Frustaci AM, Zappaterra A, Galitzia A, et al. Salvage treatment after covalent BTKi failure: An unmet need in clinical practice in Waldenstrom macroglobulinemia. HemaSphere. 2025;9(2):e70094. doi:10.1002/hem3.70094
In the Funding section, the funder was incorrectly listed. The funder statement now reads: “Open access publishing facilitated by Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, as part of the Wiley – SBBL agreement.”
The original article has been updated. We apologize for this error.
[这更正了文章DOI: 10.1002/hem3.70094.]。
{"title":"Correction to “Salvage treatment after covalent BTKi failure: An unmet need in clinical practice in Waldenstrom macroglobulinemia”","authors":"","doi":"10.1002/hem3.70219","DOIUrl":"10.1002/hem3.70219","url":null,"abstract":"<p>Frustaci AM, Zappaterra A, Galitzia A, et al. Salvage treatment after covalent BTKi failure: An unmet need in clinical practice in Waldenstrom macroglobulinemia. <i>HemaSphere</i>. 2025;9(2):e70094. doi:10.1002/hem3.70094</p><p>In the Funding section, the funder was incorrectly listed. The funder statement now reads: “Open access publishing facilitated by Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, as part of the Wiley – SBBL agreement.”</p><p>The original article has been updated. We apologize for this error.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12455678/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145137362","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}
Florence Broussais, Elise Pennings, Natacha Bolanos, Lorna Warwick, Robin Doeswijk, Gauthier Quinonez, Andrii Lebeda, Maria Gomes Da Silva, Sirpa Leppä, Georg Lenz, Anne-Ségolène Cottereau, Christopher Fox, Armando Lopez-Guillermo, Timothy Illidge, Wojciech Jurczak, Hans Eich, Igor Aurer, Marek Trneny, Andy Andreas Rosenwald, Andrew Davies, Ben (Gerben) Zwezerijnen, Jean-Philippe Jais, Martin Dreyling, Umberto Vitolo, Hervé Tilly, Catherine Thieblemont, Marie Jose Kersten
Large B-cell lymphoma (LBCL), including its most common subtype diffuse LBCL, is the most frequent aggressive lymphoma, accounting for 30%–40% of cases worldwide. It is a clinically heterogeneous disease, with outcomes influenced by molecular subtypes, comorbidities, and access to effective treatment. Although R-CHOP has long been the standard of care, approximately 30%–40% of patients relapse or are refractory to first-line therapy. Historically, salvage chemotherapy followed by autologous stem cell transplantation offered a potential cure, but only about half of patients are eligible, leaving a large unmet need for alternative therapies.1
Since 2018, the therapeutic landscape has rapidly evolved with the approval of nine new treatments across 12 indications in Europe, including CAR-T therapies, bispecific antibodies, antibody–drug conjugates, and targeted immunotherapies such as tafasitamab–lenalidomide. These advances have broadened the treatment arsenal beyond chemotherapy, offering potentially curative options for chemotherapy-resistant patients and effective alternatives for those ineligible for intensive approaches. This shift toward more targeted and less toxic therapies represents a significant step forward in addressing prior treatment limitations.
However, access to these innovations remains uneven across Europe. Disparities stem from fragmented national reimbursement systems, inconsistent use of early access programs (EAPs), and geographic variations in clinical trial availability. While 29 out of 35 European countries have implemented EAPs,2 differences in structure, funding, and transparency create unequal opportunities for early treatment access. Furthermore, health technology assessments (HTAs) and pricing negotiations often delay or restrict reimbursement, particularly in lower income or decentralized healthcare systems. The EU HTA Regulation (effective 2025) may harmonize clinical evaluations3-6 but will not resolve national pricing autonomy.7, 8 To achieve equitable access, stronger coordination, pricing transparency, and integration of real-world patient outcomes into decision-making are urgently needed to ensure that innovation benefits all LBCL patients, regardless of geography or system structure.
While developing the European LBCL guidelines,9 the writing committee identified substantial variability in access to innovative therapies across Europe. To assess how these disparities could impact guideline implementation, the committee conducted an analysis of clinical trial activity and national reimbursement patterns. Data were obtained from EU CTIS, ClinicalTrials.gov, and direct consultations with industry stakeholders on the reimbursement status of EMA-approved therapies. The analysis focused on two areas: (I) the geographic distribution of active LBCL trials and (II) national reimbur
{"title":"Bridging gaps in clinical trial accessibility and reimbursement for large B-cell lymphoma therapies across Europe","authors":"Florence Broussais, Elise Pennings, Natacha Bolanos, Lorna Warwick, Robin Doeswijk, Gauthier Quinonez, Andrii Lebeda, Maria Gomes Da Silva, Sirpa Leppä, Georg Lenz, Anne-Ségolène Cottereau, Christopher Fox, Armando Lopez-Guillermo, Timothy Illidge, Wojciech Jurczak, Hans Eich, Igor Aurer, Marek Trneny, Andy Andreas Rosenwald, Andrew Davies, Ben (Gerben) Zwezerijnen, Jean-Philippe Jais, Martin Dreyling, Umberto Vitolo, Hervé Tilly, Catherine Thieblemont, Marie Jose Kersten","doi":"10.1002/hem3.70204","DOIUrl":"10.1002/hem3.70204","url":null,"abstract":"<p>Large B-cell lymphoma (LBCL), including its most common subtype diffuse LBCL, is the most frequent aggressive lymphoma, accounting for 30%–40% of cases worldwide. It is a clinically heterogeneous disease, with outcomes influenced by molecular subtypes, comorbidities, and access to effective treatment. Although R-CHOP has long been the standard of care, approximately 30%–40% of patients relapse or are refractory to first-line therapy. Historically, salvage chemotherapy followed by autologous stem cell transplantation offered a potential cure, but only about half of patients are eligible, leaving a large unmet need for alternative therapies.<span><sup>1</sup></span></p><p>Since 2018, the therapeutic landscape has rapidly evolved with the approval of nine new treatments across 12 indications in Europe, including CAR-T therapies, bispecific antibodies, antibody–drug conjugates, and targeted immunotherapies such as tafasitamab–lenalidomide. These advances have broadened the treatment arsenal beyond chemotherapy, offering potentially curative options for chemotherapy-resistant patients and effective alternatives for those ineligible for intensive approaches. This shift toward more targeted and less toxic therapies represents a significant step forward in addressing prior treatment limitations.</p><p>However, access to these innovations remains uneven across Europe. Disparities stem from fragmented national reimbursement systems, inconsistent use of early access programs (EAPs), and geographic variations in clinical trial availability. While 29 out of 35 European countries have implemented EAPs,<span><sup>2</sup></span> differences in structure, funding, and transparency create unequal opportunities for early treatment access. Furthermore, health technology assessments (HTAs) and pricing negotiations often delay or restrict reimbursement, particularly in lower income or decentralized healthcare systems. The EU HTA Regulation (effective 2025) may harmonize clinical evaluations<span><sup>3-6</sup></span> but will not resolve national pricing autonomy.<span><sup>7, 8</sup></span> To achieve equitable access, stronger coordination, pricing transparency, and integration of real-world patient outcomes into decision-making are urgently needed to ensure that innovation benefits all LBCL patients, regardless of geography or system structure.</p><p>While developing the European LBCL guidelines,<span><sup>9</sup></span> the writing committee identified substantial variability in access to innovative therapies across Europe. To assess how these disparities could impact guideline implementation, the committee conducted an analysis of clinical trial activity and national reimbursement patterns. Data were obtained from EU CTIS, ClinicalTrials.gov, and direct consultations with industry stakeholders on the reimbursement status of EMA-approved therapies. The analysis focused on two areas: (I) the geographic distribution of active LBCL trials and (II) national reimbur","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12455876/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145137382","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>Sometimes the way research in science and medicine works seems far out of the public reach. Yet we know that it is carried out by ordinary people who happen to have followed a path which interests them, and which might carry the benefit of helping patients. As for most human activities, it can be a difficult world racked with ambition, competition, emotion, and deception. It can also be fascinating and rewarding. The historical career structure was made for men and it has always been difficult for women. In the end, the zigzag path to achievement depends on determination, talent, and on luck. One aspect not often mentioned is the importance of friendship between colleagues, often soured by competition. Our story is of a platonic partnership, in research into chronic lymphocytic leukemia (CLL), between two individuals, one an Italian male clinical scientist and the other a British female scientist (Figure 1). We did not work directly together, but in parallel, sharing the good things and supporting each other through the bad. The results of our efforts were to help to transform our knowledge of CLL, with insight into the biology providing the best prognostic indicator for patients and clinicians. The expansion of understanding then formed a foundation for novel targeted therapies. Our careers in translational hematology reflect the twists and turns of research during our time. Things have changed, but the excitement and fun of research remain for the next generations to enjoy.</p><p><i>Freda:</i> In my time it was difficult for a lone woman to make a difference in medical research. Not only did society load on all the expectations which go with being female, especially when there are children, but the male-dominated structures blocked progress. I look back at a career in what became “translational science,” operating at the interface between the laboratory and the clinic, beginning in the 1960s to the present. Many things have improved, with more awareness by institutions and male colleagues of problems facing women, but there are still many challenges. Hematology has always been open to science, but the power structure lies with clinicians, so results from the laboratory have to be first, comprehensible, and second, relevant to human disease.</p><p>It might seem odd now but, although I went through the usual undergraduate/postgraduate training, emerging with a DPhil from Oxford, I gave little thought to my career. The overwhelming ambition for women was to marry and have children so that is what I did. But, since my husband, George, was Australian and we moved there, I cheekily applied for a Lectureship in Biochemistry at Sydney University. In those days there was a shortage of applications and I forgot to mention that I was pregnant so I became a lecturer. This was pivotal because I was thrust into the academic structure and forced to find child care. It is obvious now that this is a way to go but I was a sort of pioneer, with no help availab
有时,科学和医学研究的工作方式似乎远远超出了公众的理解范围。然而,我们知道,它是由普通人进行的,他们碰巧走上了一条他们感兴趣的道路,这可能会带来帮助病人的好处。对于大多数人类活动来说,这可能是一个充满野心、竞争、情感和欺骗的艰难世界。它也可以是迷人和有益的。历史上的职业结构是为男性创造的,对女性来说一直很困难。最终,通往成就的曲折之路取决于决心、天赋和运气。一个不常被提及的方面是同事间友谊的重要性,而这种友谊往往会因竞争而恶化。我们的故事是一个柏拉图式的伙伴关系,在慢性淋巴细胞白血病(CLL)的研究中,两个人,一个是意大利男性临床科学家,另一个是英国女性科学家(图1)。我们并没有直接在一起工作,而是并行的,分享好的事情,在困难的时候互相支持。我们努力的结果是帮助改变我们对CLL的认识,深入了解为患者和临床医生提供最佳预后指标的生物学。认识的扩大为新的靶向治疗奠定了基础。我们在转化血液学的职业生涯反映了我们这个时代研究的曲折。事情已经变了,但研究的兴奋和乐趣仍然留给下一代去享受。弗蕾达:在我那个年代,一个孤独的女人很难在医学研究上有所作为。社会不仅给女性带来了所有的期望,尤其是当有孩子的时候,而且男性主导的结构阻碍了进步。我回顾了从20世纪60年代到现在,在实验室和诊所之间运作的“转化科学”的职业生涯。许多事情都有所改善,机构和男性同事对女性面临的问题有了更多的认识,但仍有许多挑战。血液学一直对科学开放,但权力结构掌握在临床医生手中,因此实验室的结果必须首先是可理解的,其次是与人类疾病相关的。现在听起来可能有些奇怪,但尽管我接受了常规的本科/研究生培训,获得了牛津大学的哲学博士学位,但我很少考虑自己的职业。女人最大的抱负就是结婚生子,所以我就是这么做的。但是,由于我的丈夫乔治是澳大利亚人,我们搬到了那里,我就厚颜无耻地申请了悉尼大学生物化学讲师的职位。在那些日子里,申请人数不足,我忘了说我怀孕了,所以我成了一名讲师。这是至关重要的,因为我被推进了学术结构,被迫寻找儿童保育。现在很明显,这是一条要走的路,但我是一个开拓者,没有任何帮助。1970年回到牛津,1973年搬到南安普顿,我决定继续这条路。在这个时候,追求细胞生物学研究事业的野心已经凝固,我继续着,现在有了三个孩子。出现了两个主要障碍:首先,南安普顿大学当时在基础血液学研究方面并不为人所知,这意味着由于声誉卓越,没有得到支持;第二,因为我最初是和乔治一起工作的(时间不长),所以大家都认为所有的想法都来自他。然而,这些缺点被来自卡迪夫的Tenovus慈善机构的强大财政支持所抵消,该慈善机构捐赠了南安普顿实验室,并接受拨款申请。在当前黯淡的日子里,它提醒我们,投资对于组建团队和提供新发现至关重要。即便如此,我还是花了数年的时间发表文章和参加会议,才建立了自己的声誉,这就是费德里科和其他人的支持很重要的地方。费德里科:意大利科学有不同但相似的问题。我出生在一个小山村,所以从某种意义上说,我是白手起家的。1973年我从都灵大学医学专业毕业。在那个年代,想要追求学术生涯,必须遵守三个几乎是一成不变的规则:男性、免兵役、家境富裕。除了男性,这不是我的情况,在军队服役后,我不得不接受额外的工作,比如夜班,以维持生计,同时每天在大学的临床部门工作。与病人的关系是最丰富的经验,激发了研究如何克服保护肿瘤的生物屏障,以及如何找到改善诊断和治疗的方法的热情。1981年,我在伦敦皇家自由医院免疫学部接受George Janossy的指导,获得了一段非常有启发意义的博士后经历。 与描述性观察性研究相反,我对患者调查的机械方法很着迷。我决定把这个策略带到都灵,并应用到CLL中。这让我参加了国际会议,在那里我遇到了弗雷达,她在说服我免疫学正在重塑临床学科,即血液学方面发挥了关键作用。事实证明,要想在有影响力的人的小圈子里得到我老板的支持,国际关系至关重要。我于1990年被任命为医学教授。2003年,我搬到了米兰的圣拉斐尔大学,在那里我成立了肿瘤系、肿瘤血液学和分子肿瘤学研究部门。我请Freda做我的导师,她是CLL和多发性骨髓瘤项目科学成功的关键,该项目由慈善机构意大利癌症协会(AIRC)资助,突出了微环境在b细胞肿瘤中的核心作用。对弗雷达来说,研究的第一个重点是针对淋巴瘤的DNA疫苗。她学到了一个重要的教训:尽管她开发了一种有效的融合疫苗,并认为临床试验自然会进行,但很快就发现制药公司对疫苗接种不感兴趣,他们(后来证明是错误的)对“基因疫苗”持否定态度。如果COVID有任何积极的影响,那就是改变了这种观点,她在南安普顿的学生现在正在利物浦进行类似设计的肺癌临床试验。面对这堵砖墙,她不得不另辟蹊径。另一个教训是,进步取决于新兴技术,她接受了免疫遗传学这门新科学。将这种方法应用于b细胞肿瘤是很明显的,她观察了CLL,以前是一种“无聊的小淋巴细胞堆积”。费德里科正在运用他在单克隆抗体方面新获得的经验来揭示CLL细胞的不寻常表型弗雷达研究了b细胞受体的免疫遗传学,结果证明这是一个金矿,她将这种疾病分为两组,分别来自生发前中心(GC)和生发后中心细胞,从特里·汉布林和尼古拉斯(尼克)·基奥拉兹的匹配临床数据中揭示了预后的重要差异。随后,Nick和Freda发表了发现CLL两种亚群重要性的历史,以庆祝2020年HemaSphere的20周年纪念日。欧洲血液学协会(EHA)看到了这一重要性,并邀请他们在EHA组织的会议上发表演讲,分享CLL集中治疗的令人兴奋的可能性。2014年,Freda获得了EHA颁发的Jean Bernard终身成就奖,以表彰她对血液学进步的贡献。虽然她后来获得了奖项,但这是对她的发现的宝贵的第一次认可。弗雷达和费德里科将他们的知识结合起来,对CLL的表型和基因型进行了回顾。5他们还与费德里科的一位才华横溢的研究员贝内代塔·阿波罗尼亚(Benedetta Apollonia)在南安普顿实验室分享了一个关于CLL能量本质的项目重要的是,b细胞受体被证明是抑制性药物的靶标,针对更具侵略性的亚群的新的有效疗法大量进入临床。在竞争激烈的世界里,男同事的支持是很少见的,弗雷达和费德里科的合作是幸运的。也许费德里科不是来自英国,因为英国的老男孩关系网经常与女性作对。虽然我们已经认识几年了,但在我们职业生涯相对较晚的阶段,也就是我们都50多岁的时候,我们变得更加亲密。有三种情况对巩固我们的友谊和相互尊重产生了重大影响。这些都说明了研究生活中的一些挑战和乐趣。在竞争激烈的研究领域生存需要很多优势。每一代人面临的挑战都在变化,但主要是资金压力,尤其是实施新技术的压力,造成了对有限资源的竞争。也许性别平等有所改善,但社会方面往往是一个相当被忽视的领域。国际会议过去很有挑战性,弗雷达不得不看着男人们一起去酒吧,然后出去吃晚饭,把她留在一个孤独的酒店里。她通过事先联系女性参与者并安排一起吃饭来解决这个问题。如果费德里科在会议上,她肯定会有社交接触和愉快的互动晚餐,有很多关于CLL和免疫学的补习。我们很幸运能成为“患难与共的朋友”,我们希望研究界的所有人都能找到这样的支持。Freda K. Stevenson:概念化;原创作品草案;写作-评论&编辑;验证。Federico
{"title":"Friends in need: The value of supportive colleagues in research","authors":"Freda K. Stevenson, Federico Caligaris Cappio","doi":"10.1002/hem3.70223","DOIUrl":"https://doi.org/10.1002/hem3.70223","url":null,"abstract":"<p>Sometimes the way research in science and medicine works seems far out of the public reach. Yet we know that it is carried out by ordinary people who happen to have followed a path which interests them, and which might carry the benefit of helping patients. As for most human activities, it can be a difficult world racked with ambition, competition, emotion, and deception. It can also be fascinating and rewarding. The historical career structure was made for men and it has always been difficult for women. In the end, the zigzag path to achievement depends on determination, talent, and on luck. One aspect not often mentioned is the importance of friendship between colleagues, often soured by competition. Our story is of a platonic partnership, in research into chronic lymphocytic leukemia (CLL), between two individuals, one an Italian male clinical scientist and the other a British female scientist (Figure 1). We did not work directly together, but in parallel, sharing the good things and supporting each other through the bad. The results of our efforts were to help to transform our knowledge of CLL, with insight into the biology providing the best prognostic indicator for patients and clinicians. The expansion of understanding then formed a foundation for novel targeted therapies. Our careers in translational hematology reflect the twists and turns of research during our time. Things have changed, but the excitement and fun of research remain for the next generations to enjoy.</p><p><i>Freda:</i> In my time it was difficult for a lone woman to make a difference in medical research. Not only did society load on all the expectations which go with being female, especially when there are children, but the male-dominated structures blocked progress. I look back at a career in what became “translational science,” operating at the interface between the laboratory and the clinic, beginning in the 1960s to the present. Many things have improved, with more awareness by institutions and male colleagues of problems facing women, but there are still many challenges. Hematology has always been open to science, but the power structure lies with clinicians, so results from the laboratory have to be first, comprehensible, and second, relevant to human disease.</p><p>It might seem odd now but, although I went through the usual undergraduate/postgraduate training, emerging with a DPhil from Oxford, I gave little thought to my career. The overwhelming ambition for women was to marry and have children so that is what I did. But, since my husband, George, was Australian and we moved there, I cheekily applied for a Lectureship in Biochemistry at Sydney University. In those days there was a shortage of applications and I forgot to mention that I was pregnant so I became a lecturer. This was pivotal because I was thrust into the academic structure and forced to find child care. It is obvious now that this is a way to go but I was a sort of pioneer, with no help availab","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70223","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145110923","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}
Anna M. Sowa, William Kudzi, Vivian Paintsil, Amma A. Benneh-Akwasi Kuma, Catherine I. Segbefia, Edeghonghon Olayemi, David Nana Adjei, Anastasia N. K. Bruce, Jeffrey R. Gruen, Ellis Owusu-Dabo, Solomon Fiifi Ofori-Acquah, The SickleGenAfrica Network
Sickle cell disease (SCD) is characterized by chronic hemolysis, resulting in the release of extracellular heme, which contributes to oxidative stress and inflammation. Heme oxygenase-1 (HO-1), an inducible enzyme that degrades heme into cytoprotective by-products, plays a critical role in mitigating heme-induced toxicity. This study analyzed serum HO-1 levels in 2309 individuals with SCD (53% female; median age: 12 years) from the SickleGenAfrica cohort, comprising 57% hemoglobin SS disease (Hb SS), 30% hemoglobin SC disease (Hb SC), 3.1% Hb sickle beta plus thalassemia (Sβ+ thalassemia), and 9.9% Hb S-hereditary persistence of fetal hemoglobin (Hb S-HPFH). Median HO-1 levels were threefold higher in children under 16 years (69.8 ng/mL; interquartile range [IQR]: 29.8–137.6) compared to adults (23.1 ng/mL; IQR: 7.8–62.4; P < 0.001), with peak levels observed in the 6–10-year age group. Across all subgroups, including sex, genotype, and hydroxyurea use, children consistently exhibited higher HO-1 levels than adults, with Hb SS patients showing the highest levels. Haptoglobin and hemopexin, key scavengers of hemoglobin and heme, respectively, were depleted in all patients, particularly in children. Overall, HO-1 levels in SCD patients were markedly elevated compared to healthy populations. These findings highlight the pronounced elevation of HO-1 in pediatric SCD patients, suggesting its potential protective role against heme-induced toxicity, especially during childhood.
{"title":"Elevated serum heme oxygenase-1 in pediatric sickle cell disease: Insights from the SickleGenAfrica Network","authors":"Anna M. Sowa, William Kudzi, Vivian Paintsil, Amma A. Benneh-Akwasi Kuma, Catherine I. Segbefia, Edeghonghon Olayemi, David Nana Adjei, Anastasia N. K. Bruce, Jeffrey R. Gruen, Ellis Owusu-Dabo, Solomon Fiifi Ofori-Acquah, The SickleGenAfrica Network","doi":"10.1002/hem3.70209","DOIUrl":"https://doi.org/10.1002/hem3.70209","url":null,"abstract":"<p>Sickle cell disease (SCD) is characterized by chronic hemolysis, resulting in the release of extracellular heme, which contributes to oxidative stress and inflammation. Heme oxygenase-1 (HO-1), an inducible enzyme that degrades heme into cytoprotective by-products, plays a critical role in mitigating heme-induced toxicity. This study analyzed serum HO-1 levels in 2309 individuals with SCD (53% female; median age: 12 years) from the SickleGenAfrica cohort, comprising 57% hemoglobin SS disease (Hb SS), 30% hemoglobin SC disease (Hb SC), 3.1% Hb sickle beta plus thalassemia (Sβ+ thalassemia), and 9.9% Hb S-hereditary persistence of fetal hemoglobin (Hb S-HPFH). Median HO-1 levels were threefold higher in children under 16 years (69.8 ng/mL; interquartile range [IQR]: 29.8–137.6) compared to adults (23.1 ng/mL; IQR: 7.8–62.4; P < 0.001), with peak levels observed in the 6–10-year age group. Across all subgroups, including sex, genotype, and hydroxyurea use, children consistently exhibited higher HO-1 levels than adults, with Hb SS patients showing the highest levels. Haptoglobin and hemopexin, key scavengers of hemoglobin and heme, respectively, were depleted in all patients, particularly in children. Overall, HO-1 levels in SCD patients were markedly elevated compared to healthy populations. These findings highlight the pronounced elevation of HO-1 in pediatric SCD patients, suggesting its potential protective role against heme-induced toxicity, especially during childhood.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70209","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145110924","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}
Enblad AP, Krali O, Gezelius H, et al. Ex vivo drug responses and molecular profiles of 597 pediatric acute lymphoblastic leukemia patients. HemaSphere. 2025;9(7):e70176. doi:10.1002/hem3.70176
The supplemental methods mentioned in the subsection titled “DNA methylation and RNA-sequencing data” was not included in the Supporting Information. This file has been added and cited at the applicable instance in the article.
We apologize for this error.
Enblad AP, Krali O, Gezelius H,等。597例小儿急性淋巴细胞白血病患者的体外药物反应和分子谱HemaSphere。2025; 9 (7): e70176。标题为“DNA甲基化和rna测序数据”小节中提到的补充方法未包含在支持信息中。该文件已在本文的适用实例中添加并引用。我们为这个错误道歉。
{"title":"Correction to “Ex vivo drug responses and molecular profiles of 597 pediatric acute lymphoblastic leukemia patients”","authors":"","doi":"10.1002/hem3.70221","DOIUrl":"https://doi.org/10.1002/hem3.70221","url":null,"abstract":"<p>Enblad AP, Krali O, Gezelius H, et al. Ex vivo drug responses and molecular profiles of 597 pediatric acute lymphoblastic leukemia patients. <i>HemaSphere</i>. 2025;9(7):e70176. doi:10.1002/hem3.70176</p><p>The supplemental methods mentioned in the subsection titled “DNA methylation and RNA-sequencing data” was not included in the Supporting Information. This file has been added and cited at the applicable instance in the article.</p><p>We apologize for this error.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70221","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145101753","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}
Ranran Zhang, Jasmin Straube, Yashaswini Janardhanan, Rohit Haldar, Leanne Cooper, Noah Hayes, Charles Laurore, Chulwoo J. Kim, Marlise R. Luskin, Robert C. Lindsley, Maximilian Stahl, Ann Mullally, Anna E. Marneth, Megan Bywater, Steven W. Lane
<p>Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are clonal disorders driven by mutations in hematopoietic stem cells (HSCs).<span><sup>1, 2</sup></span> <i>JAK2</i><sup>V617F</sup> is the most recurrent driver mutation in MPN and results in the constitutive activation of the JAK-STAT pathway.<span><sup>3-6</sup></span> Mutations in <i>ZRSR2</i> have been found in <i>JAK2</i><sup>V617F</sup>-driven MPNs and are associated with disease progression and poor prognosis. However, the functional consequences of mutations in <i>ZRSR2</i> in terms of disease progression in <i>JAK2</i><sup>V617F</sup>-driven MPN have not been determined. In MPN, somatic mutations in <i>ZRSR2</i> occur across the entire coding transcript, are predicted to confer a loss-of-function, and are primarily observed in male patients (Supporting Information: Figure S1).<span><sup>7-9</sup></span> Herein, using CRISPR-Cas9, we generated concomitant ZRSR2 loss in JAK2-mutated human megakaryoblastic cell lines (SET2, CHRF-288-11) and a mouse model of MPN to investigate its role in promoting disease progression. We confirmed ZRSR2 loss in both human cell lines and this mouse model by sequencing and immunoblotting (Supporting Information: Figures S2 and S3).</p><p><i>ZRSR2</i> is mainly involved in minor spliceosome assembly, with <i>ZRSR2</i> loss previously shown to result in the mis-splicing of U12-type introns in myelodysplastic syndromes (MDSs).<span><sup>10, 11</sup></span> We therefore performed replicate multivariate analysis of transcript splicing (rMATS)<span><sup>12</sup></span> to detect differential intron retention between <i>ZRSR2</i> knockout (KO) megakaryoblastic cells and controls, and identified significantly increased retained intron (RI) events false discovery rate [FDR] < .05) in SET-2 <i>ZRSR2</i> KO cell lines, but not in CHRF-288-11. However, in both megakaryoblastic cell lines, RI events tended to be more prominent in genes containing U12-type introns in the context of <i>ZRSR2</i> loss (Figure 1A). Differential gene expression analysis was performed to assess how <i>ZRSR2</i> loss impacts transcription. Gene set enrichment analysis (GSEA) showed enrichment for genes containing U12-type introns in both ZRSR2 KO megakaryoblastic cell lines versus control cells (Supporting Information: Figure S4A). We observed significant enrichment for megakaryocyte progenitors (MkPs) and E2F target gene sets, which are involved in MkPs and myeloid differentiation, in CHRF-288-11 <i>ZRSR2</i> KO cells, but not in SET-2 <i>ZRSR2</i> KO cells (Supporting Information: Figure S4B). Taken together, we conclude that <i>ZRSR2</i> loss causes U12-type intron retention and transcriptional changes in genes containing U12-type introns in human <i>JAK2</i>-mutant megakaryoblastic cell lines.</p><p><i>Zrsr2</i> loss in <i>Jak2</i><sup>V617F</sup>-driven murine MPN was achieved by isolating bone marro
骨髓增生性肿瘤(mpn),包括真性红细胞增多症(PV)、原发性血小板增多症(ET)和原发性骨髓纤维化(PMF),是由造血干细胞(hsc)突变驱动的克隆性疾病。1,2 JAK2V617F是MPN中最常见的驱动突变,可导致JAK-STAT通路的组成性激活。在jak2v617f驱动的mpn中发现了3-6个ZRSR2突变,并与疾病进展和不良预后相关。然而,就jak2v617f驱动的MPN的疾病进展而言,ZRSR2突变的功能后果尚未确定。在MPN中,ZRSR2的体细胞突变发生在整个编码转录本中,预计会导致功能丧失,并且主要在男性患者中观察到(支持信息:图S1)。在这里,我们使用CRISPR-Cas9,在jak2突变的人类巨核母细胞系(SET2, CHRF-288-11)和小鼠MPN模型中产生伴随的ZRSR2缺失,以研究其在促进疾病进展中的作用。我们通过测序和免疫印迹证实了ZRSR2在人类细胞系和小鼠模型中的缺失(支持信息:图S2和S3)。ZRSR2主要参与小剪接体组装,先前发现ZRSR2缺失会导致骨髓增生异常综合征(mds)中u12型内含子的错误剪接。10,11因此,我们对转录剪接(rMATS)进行了重复多变量分析12,以检测ZRSR2敲除(KO)巨核母细胞和对照组之间的内含子保留差异,并发现保留内含子(RI)事件的错误发现率显着增加[FDR] <;05)在SET-2 ZRSR2 KO细胞系中,但在CHRF-288-11中没有。然而,在两种巨核细胞系中,在ZRSR2缺失的情况下,RI事件往往在含有u12型内含子的基因中更为突出(图1A)。通过差异基因表达分析来评估ZRSR2缺失对转录的影响。基因集富集分析(GSEA)显示,与对照细胞相比,ZRSR2 KO巨核母细胞细胞系中含有u12型内含子的基因都富集(支持信息:图S4A)。我们观察到,在CHRF-288-11 ZRSR2 KO细胞中,参与MkPs和骨髓分化的巨核细胞祖细胞(MkPs)和E2F靶基因组显著富集,但在SET-2 ZRSR2 KO细胞中没有富集(支持信息:图S4B)。综上所述,我们得出结论,ZRSR2缺失导致人类jak2突变巨核母细胞系中u12型内含子保留和含有u12型内含子的基因的转录变化。通过从雄性供体小鼠(Jak2+/fl-V617F, Rosa26creER/lsl-Cas9-T2A-GFP)中分离骨髓(BM) Lineage−Sca-1+c-Kit+ (LSK)细胞,用表达sgRNA靶向Zrsr2 (sgZrsr2)的慢病毒载体转导它们,并移植到辐照的野生型受体小鼠中,实现了jak2v617f驱动小鼠MPN中Zrsr2的缺失。植入4周后,给小鼠喂食他莫昔芬2周,诱导CreER和Zrsr2编辑(图1B)。zrsr2编辑的受体和未编辑的对照组均出现类似于人PV的致死性MPN,伴有红细胞压比升高、脾肿大和红细胞前体扩增(图1C-E)。观察到的疾病表型与zrsr2编辑受体的性别无关(支持信息:图S5)。除了外周祖细胞(cKit+)增加外,Zrsr2缺失在外周MPN疾病参数、主要造血谱系或生存率方面没有显著差异(图1C-F,支持信息:图S6A-D)。据报道,ZRSR2突变经常发生在PMF中,PMF是一种以巨核细胞异常发育为特征的疾病状态然而,除了Jak2V617F-sgZrsr2小鼠的粒细胞/巨噬细胞祖细胞(GMPs)略有下降外,我们没有观察到在不同条件下转导供体细胞群中巨核细胞/红细胞祖细胞(MEPs)、普通髓系祖细胞(CMP)、MkPs或长期造血干细胞(lt - hsc)的比例有任何差异(图1G-I)。此外,在这些群体中,未检测到转导供体细胞的频率有差异(支持信息:图S6E),这表明Zrsr2的破坏不会在HSPC区室中与Jak2V617F结合带来竞争优势。两种情况下的组织病理学分析与PV表型一致,显示脾脏结构消失,红细胞和巨核细胞增生,BM中的巨核细胞显示非典型核特征和聚集,没有观察到Zrsr2丢失的疾病进展的证据(支持信息:图S6F)。我们进一步研究了Zrsr2破坏是否会影响BM LSK细胞中的内含子保留,并在Jak2V617F-sgZrsr2小鼠中发现了更高数量的RIs。 然而,这些RIs受类型限制较少,与人类ZRSR2 KO巨核母细胞相比,小鼠LSK细胞中含有U12剪接位点的基因较少发生(图1A,J)。此外,我们在两个物种中只发现了四个由ZRSR2调控的RIs基因(支持信息:图S7A),这表明ZRSR2缺失导致不同细胞类型和/或人与小鼠之间含有u12型内含子的基因存在不同程度的错剪接,而在小鼠细胞中这种情况明显较少。与此一致的是,与人类细胞相比,含有u12型内含子的小鼠基因的大量基因表达谱在两个方向上都没有显示出含有u12型内含子的转录本的显著富集(图1K)。然而,我们观察到,与Jak2V617F-EV细胞相比,Jak2V617F-sgZrsr2细胞中mkp相关基因集的显著富集以及与hsc中上调基因相关的转录变化(支持信息:图S7B),与进展为MF的慢性MPN患者表现出巨核细胞增生和hsc异常动员的遗传特征一致。综上所述,Zrsr2缺失在小鼠LSK细胞中引起适度的内含子保留和轻微的转录变化。然而,由于Zrsr2缺失而发生的转录变化并不足以在体内诱导jak2v617f驱动的小鼠MPN的疾病进展。我们的结果与Madan等人的报道相似,与人MDS或白血病细胞系相比,Zrsr2缺陷也导致小鼠造血细胞中观察到的u12型错误剪接事件较少在这项研究中,他们提出了Zrsr1的问题,Zrsr1是Zrsr2的同源物,在小鼠中表达而在人类中不表达,可能弥补Zrsr2缺失对小鼠RNA剪接调节的影响。在我们的小鼠模型中,Zrsr1的表达不受Zrsr2 KO的影响(支持信息:图S7C)。Zrsr1对Zrsr2在小鼠造血发育中的代偿作用可能解释了在我们的小鼠模型中观察到的与人类细胞相比,较弱的含子保留和较轻的疾病表型。然而,在含有ZRSR2突变的MPN样本中,表观遗传调控因子和剪接调控因子突变的频繁共存也可能表明,这些分子病变与ZRSR2的协同作用对于MPN的进展是必需的。15-18从一项已发表的MPN队列研究中发现,1289名JAK2V617F患者中包含12名同时存在ZRSR2突变的患者,7我们发现,与PV和ET相比,ZRSR2突变在JAK2V617F MF患者中明显更频繁(P < 0.0001, Pearson's卡方检验),频率分别为3.69%、0.28%和0.41%。在JAK2V617F和ZRSR2共突变的患者中,8例患者有一个或多个额外的突变,最常在表观遗传调节因子ASXL1和TET2中检测到(图2A)。值得注意的是,这些共同发生的突变更常与MF相关,而不是ET或PV,包括染色质修饰剂ASXL1和EZH2,或DNA甲基化调节剂TET2(图2A)。此外,与ZRSR2共突变的患者ASXL1、EZH2和TET2突变的频率明显高于未发生ZRSR2突变的患者(ASXL1: 33.3% vs. 6.81%, P = 0.0074; EZH2: 16.67% vs. 1.72%, P = 0.019; TET2: 33.3% vs.
{"title":"ZRSR2 loss causes aberrant splicing in JAK2V617F-driven myeloproliferative neoplasm but is not sufficient to drive disease progression","authors":"Ranran Zhang, Jasmin Straube, Yashaswini Janardhanan, Rohit Haldar, Leanne Cooper, Noah Hayes, Charles Laurore, Chulwoo J. Kim, Marlise R. Luskin, Robert C. Lindsley, Maximilian Stahl, Ann Mullally, Anna E. Marneth, Megan Bywater, Steven W. Lane","doi":"10.1002/hem3.70225","DOIUrl":"10.1002/hem3.70225","url":null,"abstract":"<p>Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are clonal disorders driven by mutations in hematopoietic stem cells (HSCs).<span><sup>1, 2</sup></span> <i>JAK2</i><sup>V617F</sup> is the most recurrent driver mutation in MPN and results in the constitutive activation of the JAK-STAT pathway.<span><sup>3-6</sup></span> Mutations in <i>ZRSR2</i> have been found in <i>JAK2</i><sup>V617F</sup>-driven MPNs and are associated with disease progression and poor prognosis. However, the functional consequences of mutations in <i>ZRSR2</i> in terms of disease progression in <i>JAK2</i><sup>V617F</sup>-driven MPN have not been determined. In MPN, somatic mutations in <i>ZRSR2</i> occur across the entire coding transcript, are predicted to confer a loss-of-function, and are primarily observed in male patients (Supporting Information: Figure S1).<span><sup>7-9</sup></span> Herein, using CRISPR-Cas9, we generated concomitant ZRSR2 loss in JAK2-mutated human megakaryoblastic cell lines (SET2, CHRF-288-11) and a mouse model of MPN to investigate its role in promoting disease progression. We confirmed ZRSR2 loss in both human cell lines and this mouse model by sequencing and immunoblotting (Supporting Information: Figures S2 and S3).</p><p><i>ZRSR2</i> is mainly involved in minor spliceosome assembly, with <i>ZRSR2</i> loss previously shown to result in the mis-splicing of U12-type introns in myelodysplastic syndromes (MDSs).<span><sup>10, 11</sup></span> We therefore performed replicate multivariate analysis of transcript splicing (rMATS)<span><sup>12</sup></span> to detect differential intron retention between <i>ZRSR2</i> knockout (KO) megakaryoblastic cells and controls, and identified significantly increased retained intron (RI) events false discovery rate [FDR] < .05) in SET-2 <i>ZRSR2</i> KO cell lines, but not in CHRF-288-11. However, in both megakaryoblastic cell lines, RI events tended to be more prominent in genes containing U12-type introns in the context of <i>ZRSR2</i> loss (Figure 1A). Differential gene expression analysis was performed to assess how <i>ZRSR2</i> loss impacts transcription. Gene set enrichment analysis (GSEA) showed enrichment for genes containing U12-type introns in both ZRSR2 KO megakaryoblastic cell lines versus control cells (Supporting Information: Figure S4A). We observed significant enrichment for megakaryocyte progenitors (MkPs) and E2F target gene sets, which are involved in MkPs and myeloid differentiation, in CHRF-288-11 <i>ZRSR2</i> KO cells, but not in SET-2 <i>ZRSR2</i> KO cells (Supporting Information: Figure S4B). Taken together, we conclude that <i>ZRSR2</i> loss causes U12-type intron retention and transcriptional changes in genes containing U12-type introns in human <i>JAK2</i>-mutant megakaryoblastic cell lines.</p><p><i>Zrsr2</i> loss in <i>Jak2</i><sup>V617F</sup>-driven murine MPN was achieved by isolating bone marro","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12439484/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145080287","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}
Joost B. Koedijk, Farnaz Barneh, Joyce E. Meesters-Ensing, Marc van Tuil, Edwin Sonneveld, Sander Lambo, Alicia Perzolli, Elizabeth K. Schweighart, Mauricio N. Ferrao Blanco, Merel van der Meulen, Anna Deli, Elize Haasjes, Kristina Bang Christensen, Hester A. de Groot-Kruseman, Soheil Meshinchi, Henrik Hasle, Mirjam E. Belderbos, Maaike Luesink, Bianca F. Goemans, Stefan Nierkens, Jayne Hehir-Kwa, C. Michel Zwaan, Olaf Heidenreich
T-cell-directed immunotherapy, which aims to boost or induce T-cell-mediated anti-tumor immunity, has shown remarkable success in various cancers, including B-cell precursor acute lymphoblastic leukemia (BCP-ALL), making it a compelling avenue for investigation in acute myeloid leukemia (AML).1, 2 Bispecific T-cell-engagers (TCEs) are a promising form of T-cell-directed immunotherapy that redirect CD3+ T-cells to tumor cells, thereby inducing T-cell activation and subsequent tumor cell lysis.3 However, bispecific TCEs, mainly targeting CD33 or CD123, have shown limited efficacy and/or high toxicity in relapsed/refractory AML.4-7 A proposed strategy to enhance TCE therapy in AML is their administration during periods of measurable residual disease, for example, in between chemotherapy courses, as demonstrated in BCP-ALL.2, 8-10 Chemotherapy may, however, significantly alter the immune landscape11: anthracyclines, for example, can promote anti-tumor immunity via immunogenic cell death,12 but chemotherapy may also deplete lymphocytes and induce T-cell dysfunction.13, 14 Since pre-treatment T-cell infiltration and dysfunction in the tumor microenvironment are key predictors of bispecific TCE efficacy,15-18 understanding how chemotherapy alters the immune landscape in the leukemic bone marrow (BM) is crucial for assessing the potential of TCEs in between chemotherapy courses in AML. Given differences in disease biology, immune system maturity, and treatment regimens between pediatric and adult AML,19, 20 pediatric-specific studies are necessary. Here, we examined the impact of chemotherapy-based regimens on the BM lymphocyte compartment in newly diagnosed pediatric AML (pAML).
We first characterized the treatment-naïve pAML BM lymphocyte compartment using diagnostic bulk RNA-sequencing (RNA-seq) data (Figure 1A). To reliably infer the lymphocyte composition from bulk RNA-seq data, we acquired a publicly-available single cell (sc) RNA-seq dataset21 to generate a healthy BM cell type signature matrix for use with CIBERSORTx.22 To validate its performance, we retrieved BM scRNA-seq data from 27 pAML cases at diagnosis, remission, and/or relapse,23 and generated pseudo-bulk profiles (n = 62). Applying CIBERSORTx with the healthy BM reference to these pseudo-bulk profiles and comparing the deconvoluted estimates with the original scRNA-seq annotations (Figure S1A), we observed strong correlations for T-, B-, and NK-cells (T-cells: r = 0.72, P < 0.001; B-cells: r = 0.87, P < 0.001; NK-cells: r = 0.68, P < 0.001; Figure 1B). Similarly, CD4+ naïve, CD8+ effector, and CD8+
{"title":"Bone marrow lymphocyte dynamics during chemotherapy in pediatric acute myeloid leukemia","authors":"Joost B. Koedijk, Farnaz Barneh, Joyce E. Meesters-Ensing, Marc van Tuil, Edwin Sonneveld, Sander Lambo, Alicia Perzolli, Elizabeth K. Schweighart, Mauricio N. Ferrao Blanco, Merel van der Meulen, Anna Deli, Elize Haasjes, Kristina Bang Christensen, Hester A. de Groot-Kruseman, Soheil Meshinchi, Henrik Hasle, Mirjam E. Belderbos, Maaike Luesink, Bianca F. Goemans, Stefan Nierkens, Jayne Hehir-Kwa, C. Michel Zwaan, Olaf Heidenreich","doi":"10.1002/hem3.70212","DOIUrl":"https://doi.org/10.1002/hem3.70212","url":null,"abstract":"<p>T-cell-directed immunotherapy, which aims to boost or induce T-cell-mediated anti-tumor immunity, has shown remarkable success in various cancers, including B-cell precursor acute lymphoblastic leukemia (BCP-ALL), making it a compelling avenue for investigation in acute myeloid leukemia (AML).<span><sup>1, 2</sup></span> Bispecific T-cell-engagers (TCEs) are a promising form of T-cell-directed immunotherapy that redirect CD3<sup>+</sup> T-cells to tumor cells, thereby inducing T-cell activation and subsequent tumor cell lysis.<span><sup>3</sup></span> However, bispecific TCEs, mainly targeting CD33 or CD123, have shown limited efficacy and/or high toxicity in relapsed/refractory AML.<span><sup>4-7</sup></span> A proposed strategy to enhance TCE therapy in AML is their administration during periods of measurable residual disease, for example, in between chemotherapy courses, as demonstrated in BCP-ALL.<span><sup>2, 8-10</sup></span> Chemotherapy may, however, significantly alter the immune landscape<span><sup>11</sup></span>: anthracyclines, for example, can promote anti-tumor immunity via immunogenic cell death,<span><sup>12</sup></span> but chemotherapy may also deplete lymphocytes and induce T-cell dysfunction.<span><sup>13, 14</sup></span> Since pre-treatment T-cell infiltration and dysfunction in the tumor microenvironment are key predictors of bispecific TCE efficacy,<span><sup>15-18</sup></span> understanding how chemotherapy alters the immune landscape in the leukemic bone marrow (BM) is crucial for assessing the potential of TCEs in between chemotherapy courses in AML. Given differences in disease biology, immune system maturity, and treatment regimens between pediatric and adult AML,<span><sup>19, 20</sup></span> pediatric-specific studies are necessary. Here, we examined the impact of chemotherapy-based regimens on the BM lymphocyte compartment in newly diagnosed pediatric AML (pAML).</p><p>We first characterized the treatment-naïve pAML BM lymphocyte compartment using diagnostic bulk RNA-sequencing (RNA-seq) data (Figure 1A). To reliably infer the lymphocyte composition from bulk RNA-seq data, we acquired a publicly-available single cell (sc) RNA-seq dataset<span><sup>21</sup></span> to generate a healthy BM cell type signature matrix for use with CIBERSORTx.<span><sup>22</sup></span> To validate its performance, we retrieved BM scRNA-seq data from 27 pAML cases at diagnosis, remission, and/or relapse,<span><sup>23</sup></span> and generated pseudo-bulk profiles (<i>n</i> = 62). Applying CIBERSORTx with the healthy BM reference to these pseudo-bulk profiles and comparing the deconvoluted estimates with the original scRNA-seq annotations (Figure S1A), we observed strong correlations for T-, B-, and NK-cells (T-cells: <i>r</i> = 0.72, <i>P</i> < 0.001; B-cells: <i>r</i> = 0.87, <i>P</i> < 0.001; NK-cells: <i>r</i> = 0.68, <i>P</i> < 0.001; Figure 1B). Similarly, CD4<sup>+</sup> naïve, CD8<sup>+</sup> effector, and CD8<sup>+</","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70212","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145037936","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}
Núria Martínez-Cibrián, Valentín Ortiz-Maldonado, Marta Español-Rego, Leticia Alserawan, Sergio Navarro, Nil Albiol, Miquel Lozano, Paola Charry, Laura Magnano, Andrea Rivero, Juan G. Correa, Pablo Mozas, Albert Cortés-Bullich, Carlos Jiménez-Vicente, Eva Giné, Mercedes Montoro-Lorite, Carla Ramos, Pilar Ayora, Hugo Calderón, María Sánchez-Castañón, Daniel Benítez-Ribas, Juan J. Mata-Molanes, Kelly Rojas, Xavier Setoaín, Sonia Rodríguez, Adrià Murias, Pau Alcubilla, Sara Varea, Eulalia Olesti, Mireia Bachiller, María Calvo-Orteu, Carla Sans-Pola, Joaquín Sáez-Peñataro, Carlos Fernández de Larrea, Armando López-Guillermo, E. Azucena González-Navarro, Manel Juan, Julio Delgado
We report the outcome of patients with relapsed/refractory (R/R) follicular lymphoma (FL) treated with varnimcabtagene autoleucel (var-cel), an academic anti-CD19 chimeric antigen receptor (CAR) T-cell product. Patients were included in the CART19-BE-01 clinical trial and a compassionate use program. Twenty-seven patients with FL were treated. Cytokine release syndrome (any grade) occurred in 55% of patients (4% Grade ≥3). Only 1 case (4%) of Grade 1 neurotoxicity was documented. The objective response rate was 100% at Day +100 (93% complete response rate), and the 3-year duration of response was 78%. The 3-year progression-free survival and overall survival were 78% and 81%, respectively. All patients developed B-cell aplasia, and the 3-year incidence of B-cell recovery was 17%. In conclusion, patients with R/R FL treated with var-cel obtained excellent disease control, with prolonged CAR T-cell survival and manageable toxicity. This trial was registered as NCT03144583.
{"title":"Efficacy and safety of the academic anti-CD19 chimeric antigen receptor T-cell product varnimcabtagene autoleucel for the treatment of relapsed/refractory follicular lymphoma","authors":"Núria Martínez-Cibrián, Valentín Ortiz-Maldonado, Marta Español-Rego, Leticia Alserawan, Sergio Navarro, Nil Albiol, Miquel Lozano, Paola Charry, Laura Magnano, Andrea Rivero, Juan G. Correa, Pablo Mozas, Albert Cortés-Bullich, Carlos Jiménez-Vicente, Eva Giné, Mercedes Montoro-Lorite, Carla Ramos, Pilar Ayora, Hugo Calderón, María Sánchez-Castañón, Daniel Benítez-Ribas, Juan J. Mata-Molanes, Kelly Rojas, Xavier Setoaín, Sonia Rodríguez, Adrià Murias, Pau Alcubilla, Sara Varea, Eulalia Olesti, Mireia Bachiller, María Calvo-Orteu, Carla Sans-Pola, Joaquín Sáez-Peñataro, Carlos Fernández de Larrea, Armando López-Guillermo, E. Azucena González-Navarro, Manel Juan, Julio Delgado","doi":"10.1002/hem3.70166","DOIUrl":"https://doi.org/10.1002/hem3.70166","url":null,"abstract":"<p>We report the outcome of patients with relapsed/refractory (R/R) follicular lymphoma (FL) treated with varnimcabtagene autoleucel (var-cel), an academic anti-CD19 chimeric antigen receptor (CAR) T-cell product. Patients were included in the CART19-BE-01 clinical trial and a compassionate use program. Twenty-seven patients with FL were treated. Cytokine release syndrome (any grade) occurred in 55% of patients (4% Grade ≥3). Only 1 case (4%) of Grade 1 neurotoxicity was documented. The objective response rate was 100% at Day +100 (93% complete response rate), and the 3-year duration of response was 78%. The 3-year progression-free survival and overall survival were 78% and 81%, respectively. All patients developed B-cell aplasia, and the 3-year incidence of B-cell recovery was 17%. In conclusion, patients with R/R FL treated with var-cel obtained excellent disease control, with prolonged CAR T-cell survival and manageable toxicity. This trial was registered as NCT03144583.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70166","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145037745","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}
Magdalena Klanova, Samuel Hricko, Michal Masar, Prokop Vodicka, David Salek, Natasa Kopalova, Petra Blahovcova, Daniela Kuruczova, Katerina Benesova, Jozef Michalka, Jan Koren, Pavel Klener, Andrea Janikova, Marek Trneny
Diffuse large B-cell lymphoma (DLBCL) is the most common hematological malignancy in adults. Although clinical outcomes of patients with DLBCL have improved over the past two decades, a significant proportion of DLBCL patients remain at risk of lymphoma relapse/progression or death due to relapse.1, 2 Patients with high international prognostic index (IPI) or central nervous system IPI (CNS-IPI) scores face the greatest risks of systemic and CNS relapse.3-5 The CNS-IPI model has been validated in numerous studies and is widely used in clinical practice to guide decisions regarding CNS prophylaxis.6-8 However, the effectiveness of CNS prophylaxis has been repeatedly questioned in the recent retrospective studies focusing on both intrathecal (IT) and intravenous high-dose methotrexate (HD MTX).9-14 There is a lack of robust, prospective clinical trials evaluating the impact of CNS prophylaxis on the incidence of CNS relapse. As such, CNS prophylaxis may still be considered for high-risk patients, in accordance with international guidelines (NCCN v5.2025). However, patients classified as high-risk by IPI or CNS-IPI are at risk of not only CNS relapses but also systemic relapses. The risk proportion of these two types of relapse in the same patient population has not been formally studied to date. To address this, we analyzed the competing risks of systemic versus CNS relapse in a real-world cohort of patients with DLBCL.
Using data from the prospective observational NiHiL project (NCT03199066), we identified consecutively diagnosed adult patients with histologically confirmed DLBCL not otherwise specified or DLBCL/high-grade B-cell lymphoma (HGBL) with BCL2/MYC or BCL6 rearrangements, HGBL NOS, or T-cell/histiocyte-rich large B-cell lymphoma. Patients were diagnosed between January 2010 and December 2021 at two academic centers in the Czech Republic and were treated with anti-CD20 monoclonal antibody plus CHOP chemotherapy as first-line therapy. Patients with biopsy-proven, treatment-naïve transformed indolent lymphoma (excluding Richter's transformation) were included. Patients with CNS involvement at diagnosis were excluded. Causes of death were uniformly categorized as CNS (±systemic) relapse-related, systemic relapse-related, first-line treatment toxicity-related, other, or unknown. Deaths were considered first-line treatment toxicity-related if they occurred during first-line therapy or shortly thereafter (up to Day +60 after the last treatment administration) and were attributed to treatment toxicity in the absence of progressive or refractory disease. Deaths occurring after second or subsequent lines of therapy were considered relapse-related (CNS ± systemic or systemic), unless there was clear documentation of a non-lymphoma-related cause unrelated to active disease or its treatment (e.g., unrelated comorbidity). In such cases, the cause
{"title":"Risk of central nervous system versus systemic relapse in patients with diffuse large B-cell lymphoma treated with R-CHOP: What should we focus on?","authors":"Magdalena Klanova, Samuel Hricko, Michal Masar, Prokop Vodicka, David Salek, Natasa Kopalova, Petra Blahovcova, Daniela Kuruczova, Katerina Benesova, Jozef Michalka, Jan Koren, Pavel Klener, Andrea Janikova, Marek Trneny","doi":"10.1002/hem3.70201","DOIUrl":"https://doi.org/10.1002/hem3.70201","url":null,"abstract":"<p>Diffuse large B-cell lymphoma (DLBCL) is the most common hematological malignancy in adults. Although clinical outcomes of patients with DLBCL have improved over the past two decades, a significant proportion of DLBCL patients remain at risk of lymphoma relapse/progression or death due to relapse.<span><sup>1, 2</sup></span> Patients with high international prognostic index (IPI) or central nervous system IPI (CNS-IPI) scores face the greatest risks of systemic and CNS relapse.<span><sup>3-5</sup></span> The CNS-IPI model has been validated in numerous studies and is widely used in clinical practice to guide decisions regarding CNS prophylaxis.<span><sup>6-8</sup></span> However, the effectiveness of CNS prophylaxis has been repeatedly questioned in the recent retrospective studies focusing on both intrathecal (IT) and intravenous high-dose methotrexate (HD MTX).<span><sup>9-14</sup></span> There is a lack of robust, prospective clinical trials evaluating the impact of CNS prophylaxis on the incidence of CNS relapse. As such, CNS prophylaxis may still be considered for high-risk patients, in accordance with international guidelines (NCCN v5.2025). However, patients classified as high-risk by IPI or CNS-IPI are at risk of not only CNS relapses but also systemic relapses. The risk proportion of these two types of relapse in the same patient population has not been formally studied to date. To address this, we analyzed the competing risks of systemic versus CNS relapse in a real-world cohort of patients with DLBCL.</p><p>Using data from the prospective observational NiHiL project (NCT03199066), we identified consecutively diagnosed adult patients with histologically confirmed DLBCL not otherwise specified or DLBCL/high-grade B-cell lymphoma (HGBL) with BCL2/MYC or BCL6 rearrangements, HGBL NOS, or T-cell/histiocyte-rich large B-cell lymphoma. Patients were diagnosed between January 2010 and December 2021 at two academic centers in the Czech Republic and were treated with anti-CD20 monoclonal antibody plus CHOP chemotherapy as first-line therapy. Patients with biopsy-proven, treatment-naïve transformed indolent lymphoma (excluding Richter's transformation) were included. Patients with CNS involvement at diagnosis were excluded. Causes of death were uniformly categorized as CNS (±systemic) relapse-related, systemic relapse-related, first-line treatment toxicity-related, other, or unknown. Deaths were considered first-line treatment toxicity-related if they occurred during first-line therapy or shortly thereafter (up to Day +60 after the last treatment administration) and were attributed to treatment toxicity in the absence of progressive or refractory disease. Deaths occurring after second or subsequent lines of therapy were considered relapse-related (CNS ± systemic or systemic), unless there was clear documentation of a non-lymphoma-related cause unrelated to active disease or its treatment (e.g., unrelated comorbidity). In such cases, the cause ","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70201","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145012710","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}
Corentin Orvain, Suzanne Tavitian, Clémence Mediavilla, Françoise Boyer, Alberto Santagostino, Geoffroy Venton, Samia Madene, Tony Marchand, Pascal Turlure, Diane Lara, Lenaig Le Clech, Katell Le Du, Jean-Baptiste Robin, Lise Willems, Anaïse Blouet, Thomas Systchenko, Mathieu Wemeau, Hélène Pasquer, Mélanie Mercier, Christophe Nicol, Laurence Legros, Antoine Machet, Franck-Emmanuel Nicolini, Lydia Roy, Clémentine Salvado, Guillaume Denis, Elsa Lestang, Kamel Laribi, Damien Luque Paz, Jean-Jacques Kiladjian, Eric Lippert, Lina Benajiba, Jean-Christophe Ianotto
Accelerated-phase (AP) or blast-phase (BP) myeloproliferative neoplasms (MPNs) are associated with dismal prognosis, with non-curative therapies such as hypomethylating agents (HMAs) considered in patients not eligible for intensive therapy, while some studies advocate for combination therapy with either ruxolitinib (RUXO) or venetoclax (VEN). To assess the relationship between treatment modalities and outcome, herein, we report a multicentric cohort of 149 patients (median age, 75 years) with AP/BP MPN not eligible for intensive therapy and/or allogeneic hematopoietic cell transplantation who received azacitidine (AZA) alone (n = 60) or in combination (n = 89; VEN [n = 51], RUXO [n = 27], or both [n = 9], isocitrate dehydrogenase inhibitors [n = 2]) between January 2019 and October 2023. With a median follow-up of 15 months, the median overall survival of the full cohort was 8.04 months, with a 3-year overall survival (OS) of 13%. Among disease characteristics, OS was lower in patients with BP (6.24 vs. 18.00 months in patients with AP disease, P = 0.03), complex karyotype (6.00 vs. 13.08 months, P = 0.005), and TP53 mutations (8.04 vs. 11.04 months, P = 0.009). OS was nonsignificantly higher in patients receiving AZA combinations (10.08 vs. 6.96 months in patients receiving AZA monotherapy, P = 0.12). When analyzing AZA combinations separately, patients who were treated with AZA–RUXO had higher OS (18.00 vs. 9.00 vs. 10.08 months in patients receiving AZA–VEN and AZA–VEN–RUXO, P = 0.015). The improved survival with AZA–RUXO in the absence of complex karyotype and/or TP53 mutations warrants further prospective validation. New therapeutic options are urgently needed, especially in patients with complex karyotype and/or TP53 mutations.
加速期(AP)或母细胞期(BP)骨髓增生性肿瘤(mpn)预后不佳,非根治性治疗如低甲基化药物(HMAs)被认为不适合进行强化治疗,而一些研究主张与鲁索利替尼(ruxolitinib, RUXO)或venetoclax (VEN)联合治疗。为了评估治疗方式与结果之间的关系,我们报告了一项多中心队列研究,包括149例AP/BP MPN患者(中位年龄75岁),这些患者不适合强化治疗和/或异基因造血细胞移植,他们在2019年1月至2023年10月期间接受了阿扎胞苷(AZA)单独治疗(n = 60)或联合治疗(n = 89; VEN [n = 51], RUXO [n = 27],或两者联合治疗[n = 9],异柠檬酸脱氢酶抑制剂[n = 2])。中位随访15个月,整个队列的中位总生存期为8.04个月,3年总生存期(OS)为13%。在疾病特征中,BP患者(6.24 vs 18.00个月,P = 0.03)、复杂核型患者(6.00 vs 13.08个月,P = 0.005)和TP53突变患者(8.04 vs 11.04个月,P = 0.009)的OS较低。接受AZA联合治疗的患者OS无显著性升高(10.08 vs.接受AZA单药治疗的患者6.96个月,P = 0.12)。单独分析AZA联合用药时,AZA - ruxo组患者的生存期更高(分别为18.00个月、9.00个月和10.08个月,P = 0.015)。在没有复杂核型和/或TP53突变的情况下,AZA-RUXO的生存率提高值得进一步的前瞻性验证。迫切需要新的治疗选择,特别是对于具有复杂核型和/或TP53突变的患者。
{"title":"Outcome of patients with accelerated and blast-phase myeloproliferative neoplasms not eligible for intensive chemotherapy or allogeneic hematopoietic cell transplantation treated by azacitidine alone or in combination—A FIM study","authors":"Corentin Orvain, Suzanne Tavitian, Clémence Mediavilla, Françoise Boyer, Alberto Santagostino, Geoffroy Venton, Samia Madene, Tony Marchand, Pascal Turlure, Diane Lara, Lenaig Le Clech, Katell Le Du, Jean-Baptiste Robin, Lise Willems, Anaïse Blouet, Thomas Systchenko, Mathieu Wemeau, Hélène Pasquer, Mélanie Mercier, Christophe Nicol, Laurence Legros, Antoine Machet, Franck-Emmanuel Nicolini, Lydia Roy, Clémentine Salvado, Guillaume Denis, Elsa Lestang, Kamel Laribi, Damien Luque Paz, Jean-Jacques Kiladjian, Eric Lippert, Lina Benajiba, Jean-Christophe Ianotto","doi":"10.1002/hem3.70202","DOIUrl":"https://doi.org/10.1002/hem3.70202","url":null,"abstract":"<p>Accelerated-phase (AP) or blast-phase (BP) myeloproliferative neoplasms (MPNs) are associated with dismal prognosis, with non-curative therapies such as hypomethylating agents (HMAs) considered in patients not eligible for intensive therapy, while some studies advocate for combination therapy with either ruxolitinib (RUXO) or venetoclax (VEN). To assess the relationship between treatment modalities and outcome, herein, we report a multicentric cohort of 149 patients (median age, 75 years) with AP/BP MPN not eligible for intensive therapy and/or allogeneic hematopoietic cell transplantation who received azacitidine (AZA) alone (<i>n</i> = 60) or in combination (<i>n</i> = 89; VEN [<i>n</i> = 51], RUXO [<i>n</i> = 27], or both [<i>n</i> = 9], isocitrate dehydrogenase inhibitors [<i>n</i> = 2]) between January 2019 and October 2023. With a median follow-up of 15 months, the median overall survival of the full cohort was 8.04 months, with a 3-year overall survival (OS) of 13%. Among disease characteristics, OS was lower in patients with BP (6.24 vs. 18.00 months in patients with AP disease, P = 0.03), complex karyotype (6.00 vs. 13.08 months, P = 0.005), and <i>TP53</i> mutations (8.04 vs. 11.04 months, P = 0.009). OS was nonsignificantly higher in patients receiving AZA combinations (10.08 vs. 6.96 months in patients receiving AZA monotherapy, P = 0.12). When analyzing AZA combinations separately, patients who were treated with AZA–RUXO had higher OS (18.00 vs. 9.00 vs. 10.08 months in patients receiving AZA–VEN and AZA–VEN–RUXO, P = 0.015). The improved survival with AZA–RUXO in the absence of complex karyotype and/or <i>TP53</i> mutations warrants further prospective validation. New therapeutic options are urgently needed, especially in patients with complex karyotype and/or <i>TP53</i> mutations.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 9","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70202","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144934874","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}