Christopher P. Fox, Khê Hoang-Xuan, Denis Malaise, Maurilio Ponzoni, Elisabeth Schorb, Carole Soussain, Lena Specht, Emanuele Zucca, Christian Buske, Mats Jerkeman, Martin Dreyling, EHA and ESMO Guidelines Committees
<p>Primary diffuse large B-cell lymphoma (DLBCL) of the central nervous system (CNS), termed primary CNS lymphoma (PCNSL), is an aggressive neoplasm presenting with disease limited to the CNS. PCNSL was recognised as a distinct entity by the 2017 World Health Organization (WHO) Classification of Tumours of Haematopoietic and Lymphoid Tissues.<span><sup>1</sup></span> In the 2022 edition of the WHO classification,<span><sup>2</sup></span> this neoplasm is classified in the ‘Large B-cell lymphomas of immune-privileged sites' group, whereas it is considered a specific entity in the International Consensus Classification of Mature Lymphoid Neoplasms.<span><sup>3</sup></span> This entity is also recognised by the WHO classification of CNS tumours.<span><sup>4</sup></span> PCNSL can arise in both immunocompetent individuals and in those who are immunosuppressed (e.g. individuals living with human immunodeficiency virus and patients receiving immunosuppressive therapies following organ transplant). While no clear predisposing factors have been recognised in immunocompetent individuals, the nature, intensity and duration of immune suppression can influence the risk of PCNSL in those who are immunocompromised.<span><sup>5</sup></span></p><p>This European Hematology Association (EHA)–European Society for Medical Oncology (ESMO) Clinical Practice Guideline (CPG) includes recommendations for the management of immunocompetent patients with PCNSL. In this population, PCNSL accounts for 2% of all primary CNS tumours and 4%-6% of extranodal lymphomas, with an incidence of 0.47/100 000 person-years.<span><sup>6</sup></span> PCNSL is typically diagnosed in the sixth or seventh decade of life, with a median age at diagnosis of 68 years and a slightly higher frequency in males.<span><sup>7</sup></span> Notably, the recent increase in incidence is limited to patients of >60 years. The incidence of PCNSL in African-American males of <50 years is more than twofold higher than that in Caucasian males of the same age.<span><sup>6</sup></span> Among elderly patients, however, incidence in Caucasian males is twofold higher than that in African-American males. Similar patterns, but with a lesser magnitude, are evident among females.<span><sup>6</sup></span></p><p>A comprehensive assessment of the extent of lymphoma involvement (see Supplementary Table S5, available at https://doi.org/10.1016/10.1016/j.annonc.2023.11.010) is mandatory to determine both the compartments involved within the CNS and the presence of concomitant systemic disease, as recommended by the IPCG guidelines.<span><sup>13</sup></span> Full neurological and oncohaematological evaluation is crucial before treatment planning. Gadolinium-enhanced MRI is the most relevant tool to define an extension of disease in the brain and spinal cord. Brain MRI should be repeated after biopsy and ideally within 14 days before starting treatment<span><sup>12</sup></span>; this is supported by extremely high prolifer
{"title":"Primary central nervous system lymphomas: EHA–ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up","authors":"Andreas J. M. Ferreri, Gerald Illerhaus, Jeanette K. Doorduijn, Dorothee P. Auer, Jacoline E. C. Bromberg, Teresa Calimeri, Kate Cwynarski, Christopher P. Fox, Khê Hoang-Xuan, Denis Malaise, Maurilio Ponzoni, Elisabeth Schorb, Carole Soussain, Lena Specht, Emanuele Zucca, Christian Buske, Mats Jerkeman, Martin Dreyling, EHA and ESMO Guidelines Committees","doi":"10.1002/hem3.89","DOIUrl":"https://doi.org/10.1002/hem3.89","url":null,"abstract":"<p>Primary diffuse large B-cell lymphoma (DLBCL) of the central nervous system (CNS), termed primary CNS lymphoma (PCNSL), is an aggressive neoplasm presenting with disease limited to the CNS. PCNSL was recognised as a distinct entity by the 2017 World Health Organization (WHO) Classification of Tumours of Haematopoietic and Lymphoid Tissues.<span><sup>1</sup></span> In the 2022 edition of the WHO classification,<span><sup>2</sup></span> this neoplasm is classified in the ‘Large B-cell lymphomas of immune-privileged sites' group, whereas it is considered a specific entity in the International Consensus Classification of Mature Lymphoid Neoplasms.<span><sup>3</sup></span> This entity is also recognised by the WHO classification of CNS tumours.<span><sup>4</sup></span> PCNSL can arise in both immunocompetent individuals and in those who are immunosuppressed (e.g. individuals living with human immunodeficiency virus and patients receiving immunosuppressive therapies following organ transplant). While no clear predisposing factors have been recognised in immunocompetent individuals, the nature, intensity and duration of immune suppression can influence the risk of PCNSL in those who are immunocompromised.<span><sup>5</sup></span></p><p>This European Hematology Association (EHA)–European Society for Medical Oncology (ESMO) Clinical Practice Guideline (CPG) includes recommendations for the management of immunocompetent patients with PCNSL. In this population, PCNSL accounts for 2% of all primary CNS tumours and 4%-6% of extranodal lymphomas, with an incidence of 0.47/100 000 person-years.<span><sup>6</sup></span> PCNSL is typically diagnosed in the sixth or seventh decade of life, with a median age at diagnosis of 68 years and a slightly higher frequency in males.<span><sup>7</sup></span> Notably, the recent increase in incidence is limited to patients of >60 years. The incidence of PCNSL in African-American males of <50 years is more than twofold higher than that in Caucasian males of the same age.<span><sup>6</sup></span> Among elderly patients, however, incidence in Caucasian males is twofold higher than that in African-American males. Similar patterns, but with a lesser magnitude, are evident among females.<span><sup>6</sup></span></p><p>A comprehensive assessment of the extent of lymphoma involvement (see Supplementary Table S5, available at https://doi.org/10.1016/10.1016/j.annonc.2023.11.010) is mandatory to determine both the compartments involved within the CNS and the presence of concomitant systemic disease, as recommended by the IPCG guidelines.<span><sup>13</sup></span> Full neurological and oncohaematological evaluation is crucial before treatment planning. Gadolinium-enhanced MRI is the most relevant tool to define an extension of disease in the brain and spinal cord. Brain MRI should be repeated after biopsy and ideally within 14 days before starting treatment<span><sup>12</sup></span>; this is supported by extremely high prolifer","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.89","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141245916","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}
Berit Müller-Meinhard, Nicole Seifert, Johanna Grund, Sarah Reinke, Fatih Yalcin, Helen Kaul, Sven Borchmann, Bastian von Tresckow, Peter Borchmann, Annette Plütschow, Julia Richter, Andreas Engert, Michael Altenbuchinger, Paul J. Bröckelmann, Wolfram Klapper
Hodgkin–Reed–Sternberg cells (HRSCs) in classic Hodgkin Lymphoma (HL) frequently lack expression of human leukocyte antigen class I (HLA-I), considered to hamper activation of cytotoxic T cells in the tumor microenvironment (TME). Here, we demonstrate HLA-I expression on HRSCs to be a strong determinant of TME composition whereas expression of HLA-II was associated with only minor differential gene expression in the TME. In HLA-I-positive HL the HRSC content and expression of CCL17/TARC in HRSCs are low, independent of the presence of Epstein–Barr virus in HRSCs. Additionally, HLA-I-positive HL shows a high content of CD8+ cytotoxic T cells. However, an increased expression of the inhibitory immune checkpoint LAG3 on CD8+ T cells in close proximity to HRSCs is observed. Suggesting interference with cytotoxic activity, we observed an absence of clonally expanded T cells in the TME. While HLA-I-positive HL is not associated with an unfavorable clinical course in our cohorts, they share features with the recently described H2 subtype of HL. Given the major differences in TME composition, immune checkpoint inhibitors may differ in their mechanism of action in HLA-I-positive compared to HLA-I-negative HL.
{"title":"Human leukocyte antigen (HLA) class I expression on Hodgkin–Reed–Sternberg cells is an EBV-independent major determinant of microenvironment composition in classic Hodgkin lymphoma","authors":"Berit Müller-Meinhard, Nicole Seifert, Johanna Grund, Sarah Reinke, Fatih Yalcin, Helen Kaul, Sven Borchmann, Bastian von Tresckow, Peter Borchmann, Annette Plütschow, Julia Richter, Andreas Engert, Michael Altenbuchinger, Paul J. Bröckelmann, Wolfram Klapper","doi":"10.1002/hem3.84","DOIUrl":"https://doi.org/10.1002/hem3.84","url":null,"abstract":"<p>Hodgkin–Reed–Sternberg cells (HRSCs) in classic Hodgkin Lymphoma (HL) frequently lack expression of human leukocyte antigen class I (HLA-I), considered to hamper activation of cytotoxic T cells in the tumor microenvironment (TME). Here, we demonstrate HLA-I expression on HRSCs to be a strong determinant of TME composition whereas expression of HLA-II was associated with only minor differential gene expression in the TME. In HLA-I-positive HL the HRSC content and expression of CCL17/TARC in HRSCs are low, independent of the presence of Epstein–Barr virus in HRSCs. Additionally, HLA-I-positive HL shows a high content of CD8+ cytotoxic T cells. However, an increased expression of the inhibitory immune checkpoint LAG3 on CD8+ T cells in close proximity to HRSCs is observed. Suggesting interference with cytotoxic activity, we observed an absence of clonally expanded T cells in the TME. While HLA-I-positive HL is not associated with an unfavorable clinical course in our cohorts, they share features with the recently described H2 subtype of HL. Given the major differences in TME composition, immune checkpoint inhibitors may differ in their mechanism of action in HLA-I-positive compared to HLA-I-negative HL.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.84","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141245671","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>HOX-homeobox transcription factors are best known for their prominent role in embryogenesis where they control body segment identity. This regulatory principle has been “recycled” in adult tissues. HOX proteins frequently regulate the differentiation of tissue stem cells and aberrant HOX function can induce derailed maturation and tumorigenesis. A paradigm for this principle is hematopoiesis where HOXA9, a member of the so-called abdominal HOX proteins, has acquired a notorious reputation for its frequent involvement in leukemogenesis. A number of recurrent genomic aberrations in acute leukemia are associated with elevated HOXA9 expression. Examples are KMT2A (MLL) and NUP98 fusion proteins as well as the very common NPM mutations, which all induce HOXA9 overproduction. Besides, a sizable portion of acute myeloid leukemia with normal karyotype shows abnormally high levels of HOXA9. Overall, HOXA9 dysregulation can be observed in more than 50% of all cases of myeloid leukemia, and generally, this is associated with a negative prognosis (Figure 1).<span><sup>1</sup></span></p><p>At a molecular level, HOXA9 acts as a so-called pioneer transcription factor that binds to a variety of AT-rich binding sites that mark prototypical enhancers and promoters important for hematopoietic precursor cells.<span><sup>2</sup></span> On a subset of these sequences, HOXA9 assembles a trimeric complex with two other homeodomain transcription factors MEIS1 and PBX3, which contribute further DNA binding specificity. This stabilizes the trimer and in conjunction with other transcription factors leads to the establishment of specific enhancer/promoter sequences in a process that has been called enhancer sharpening.<span><sup>3</sup></span> In consequence, genes necessary for the growth, proliferation, and survival of hematopoietic precursor cells are strongly activated. Prominent examples of HOXA9/MEIS1/PBX3 targets are <i>MYB</i>, <i>MYC</i>, <i>CDK6</i>, <i>BCL2</i>, and ribosomal genes to name just a few.<span><sup>4, 5</sup></span> This explains why the constitutive expression of HOXA9 is such a strong cancer driver. In normal cells, the production of HOXA9 and its binding partners is extinguished during differentiation.</p><p>These properties make HOXA9 an attractive target for pharmacological intervention. Unfortunately, transcription factors are notoriously hard to target with pharmaceutically applicable substances.<span><sup>6</sup></span> The most advanced attempts to derail the HOX network relies on an indirect approach by blocking the function of menin.<span><sup>7</sup></span> Menin is the product of the gene <i>multiple endocrine neoplasia</i> and it physically binds to the histone methyltransferase KMT2A and its fusion derivatives. This association is necessary for the proper localization on chromatin. Menin inhibitors disrupt the menin KMT2A interaction and show early clinical promise in KMT2A-rearranged and NPM-mutated leukemia. For unknown reasons, ho
{"title":"How to put a hex on HOX","authors":"Robert K. Slany","doi":"10.1002/hem3.83","DOIUrl":"https://doi.org/10.1002/hem3.83","url":null,"abstract":"<p>HOX-homeobox transcription factors are best known for their prominent role in embryogenesis where they control body segment identity. This regulatory principle has been “recycled” in adult tissues. HOX proteins frequently regulate the differentiation of tissue stem cells and aberrant HOX function can induce derailed maturation and tumorigenesis. A paradigm for this principle is hematopoiesis where HOXA9, a member of the so-called abdominal HOX proteins, has acquired a notorious reputation for its frequent involvement in leukemogenesis. A number of recurrent genomic aberrations in acute leukemia are associated with elevated HOXA9 expression. Examples are KMT2A (MLL) and NUP98 fusion proteins as well as the very common NPM mutations, which all induce HOXA9 overproduction. Besides, a sizable portion of acute myeloid leukemia with normal karyotype shows abnormally high levels of HOXA9. Overall, HOXA9 dysregulation can be observed in more than 50% of all cases of myeloid leukemia, and generally, this is associated with a negative prognosis (Figure 1).<span><sup>1</sup></span></p><p>At a molecular level, HOXA9 acts as a so-called pioneer transcription factor that binds to a variety of AT-rich binding sites that mark prototypical enhancers and promoters important for hematopoietic precursor cells.<span><sup>2</sup></span> On a subset of these sequences, HOXA9 assembles a trimeric complex with two other homeodomain transcription factors MEIS1 and PBX3, which contribute further DNA binding specificity. This stabilizes the trimer and in conjunction with other transcription factors leads to the establishment of specific enhancer/promoter sequences in a process that has been called enhancer sharpening.<span><sup>3</sup></span> In consequence, genes necessary for the growth, proliferation, and survival of hematopoietic precursor cells are strongly activated. Prominent examples of HOXA9/MEIS1/PBX3 targets are <i>MYB</i>, <i>MYC</i>, <i>CDK6</i>, <i>BCL2</i>, and ribosomal genes to name just a few.<span><sup>4, 5</sup></span> This explains why the constitutive expression of HOXA9 is such a strong cancer driver. In normal cells, the production of HOXA9 and its binding partners is extinguished during differentiation.</p><p>These properties make HOXA9 an attractive target for pharmacological intervention. Unfortunately, transcription factors are notoriously hard to target with pharmaceutically applicable substances.<span><sup>6</sup></span> The most advanced attempts to derail the HOX network relies on an indirect approach by blocking the function of menin.<span><sup>7</sup></span> Menin is the product of the gene <i>multiple endocrine neoplasia</i> and it physically binds to the histone methyltransferase KMT2A and its fusion derivatives. This association is necessary for the proper localization on chromatin. Menin inhibitors disrupt the menin KMT2A interaction and show early clinical promise in KMT2A-rearranged and NPM-mutated leukemia. For unknown reasons, ho","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 6","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.83","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141245670","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>When working within a haematology day unit I met patients receiving regular blood products as part of a ‘best supportive care’ approach. For some, this was because their disease had not responded to rounds of treatment; others had comorbidities or frailty that made chemotherapy inappropriate. I was often presented with a stack of blood product prescription charts to fill in for the following day. Often I had never met the patients in question, and sometimes there was no documentation about their transfusion regime, or most importantly about the aims of transfusion. For some patients having best supportive care for conditions like myelodysplastic syndrome, myelofibrosis, or aplastic anaemia, there are no prescribing guidelines, and the goal of treatment may never have been discussed. The easiest thing, especially when I was multitasking and under pressure to make quick decisions, was to repeat the previous prescription.</p><p>But as I got to know particular individuals, questions began to arise in my mind. An elderly patient called Bill* had been attending the day unit every 2 weeks for blood transfusions to relieve symptoms of anaemia, caused by myelodysplastic syndrome. Bill had tried oral chemotherapy but had suffered severe infections requiring hospitalisation as a result, and so his treatment had been stopped. Initially, his fatigue and breathlessness were managed well with transfusions every month, but within 6 months, he found the symptom relief would wane after just a week, leaving him lethargic. Accumulation of excess iron from blood transfusions added to his symptom burden. He went from walking into the day unit looking upbeat to being wheeled in looking withdrawn. I started to wonder whether his treatment made him feel better. How could we measure this? Was it right to keep giving him blood transfusions if they didn't help him feel better?</p><p>Since medical school, I have used art and comics to reflect. I have been drawn to patient stories in the realm of graphic medicine, using art and cartoons to explore illness and health narratives. These visual representations are unique reflections in that they allow me to view the artist's perspective in a way that prose does not. Creating something like a comic also allows me time to order my thoughts, to orient and contextualise experiences, and to assign meaning to them. Below is a depiction of an interaction I had with Bill**:</p><p></p><p></p><p></p><p>When I revisit and reflect on this comic, I am clear that Bill needed the time and space to explore his feelings and motivations for treatment. My suggestion of a simple binary choice—to stop or to continue transfusions—looked immature and ill-considered when Bill opened up to me. Without knowing ‘the right thing' to say I chose to sit with him until my pager inevitably went off. I felt that I let him down by not giving him an answer or a plan. Bill had clearly expressed his misery in the hours spent at the day unit, but did I have the ri
这种调整并不是一个被动的过程。反思和在变化中寻找意义的行为可以帮助个人塑造自己的故事。临床医生可以通过直接讨论以及建议写日记、写诗或绘画等创造性方式为患者创造空间。进一步的临床证据可能会为患者提供更多选择信息,但也不会给出'答案'。通过允许像比尔这样的患者与可信赖的临床医生讨论这些决定,我们挑战了 "必须继续治疗 "和 "必须在每次就诊结束后预约下一次输血 "的隐含信息。我在想,当治疗手段有限且缺乏证据时,让患者探索并塑造自己的疾病故事是否能帮助他们找到出路。索菲-埃文斯(Sophie Evans)撰写了文章初稿并绘制了图表。Stephen P. Hibbs对文章进行了严格审阅和修改。所有作者均同意最终版本。作者声明不存在利益冲突。SPH由HARP博士研究奖学金资助,该奖学金由威康信托基金会资助(资助编号为223500/Z/21/Z)。本出版物未获得任何资助。
{"title":"When to stop: Transfusions, difficult conversations and creativity","authors":"Sophie Evans, Stephen P. Hibbs","doi":"10.1002/hem3.79","DOIUrl":"10.1002/hem3.79","url":null,"abstract":"<p>When working within a haematology day unit I met patients receiving regular blood products as part of a ‘best supportive care’ approach. For some, this was because their disease had not responded to rounds of treatment; others had comorbidities or frailty that made chemotherapy inappropriate. I was often presented with a stack of blood product prescription charts to fill in for the following day. Often I had never met the patients in question, and sometimes there was no documentation about their transfusion regime, or most importantly about the aims of transfusion. For some patients having best supportive care for conditions like myelodysplastic syndrome, myelofibrosis, or aplastic anaemia, there are no prescribing guidelines, and the goal of treatment may never have been discussed. The easiest thing, especially when I was multitasking and under pressure to make quick decisions, was to repeat the previous prescription.</p><p>But as I got to know particular individuals, questions began to arise in my mind. An elderly patient called Bill* had been attending the day unit every 2 weeks for blood transfusions to relieve symptoms of anaemia, caused by myelodysplastic syndrome. Bill had tried oral chemotherapy but had suffered severe infections requiring hospitalisation as a result, and so his treatment had been stopped. Initially, his fatigue and breathlessness were managed well with transfusions every month, but within 6 months, he found the symptom relief would wane after just a week, leaving him lethargic. Accumulation of excess iron from blood transfusions added to his symptom burden. He went from walking into the day unit looking upbeat to being wheeled in looking withdrawn. I started to wonder whether his treatment made him feel better. How could we measure this? Was it right to keep giving him blood transfusions if they didn't help him feel better?</p><p>Since medical school, I have used art and comics to reflect. I have been drawn to patient stories in the realm of graphic medicine, using art and cartoons to explore illness and health narratives. These visual representations are unique reflections in that they allow me to view the artist's perspective in a way that prose does not. Creating something like a comic also allows me time to order my thoughts, to orient and contextualise experiences, and to assign meaning to them. Below is a depiction of an interaction I had with Bill**:</p><p></p><p></p><p></p><p>When I revisit and reflect on this comic, I am clear that Bill needed the time and space to explore his feelings and motivations for treatment. My suggestion of a simple binary choice—to stop or to continue transfusions—looked immature and ill-considered when Bill opened up to me. Without knowing ‘the right thing' to say I chose to sit with him until my pager inevitably went off. I felt that I let him down by not giving him an answer or a plan. Bill had clearly expressed his misery in the hours spent at the day unit, but did I have the ri","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 5","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11130759/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141160080","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}