Pub Date : 2024-12-11DOI: 10.1038/s41375-024-02474-6
Eleni A. Argyriadi, Ingo G. Steffen, Christiane Chen-Santel, Andrej Lissat, Andishe Attarbaschi, Jean-Pierre Bourquin, Guenter Henze, Arend von Stackelberg
Relapsed Acute Lymphoblastic Leukemia (ALL) is among the most common causes of cancer-associated deaths in children. However, little is known about the implications of deviations from ALL treatment protocols on survival rates. The present study elucidates the various characteristics of treatment deviations in children with relapsed ALL included in the ALL-REZ BFM 2002 (i.e., Relapse Berlin-Frankfurt- Münster) trial and determines their prognostic relevance for relapse and death rates. Among 687 patients, 100 were identified with treatment deviations, further classified, and examined by occurrence time, cause and type. Protocol deviation was considered a time-dependent variable and its impact on Disease Free Survival (DFS) and Overall Survival (OS) was examined using the time-dependent model Mantel Byar. Five years after the relapse diagnosis, deviations were significantly related to both inferior DFS (38%) and OS (57%) rates compared to protocol conformed treatment (DFS = 61%; OS = 70%, P < 0.001). Based on multivariate analyses, protocol deviation proved to be an independent adverse prognostic factor of DFS. Moreover, deviations triggered by chemotherapy-induced toxicity were associated with a higher relapse rate compared to deviations due to insufficient response. Therefore, to avoid impairment of results by deviations, future clinical trials, and treatment strategies should focus on less toxic treatments and stricter protocol compliance.
{"title":"Prognostic relevance of treatment deviations in children with relapsed acute lymphoblastic leukemia who were treated in the ALL-REZ BFM 2002 study","authors":"Eleni A. Argyriadi, Ingo G. Steffen, Christiane Chen-Santel, Andrej Lissat, Andishe Attarbaschi, Jean-Pierre Bourquin, Guenter Henze, Arend von Stackelberg","doi":"10.1038/s41375-024-02474-6","DOIUrl":"https://doi.org/10.1038/s41375-024-02474-6","url":null,"abstract":"<p>Relapsed Acute Lymphoblastic Leukemia (ALL) is among the most common causes of cancer-associated deaths in children. However, little is known about the implications of deviations from ALL treatment protocols on survival rates. The present study elucidates the various characteristics of treatment deviations in children with relapsed ALL included in the ALL-REZ BFM 2002 (i.e., Relapse Berlin-Frankfurt- Münster) trial and determines their prognostic relevance for relapse and death rates. Among 687 patients, 100 were identified with treatment deviations, further classified, and examined by occurrence time, cause and type. Protocol deviation was considered a time-dependent variable and its impact on Disease Free Survival (DFS) and Overall Survival (OS) was examined using the time-dependent model Mantel Byar. Five years after the relapse diagnosis, deviations were significantly related to both inferior DFS (38%) and OS (57%) rates compared to protocol conformed treatment (DFS = 61%; OS = 70%, <i>P</i> < 0.001). Based on multivariate analyses, protocol deviation proved to be an independent adverse prognostic factor of DFS. Moreover, deviations triggered by chemotherapy-induced toxicity were associated with a higher relapse rate compared to deviations due to insufficient response. Therefore, to avoid impairment of results by deviations, future clinical trials, and treatment strategies should focus on less toxic treatments and stricter protocol compliance.</p>","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":"40 1","pages":""},"PeriodicalIF":11.4,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142809726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-10DOI: 10.1038/s41375-024-02483-5
Florian Simon, Othman Al-Sawaf, John F. Seymour, Barbara Eichhorst
Indolent mature B-cell neoplasms are a group of diseases in which recent therapeutic advances have led to an improved overall survival (OS) extending beyond several years. While cause of celebration for patients and caregivers, the increasingly long observation periods necessary to capture treatment effects are complicating trial design and possibly hindering swift access to more effective therapies. Surrogate endpoints are a tool with the potential of earlier study readouts, however, their validity needs to be proven in each individual disease and therapeutic setting. The validation of surrogate endpoints and available data for mature B-cell neoplasms are discussed within this perspective article, followed by an outlook on the potential of precise tools such as measurable residual disease assessment as novel surrogate candidates.
{"title":"Surrogate endpoints in mature B-cell neoplasms – meaningful or misleading?","authors":"Florian Simon, Othman Al-Sawaf, John F. Seymour, Barbara Eichhorst","doi":"10.1038/s41375-024-02483-5","DOIUrl":"10.1038/s41375-024-02483-5","url":null,"abstract":"Indolent mature B-cell neoplasms are a group of diseases in which recent therapeutic advances have led to an improved overall survival (OS) extending beyond several years. While cause of celebration for patients and caregivers, the increasingly long observation periods necessary to capture treatment effects are complicating trial design and possibly hindering swift access to more effective therapies. Surrogate endpoints are a tool with the potential of earlier study readouts, however, their validity needs to be proven in each individual disease and therapeutic setting. The validation of surrogate endpoints and available data for mature B-cell neoplasms are discussed within this perspective article, followed by an outlook on the potential of precise tools such as measurable residual disease assessment as novel surrogate candidates.","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":"39 1","pages":"25-28"},"PeriodicalIF":12.8,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41375-024-02483-5.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142796999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-10DOI: 10.1038/s41375-024-02492-4
Paul J. Hampel, Kari G. Rabe, Yucai Wang, Steven R. Hwang, Saad S. Kenderian, Eli Muchtar, Jose F. Leis, Amber B. Koehler, Mazie Tsang, Talal Hilal, Ricardo Parrondo, Rachel J. Bailen, Susan M. Schwager, Curtis A. Hanson, Esteban Braggio, Susan L. Slager, Min Shi, Cinthya J. Zepeda-Mendoza, Daniel L. Van Dyke, Tait D. Shanafelt, Rebecca L. King, Timothy G. Call, Neil E. Kay, Wei Ding, Sameer A. Parikh
Richter transformation (RT) is the histologic transformation of chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) into an aggressive lymphoma, most commonly diffuse large B-cell lymphoma (DLBCL). RT has devastating consequences with survival after transformation typically <1 year [1]. Estimates of RT frequency vary by study design, setting, and duration of follow-up. In several large studies from time periods when chemoimmunotherapy (CIT) was used to treat patients with CLL, RT rates have been reported between 2 and 10% after a median follow-up of 4–6 years from the time of diagnosis; these studies included patients with both newly diagnosed CLL and treated CLL [2,3,4,5,6]. Targeted therapies, including Bruton tyrosine kinase inhibitors (BTKi) and B-cell lymphoma 2 inhibitors (BCL2i), are now standard of care treatment for patients with CLL. High RT rates (up to 25%) reported in early studies with BTKi and BCL2i in relapsed CLL patients likely reflect increased RT risk among heavily pre-treated patients rather than risk due to these therapies themselves [7, 8]. Comparison of risk after specific treatment exposures from first-line clinical trials with CIT and targeted therapy arms is limited by the low numbers of RT events reported [9,10,11,12,13]. As the treatment paradigm for CLL has changed and CLL patients are living longer, we aimed to assess the current risk of developing RT and the potential impact of targeted therapies on that risk by comparing cohorts of patients with newly diagnosed CLL across different time periods.
Following IRB approval, we identified patients with previously untreated CLL in the Mayo Clinic CLL Database who were seen within 12 months of diagnosis. Only cases of biopsy proven DLBCL with histopathologic confirmation at Mayo Clinic were considered an RT event. Cumulative incidence methodology was used to display the time to development of RT, both from time of initial CLL diagnosis and from start of CLL-directed therapy, with death as a competing risk. We defined the period prior to February 2014 (FDA approval of ibrutinib for CLL) as the pre-targeted therapy era and the period after February 2014 as the targeted therapy era. Using Cox proportional hazards regression analysis, we compared the incidence of RT between patients diagnosed with CLL in the pre-targeted therapy era versus the targeted therapy era. Cox regression analysis was also used to investigate the association of effects of type of treatment exposure (time-dependent variable) on risk of RT. Statistical analyses were conducted using SAS 9.4 (SAS Institute, Cary, NC, USA).
Richter转化(RT)是慢性淋巴细胞白血病/小淋巴细胞淋巴瘤(CLL)向侵袭性淋巴瘤的组织学转化,最常见的是弥漫性大b细胞淋巴瘤(DLBCL)。RT对转化后的生存具有毁灭性的影响,通常为1年。RT频率的估计因研究设计、环境和随访时间而异。在使用化学免疫疗法(CIT)治疗CLL患者的几项大型研究中,在诊断后4-6年的中位随访后,RT率报道在2%至10%之间;这些研究既包括新诊断的CLL患者,也包括已治疗的CLL患者[2,3,4,5,6]。靶向治疗,包括布鲁顿酪氨酸激酶抑制剂(BTKi)和b细胞淋巴瘤2抑制剂(BCL2i),现在是CLL患者的标准护理治疗。BTKi和BCL2i在复发CLL患者中的早期研究中报道的高RT率(高达25%)可能反映了大量预处理患者RT风险的增加,而不是由于这些治疗本身的风险[7,8]。CIT和靶向治疗组一线临床试验特异性治疗暴露后的风险比较受到报道的低RT事件数量的限制[9,10,11,12,13]。由于CLL的治疗模式发生了变化,CLL患者的寿命更长,我们旨在通过比较不同时期新诊断的CLL患者队列,评估目前发展为RT的风险以及靶向治疗对该风险的潜在影响。在IRB批准后,我们在梅奥诊所CLL数据库中发现了诊断后12个月内未治疗的CLL患者。只有在梅奥诊所活检证实DLBCL并经组织病理学证实的病例才被认为是RT事件。累积发病率方法学用于显示从初始CLL诊断时间和从CLL定向治疗开始到RT发展的时间,死亡是一个竞争风险。我们将2014年2月(FDA批准ibrutinib治疗CLL)之前的时期定义为预靶向治疗时代,2014年2月之后的时期定义为靶向治疗时代。使用Cox比例风险回归分析,我们比较了靶向治疗前和靶向治疗时期诊断为CLL患者的RT发生率。还采用Cox回归分析来调查治疗暴露类型(时间相关变量)对rt风险的影响。使用SAS 9.4 (SAS Institute, Cary, NC, USA)进行统计分析。
{"title":"Incidence of Richter transformation of chronic lymphocytic leukemia/small lymphocytic lymphoma in the targeted therapy era","authors":"Paul J. Hampel, Kari G. Rabe, Yucai Wang, Steven R. Hwang, Saad S. Kenderian, Eli Muchtar, Jose F. Leis, Amber B. Koehler, Mazie Tsang, Talal Hilal, Ricardo Parrondo, Rachel J. Bailen, Susan M. Schwager, Curtis A. Hanson, Esteban Braggio, Susan L. Slager, Min Shi, Cinthya J. Zepeda-Mendoza, Daniel L. Van Dyke, Tait D. Shanafelt, Rebecca L. King, Timothy G. Call, Neil E. Kay, Wei Ding, Sameer A. Parikh","doi":"10.1038/s41375-024-02492-4","DOIUrl":"https://doi.org/10.1038/s41375-024-02492-4","url":null,"abstract":"<p>Richter transformation (RT) is the histologic transformation of chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) into an aggressive lymphoma, most commonly diffuse large B-cell lymphoma (DLBCL). RT has devastating consequences with survival after transformation typically <1 year [1]. Estimates of RT frequency vary by study design, setting, and duration of follow-up. In several large studies from time periods when chemoimmunotherapy (CIT) was used to treat patients with CLL, RT rates have been reported between 2 and 10% after a median follow-up of 4–6 years from the time of diagnosis; these studies included patients with both newly diagnosed CLL and treated CLL [2,3,4,5,6]. Targeted therapies, including Bruton tyrosine kinase inhibitors (BTKi) and B-cell lymphoma 2 inhibitors (BCL2i), are now standard of care treatment for patients with CLL. High RT rates (up to 25%) reported in early studies with BTKi and BCL2i in relapsed CLL patients likely reflect increased RT risk among heavily pre-treated patients rather than risk due to these therapies themselves [7, 8]. Comparison of risk after specific treatment exposures from first-line clinical trials with CIT and targeted therapy arms is limited by the low numbers of RT events reported [9,10,11,12,13]. As the treatment paradigm for CLL has changed and CLL patients are living longer, we aimed to assess the current risk of developing RT and the potential impact of targeted therapies on that risk by comparing cohorts of patients with newly diagnosed CLL across different time periods.</p><p>Following IRB approval, we identified patients with previously untreated CLL in the Mayo Clinic CLL Database who were seen within 12 months of diagnosis. Only cases of biopsy proven DLBCL with histopathologic confirmation at Mayo Clinic were considered an RT event. Cumulative incidence methodology was used to display the time to development of RT, both from time of initial CLL diagnosis and from start of CLL-directed therapy, with death as a competing risk. We defined the period prior to February 2014 (FDA approval of ibrutinib for CLL) as the pre-targeted therapy era and the period after February 2014 as the targeted therapy era. Using Cox proportional hazards regression analysis, we compared the incidence of RT between patients diagnosed with CLL in the pre-targeted therapy era versus the targeted therapy era. Cox regression analysis was also used to investigate the association of effects of type of treatment exposure (time-dependent variable) on risk of RT. Statistical analyses were conducted using SAS 9.4 (SAS Institute, Cary, NC, USA).</p>","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":"93 1","pages":""},"PeriodicalIF":11.4,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142796997","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>The t(16;21)(p11;q22) translocation is a rare but non-random chromosomal abnormality that accounts for approximately 1% of cases of acute myeloid leukemia (AML) [1, 2]. This chromosomal translocation produces a fusion gene between the <i>FUS</i> gene at chromosome 16p11 and the <i>ERG</i> gene at chromosome 21q22. Recent cytogenetic stratification classifies the sole abnormality of t(16;21)(p11;q22) as intermediate risk [1]. However, previous reports [2,3,4] suggest that the prognosis for AML patients with t(16;21)(p11;q22) is generally significantly worse, and evidence regarding allogeneic hematopoietic stem cell transplantation (HSCT) for AML patients with t(16;21)(p11;q22) is still lacking. Therefore, this study evaluated the survival outcomes of AML with t(16;21)(p11;q22) after allogeneic HSCT and compared them with those of other AML patients using Japanese transplant registry data.</p><p>The clinical data were provided by the Transplant Registry Unified Management Program 2 of the Japan Society for Transplantation and the Cellular Therapy and the Japanese Data Center for Hematopoietic Cell Transplantation [5,6,7]. Our study included patients with AML aged ≥16 years who received their first allogeneic HSCT between 1986 and 2021. Patients with a favorable-risk karyotype were excluded, as t(16;21)(p11;q22) is not classified as favorable according to the karyotype risk classification. This study was approved by the Data Management Committee of the Japanese Data Center for Hematopoietic Cell Transplantation and the Ethics Committee of Kobe City Medical Center General Hospital. Cytogenetic information was obtained at diagnosis or before allogeneic HSCT. The cytogenetic risk was classified as favorable, intermediate, or poor in accordance with the criteria provided by the National Comprehensive Cancer Network Guidelines (Version 1.2016) [8]. Due to insufficient data, in some cases, the t(16;21) translocation was identified, but their breakpoints were unknown (t(16;21) BU [breakpoint unknown]). Chromosomal translocations involving chromosomes 16 and 21 seen in AML include t(16;21)(q24;q22), as well as t(16;21)(p11;q22) [9]. However, t(16;21)(q24;q22) is notably rare in adult AML [3, 4, 9]; therefore, we considered t(16;21) BU as t(16;21)(p11;q22) in our analysis. Additionally, we performed a sensitivity analysis comparing patient characteristics and survival data between the t(16;21) BU and t(16;21)(p11;q22) groups to identify potential misclassifications. Other detailed definitions of variables and endpoints are described in Supplementary materials. The study endpoints were progression-free survival (PFS), overall survival (OS), relapse, and non-relapse mortality (NRM). For survival analysis, the Kaplan–Meier method was used for OS and PFS, while the Gray’s method was used for relapse and NRM to account for competing risks. Multivariable analysis was performed using a Cox proportional hazard model for OS and PFS or a Fine and Gray proportional
t(16;21)(p11;q22)易位是一种罕见但非随机的染色体异常,约占急性髓性白血病(AML)病例的1%[1,2]。这种染色体易位在16p11染色体上的FUS基因和21q22染色体上的ERG基因之间产生融合基因。最近的细胞遗传学分层将t(16;21)(p11;q22)唯一的异常归为中危[1]。然而,以往的报道[2,3,4]表明,合并t(16;21)(p11;q22)的AML患者的预后通常明显较差,并且关于同种异体造血干细胞移植(HSCT)治疗合并t(16;21)(p11;q22)的证据仍然缺乏。因此,本研究评估了同种异体造血干细胞移植后AML伴t(16;21)(p11;q22)的生存结果,并使用日本移植登记数据将其与其他AML患者的生存结果进行了比较。临床数据由日本移植和细胞治疗学会移植登记统一管理计划2和日本造血细胞移植数据中心提供[5,6,7]。我们的研究纳入了年龄≥16岁的AML患者,这些患者在1986年至2021年间接受了第一次同种异体造血干细胞移植。由于t(16;21)(p11;q22)未按核型风险分类为有利,因此排除了有利核型的患者。本研究经日本造血细胞移植数据中心数据管理委员会和神户市医疗中心总医院伦理委员会批准。在诊断时或同种异体造血干细胞移植前获得细胞遗传学信息。根据美国国家综合癌症网络指南(Version 1.2016)[8]提供的标准,细胞遗传学风险分为有利、中等或较差。由于数据不足,在某些情况下,虽然发现了t(16;21)易位,但断点未知(t(16;21) BU[断点未知])。AML中涉及16号和21号染色体的染色体易位包括t(16;21)(q24;q22),以及t(16;21)(p11;q22)[9]。然而,t(16;21)(q24;q22)在成人AML中非常罕见[3,4,9];因此,我们认为t(16;21) BU在我们的分析中为t(16;21)(p11;q22)。此外,我们进行了敏感性分析,比较t(16;21) BU组和t(16;21)(p11;q22)组之间的患者特征和生存数据,以识别潜在的错误分类。其他变量和端点的详细定义在补充材料中描述。研究终点为无进展生存期(PFS)、总生存期(OS)、复发和非复发死亡率(NRM)。对于生存分析,Kaplan-Meier法用于OS和PFS,而Gray法用于复发和NRM,以解释竞争风险。多变量分析采用OS和PFS的Cox比例风险模型或复发和NRM的Fine和Gray比例风险模型。采用Fine and Gray方法来考虑竞争风险。P <; 0.05为统计学意义。所有统计分析均采用R软件(版本4.3.2;R开发核心团队)。
{"title":"Allogeneic hematopoietic stem cell transplantation in adult acute myeloid leukemia with t(16;21)(p11;q22)/FUS::ERG","authors":"Satoshi Mitsuyuki, Yoshimitsu Shimomura, Hiroki Mizumaki, Masamitsu Yanada, Shohei Mizuno, Naoyuki Uchida, Noriko Doki, Ayumu Ito, Masatsugu Tanaka, Tetsuya Nishida, Yuta Katayama, Satoshi Yoshihara, Tetsuya Eto, Satoru Takada, Shuichi Ota, Masako Toyosaki, Yuta Hasegawa, Hirohisa Nakamae, Koji Kawamura, Makoto Onizuka, Takahiro Fukuda, Marie Ohbiki, Yoshiko Atsuta, Takaaki Konuma","doi":"10.1038/s41375-024-02495-1","DOIUrl":"https://doi.org/10.1038/s41375-024-02495-1","url":null,"abstract":"<p>The t(16;21)(p11;q22) translocation is a rare but non-random chromosomal abnormality that accounts for approximately 1% of cases of acute myeloid leukemia (AML) [1, 2]. This chromosomal translocation produces a fusion gene between the <i>FUS</i> gene at chromosome 16p11 and the <i>ERG</i> gene at chromosome 21q22. Recent cytogenetic stratification classifies the sole abnormality of t(16;21)(p11;q22) as intermediate risk [1]. However, previous reports [2,3,4] suggest that the prognosis for AML patients with t(16;21)(p11;q22) is generally significantly worse, and evidence regarding allogeneic hematopoietic stem cell transplantation (HSCT) for AML patients with t(16;21)(p11;q22) is still lacking. Therefore, this study evaluated the survival outcomes of AML with t(16;21)(p11;q22) after allogeneic HSCT and compared them with those of other AML patients using Japanese transplant registry data.</p><p>The clinical data were provided by the Transplant Registry Unified Management Program 2 of the Japan Society for Transplantation and the Cellular Therapy and the Japanese Data Center for Hematopoietic Cell Transplantation [5,6,7]. Our study included patients with AML aged ≥16 years who received their first allogeneic HSCT between 1986 and 2021. Patients with a favorable-risk karyotype were excluded, as t(16;21)(p11;q22) is not classified as favorable according to the karyotype risk classification. This study was approved by the Data Management Committee of the Japanese Data Center for Hematopoietic Cell Transplantation and the Ethics Committee of Kobe City Medical Center General Hospital. Cytogenetic information was obtained at diagnosis or before allogeneic HSCT. The cytogenetic risk was classified as favorable, intermediate, or poor in accordance with the criteria provided by the National Comprehensive Cancer Network Guidelines (Version 1.2016) [8]. Due to insufficient data, in some cases, the t(16;21) translocation was identified, but their breakpoints were unknown (t(16;21) BU [breakpoint unknown]). Chromosomal translocations involving chromosomes 16 and 21 seen in AML include t(16;21)(q24;q22), as well as t(16;21)(p11;q22) [9]. However, t(16;21)(q24;q22) is notably rare in adult AML [3, 4, 9]; therefore, we considered t(16;21) BU as t(16;21)(p11;q22) in our analysis. Additionally, we performed a sensitivity analysis comparing patient characteristics and survival data between the t(16;21) BU and t(16;21)(p11;q22) groups to identify potential misclassifications. Other detailed definitions of variables and endpoints are described in Supplementary materials. The study endpoints were progression-free survival (PFS), overall survival (OS), relapse, and non-relapse mortality (NRM). For survival analysis, the Kaplan–Meier method was used for OS and PFS, while the Gray’s method was used for relapse and NRM to account for competing risks. Multivariable analysis was performed using a Cox proportional hazard model for OS and PFS or a Fine and Gray proportional ","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":"27 1","pages":""},"PeriodicalIF":11.4,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142796998","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-04DOI: 10.1038/s41375-024-02486-2
Sen Yang, Xiaoshuai Zhang, Robert Peter Gale, Xiaojun Huang, Qian Jiang
Whether there is really a distinct accelerated phase (AP) at diagnosis in chronic myeloid leukaemia (CML) in the context of tyrosine kinase-inhibitor (TKI)-therapy is controversial. We studied 2122 consecutive subjects in chronic phase (CP, n = 1837) or AP (n = 285) at diagnosis classified according to the 2020 European LeukemiaNet (ELN) classification. AP subjects with increased basophils only had similar transformation-free survival (TFS) and survival compared with CP subjects classified as ELTS intermediate-risk. Those with increased blasts only had worse TFS but similar survival compared with CP subjects classified as ELTS high-risk. AP subjects with decreased platelets only had similar TFS but worse survival compared with subjects classified as ELTS high-risk. Proportions of CP and AP subjects meeting the 2020 ELN TKI-response milestones were similar. However, worse TFS at 3-month and survival at 6- or 12-month were only in AP subjects failing to meet ELN milestones. Findings were similar using the 2022 International Consensus Classification (ICC) criteria for AP replacing decreased platelets with additional cytogenetic abnormalities. Our data support the 2022 WHO classification of CML eliminating AP. We suggest adding a very high-risk cohort to the ELTS score including people with increased blasts or decreased platelets and dividing CML into 2 phases at diagnosis: CP and acute or blast phases.
在酪氨酸激酶抑制剂(TKI)治疗的背景下,慢性髓性白血病(CML)的诊断是否真的存在明显的加速期(AP)是有争议的。我们研究了2122名连续受试者,根据2020年欧洲白血病网(ELN)分类,诊断为慢年期(CP, n = 1837)或AP (n = 285)。嗜碱性粒细胞增加的AP受试者与被归类为elt中风险的CP受试者相比,只有相似的无转化生存期(TFS)和生存期。与被归类为ELTS高风险的CP受试者相比,那些爆炸增加的患者只有更差的TFS,但生存率相似。血小板减少的AP患者与ELTS高危患者相比,TFS相似,但生存率更差。CP和AP受试者达到2020年ELN tki反应里程碑的比例相似。然而,只有未达到ELN里程碑的AP受试者在3个月时的TFS和6或12个月时的生存率较差。使用2022年国际共识分类(ICC)标准,AP取代血小板减少并伴有额外的细胞遗传学异常,结果相似。我们的数据支持WHO对2022年排除AP的CML的分类。我们建议在ELTS评分中加入一个非常高风险的队列,包括血小板增加或血小板减少的患者,并在诊断时将CML分为2期:CP期和急性期或blast期。
{"title":"Is there really an accelerated phase of chronic myeloid leukaemia at presentation?","authors":"Sen Yang, Xiaoshuai Zhang, Robert Peter Gale, Xiaojun Huang, Qian Jiang","doi":"10.1038/s41375-024-02486-2","DOIUrl":"https://doi.org/10.1038/s41375-024-02486-2","url":null,"abstract":"<p>Whether there is really a distinct accelerated phase (AP) at diagnosis in chronic myeloid leukaemia (CML) in the context of tyrosine kinase-inhibitor (TKI)-therapy is controversial. We studied 2122 consecutive subjects in chronic phase (CP, <i>n</i> = 1837) or AP (<i>n</i> = 285) at diagnosis classified according to the 2020 European LeukemiaNet (ELN) classification. AP subjects with increased basophils only had similar transformation-free survival (TFS) and survival compared with CP subjects classified as ELTS intermediate-risk. Those with increased blasts only had worse TFS but similar survival compared with CP subjects classified as ELTS high-risk. AP subjects with decreased platelets only had similar TFS but worse survival compared with subjects classified as ELTS high-risk. Proportions of CP and AP subjects meeting the 2020 ELN TKI-response milestones were similar. However, worse TFS at 3-month and survival at 6- or 12-month were only in AP subjects failing to meet ELN milestones. Findings were similar using the 2022 International Consensus Classification (ICC) criteria for AP replacing decreased platelets with additional cytogenetic abnormalities. Our data support the 2022 WHO classification of CML eliminating AP. We suggest adding a very high-risk cohort to the ELTS score including people with increased blasts or decreased platelets and dividing CML into 2 phases at diagnosis: CP and acute or blast phases.</p>","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":"14 1","pages":""},"PeriodicalIF":11.4,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142763190","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-04DOI: 10.1038/s41375-024-02485-3
Saioa Arza-Apalategi, Branco M. H. Heuts, Saskia M. Bergevoet, Roos Meering, Daan Gilissen, Pascal W. T. C. Jansen, Anja Krippner-Heidenreich, Peter J. M. Valk, Michiel Vermeulen, Olaf Heidenreich, Torsten Haferlach, Joop H. Jansen, Joost H. A. Martens, Bert A. van der Reijden
KMT2A::MLLT3 acute myelomonocytic leukemia (AML) comes in two clinically and biologically different subtypes. One is characterized by inferior outcome, older age, and MECOM oncogene expression. The other is mainly observed in children and young adults, associates with better clinical outcome, but lacks MECOM. To identify cell fate determining transcription factors downstream of KMT2A::MLLT3, we applied a bioinformatic algorithm that integrates gene and enhancer expression from primary MECOM-positive and -negative KMT2A::MLLT3 AML samples. This identified MECOM to be most influential in the MECOM-positive group, while neuronal transcription factor HMX3 was most influential in the MECOM-negative group. In large AML cohorts, HMX3 expression associated with a unique gene expression profile, younger age (p < 0.002) and KMT2A-rearranged and KAT6A-CREBBP leukemia (p < 0.00001). HMX3 was not expressed in other major genetic risk groups and healthy blood cells. RNA-sequencing analyses following forced HMX3 expression in healthy CD34+ cells and its silencing in KMT2A::MLT3 cells showed that HMX3 drives cancer-associated E2F and MYC gene programs (p < 0.001). HMX3 expression in healthy CD34+ cells blocked monocytic but not granulocytic colony formation. Strikingly, HMX3 silencing in KMT2A::MLLT3 patient cells resulted in cell cycle arrest, monocytic differentiation and apoptosis. Thus, the neuronal transcription factor HMX3 is a leukemia-specific vulnerability in KMT2A::MLLT3 AML.
{"title":"HMX3 is a critical vulnerability in MECOM-negative KMT2A::MLLT3 acute myelomonocytic leukemia","authors":"Saioa Arza-Apalategi, Branco M. H. Heuts, Saskia M. Bergevoet, Roos Meering, Daan Gilissen, Pascal W. T. C. Jansen, Anja Krippner-Heidenreich, Peter J. M. Valk, Michiel Vermeulen, Olaf Heidenreich, Torsten Haferlach, Joop H. Jansen, Joost H. A. Martens, Bert A. van der Reijden","doi":"10.1038/s41375-024-02485-3","DOIUrl":"https://doi.org/10.1038/s41375-024-02485-3","url":null,"abstract":"<p>KMT2A::MLLT3 acute myelomonocytic leukemia (AML) comes in two clinically and biologically different subtypes. One is characterized by inferior outcome, older age, and MECOM oncogene expression. The other is mainly observed in children and young adults, associates with better clinical outcome, but lacks MECOM. To identify cell fate determining transcription factors downstream of KMT2A::MLLT3, we applied a bioinformatic algorithm that integrates gene and enhancer expression from primary MECOM-positive and -negative KMT2A::MLLT3 AML samples. This identified MECOM to be most influential in the MECOM-positive group, while neuronal transcription factor HMX3 was most influential in the MECOM-negative group. In large AML cohorts, HMX3 expression associated with a unique gene expression profile, younger age (<i>p</i> < 0.002) and KMT2A-rearranged and KAT6A-CREBBP leukemia (<i>p</i> < 0.00001). HMX3 was not expressed in other major genetic risk groups and healthy blood cells. RNA-sequencing analyses following forced HMX3 expression in healthy CD34+ cells and its silencing in KMT2A::MLT3 cells showed that HMX3 drives cancer-associated E2F and MYC gene programs (<i>p</i> < 0.001). HMX3 expression in healthy CD34+ cells blocked monocytic but not granulocytic colony formation. Strikingly, HMX3 silencing in KMT2A::MLLT3 patient cells resulted in cell cycle arrest, monocytic differentiation and apoptosis. Thus, the neuronal transcription factor HMX3 is a leukemia-specific vulnerability in KMT2A::MLLT3 AML.</p>","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":"26 1","pages":""},"PeriodicalIF":11.4,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142763191","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-03DOI: 10.1038/s41375-024-02487-1
Jeff P Sharman, Miklos Egyed, Wojciech Jurczak, Alan Skarbnik, John M Pagel, Ian W Flinn, Manali Kamdar, Talha Munir, Renata Walewska, Gillian Corbett, Laura Maria Fogliatto, Yair Herishanu, Versha Banerji, Steven Coutre, George Follows, Patricia Walker, Karin Karlsson, Paolo Ghia, Ann Janssens, Florence Cymbalista, Jennifer A Woyach, Emmanuelle Ferrant, William G Wierda, Veerendra Munugalavadla, Ting Yu, Min Hui Wang, John C Byrd
{"title":"Correction: Efficacy and safety in a 4-year follow-up of the ELEVATE-TN study comparing acalabrutinib with or without obinutuzumab versus obinutuzumab plus chlorambucil in treatment-naïve chronic lymphocytic leukemia.","authors":"Jeff P Sharman, Miklos Egyed, Wojciech Jurczak, Alan Skarbnik, John M Pagel, Ian W Flinn, Manali Kamdar, Talha Munir, Renata Walewska, Gillian Corbett, Laura Maria Fogliatto, Yair Herishanu, Versha Banerji, Steven Coutre, George Follows, Patricia Walker, Karin Karlsson, Paolo Ghia, Ann Janssens, Florence Cymbalista, Jennifer A Woyach, Emmanuelle Ferrant, William G Wierda, Veerendra Munugalavadla, Ting Yu, Min Hui Wang, John C Byrd","doi":"10.1038/s41375-024-02487-1","DOIUrl":"https://doi.org/10.1038/s41375-024-02487-1","url":null,"abstract":"","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142770615","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-28DOI: 10.1038/s41375-024-02480-8
Rosalin A Cooper, Emily Thomas, Anna M Sozanska, Carlo Pescia, Daniel J Royston
{"title":"Spatial transcriptomic approaches for characterising the bone marrow landscape: pitfalls and potential.","authors":"Rosalin A Cooper, Emily Thomas, Anna M Sozanska, Carlo Pescia, Daniel J Royston","doi":"10.1038/s41375-024-02480-8","DOIUrl":"https://doi.org/10.1038/s41375-024-02480-8","url":null,"abstract":"","PeriodicalId":18109,"journal":{"name":"Leukemia","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142751322","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}