Dimitrios Farmakis, Michael Angastiniotis, Androulla Eleftheriou
<p>The 16th International Conference on Thalassemia & Hemoglobinopathies was held together with the 18th International Conference for Patients and Parents in Kuala Lumpur, Malaysia, on November 3–5, 2023. Both congresses were organized by the Thalassaemia International Federation (TIF), a patient-oriented, nonprofit, nongovernmental umbrella federation with 231 member associations from 69 countries. TIF works in official relations with the World Health Organization (WHO), the European Council, and the United Nations Economic and Social Council to promote access to quality care for all patients with thalassemia or other hemoglobinopathies worldwide.</p><p>The scientific program of the conference addressed a broad range of topics concerning thalassemia, sickle cell disease (SCD), and other rare anemias, providing a comprehensive review of the current state of the art, recent advances, and persisting or emerging challenges on diagnosis, management, and prevention of hemoglobinopathies.</p><p>Impaired maturation of erythroid progenitors to red cells in bone marrow is a key component of β-thalassemia pathophysiology. Luspatercept is a novel drug that promotes erythroid maturation, improving anemia and reducing transfusional requirements. Its efficacy and safety have been documented in transfusion-dependent thalassemia (TDT) and nontransfusion-dependent thalassemia (NTDT) by the randomized trials BELIEVE and BEYOND, respectively,<span><sup>1, 2</sup></span> resulting in the approval of the drug for TDT in 2020 and for either TDT or NTDT in 2023. Open issues include the efficacy and safety of luspatercept in children, patients with alpha-thalassemia, and in combination with other drugs and the identification of predictors of response and side effects. The high cost of the drug poses a challenge to healthcare systems and creates the need for proper patient selection.</p><p>Mitapivat is an oral small-molecule activator of red cell-specific pyruvate kinase (PK). PK is crucial for the energetic supply, function, and survival of red cells, which are compromised in patients with PK deficiency, thalassemia, and SCD. A series of clinical trials have addressed the efficacy and safety of mitapivat in these three hemolytic conditions. Two phase 2 studies in NTDT and SCD, respectively, documented improvements in hemoglobin levels, hemolysis, and sickling, with adequate safety.<span><sup>3, 4</sup></span> Two phase 3 trials in TDT and NTDT and a larger phase 2/3 study in SCD are ongoing.</p><p>Drugs and interventions promoting hemoglobin F (HbF) synthesis have been tested as potential therapies for β-thalassemia. BCL11A is a transcription factor inhibiting HbF synthesis and its genetic manipulation with the gene-editing technique CRISPR-Cas9, followed by autologous stem cell transplantation with BCL11A-edited cells, is being evaluated in β-thalassemia and SCD.<span><sup>5</sup></span></p><p>The TMPRSS6 antisense oligonucleotide (ASO) inhibits the expression of T
{"title":"Exciting science from the 16th International Conference on Thalassemia and Hemoglobinopathies","authors":"Dimitrios Farmakis, Michael Angastiniotis, Androulla Eleftheriou","doi":"10.1002/hem3.114","DOIUrl":"https://doi.org/10.1002/hem3.114","url":null,"abstract":"<p>The 16th International Conference on Thalassemia & Hemoglobinopathies was held together with the 18th International Conference for Patients and Parents in Kuala Lumpur, Malaysia, on November 3–5, 2023. Both congresses were organized by the Thalassaemia International Federation (TIF), a patient-oriented, nonprofit, nongovernmental umbrella federation with 231 member associations from 69 countries. TIF works in official relations with the World Health Organization (WHO), the European Council, and the United Nations Economic and Social Council to promote access to quality care for all patients with thalassemia or other hemoglobinopathies worldwide.</p><p>The scientific program of the conference addressed a broad range of topics concerning thalassemia, sickle cell disease (SCD), and other rare anemias, providing a comprehensive review of the current state of the art, recent advances, and persisting or emerging challenges on diagnosis, management, and prevention of hemoglobinopathies.</p><p>Impaired maturation of erythroid progenitors to red cells in bone marrow is a key component of β-thalassemia pathophysiology. Luspatercept is a novel drug that promotes erythroid maturation, improving anemia and reducing transfusional requirements. Its efficacy and safety have been documented in transfusion-dependent thalassemia (TDT) and nontransfusion-dependent thalassemia (NTDT) by the randomized trials BELIEVE and BEYOND, respectively,<span><sup>1, 2</sup></span> resulting in the approval of the drug for TDT in 2020 and for either TDT or NTDT in 2023. Open issues include the efficacy and safety of luspatercept in children, patients with alpha-thalassemia, and in combination with other drugs and the identification of predictors of response and side effects. The high cost of the drug poses a challenge to healthcare systems and creates the need for proper patient selection.</p><p>Mitapivat is an oral small-molecule activator of red cell-specific pyruvate kinase (PK). PK is crucial for the energetic supply, function, and survival of red cells, which are compromised in patients with PK deficiency, thalassemia, and SCD. A series of clinical trials have addressed the efficacy and safety of mitapivat in these three hemolytic conditions. Two phase 2 studies in NTDT and SCD, respectively, documented improvements in hemoglobin levels, hemolysis, and sickling, with adequate safety.<span><sup>3, 4</sup></span> Two phase 3 trials in TDT and NTDT and a larger phase 2/3 study in SCD are ongoing.</p><p>Drugs and interventions promoting hemoglobin F (HbF) synthesis have been tested as potential therapies for β-thalassemia. BCL11A is a transcription factor inhibiting HbF synthesis and its genetic manipulation with the gene-editing technique CRISPR-Cas9, followed by autologous stem cell transplantation with BCL11A-edited cells, is being evaluated in β-thalassemia and SCD.<span><sup>5</sup></span></p><p>The TMPRSS6 antisense oligonucleotide (ASO) inhibits the expression of T","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.114","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141730109","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>Laboratories studying hematopoietic stem cell (HSC) biology from across the world all have their own way of doing things, and this extends not just to the various cell surface marker combinations used to isolate HSCs but also to the more mundane aspects of cell isolation and preparation. At the very beginning of an HSC experiment, all researchers are faced with the question of which bones to obtain their bone marrow sample from. In the mouse, where options are more numerous, researchers commonly obtain marrow samples from the tibia, femur, hip, sternum, or spine. Some researchers then proceed to isolate the marrow by flushing, others by centrifuging bones, and still others by crushing. Yet, the vast majority of studies simply report that mouse bone marrow was isolated, and where it is specified, it is rarely looked at as a major experimental variable (e.g., a study with bone marrow from the tibia and femur is not viewed differently to one that also obtains marrow from spine and hip). Does it really matter which bone (or which part of which bone) cells are isolated from? Recent evidence suggests that the answer is a resounding “yes.”</p><p>While numerous anecdotes have circulated around various conference circuits and between highly specialized HSC labs, it is rare to see published studies that go into the fine detail of differences in the anatomical location of bone marrow blood cells. That said, studies have been cropping up in various guises over the years, and a major landmark study came out this year from Daniel Lucas' group<span><sup>1</sup></span> that added fuel to the fire and appears to cement the unique roles of different bones in blood cell production. One of the earliest studies was from Brian Lord in the 1970s where colony-forming cells were isolated from either the central marrow shaft (dubbed “axial”) or the edges of the bone (named “marginal”) and distinct numbers and types of colonies resided in each cell preparation.<span><sup>2</sup></span> This study preceded a large number of studies that focused on different types of bone and bone matrix, different proposed HSC niches, and different neighboring cell types (reviewed in Comazzetto et al.<span><sup>3</sup></span>).</p><p>Another provocative study emerged from David Bryder's lab in 2015<span><sup>4</sup></span> that challenged the notion of transplantable HSCs being equally distributed across the skeleton of a repopulated mouse. Following 16 weeks of transplantation and monitoring of overall chimerism in the blood, each of the right and left legs (tibia, femur, and hip) were assessed for donor chimerism, and the differences between bones were substantial, with some animals showing chimerism nearly exclusively in a single bone. Experimentally, this again challenges researchers to not consider that all marrow is equal irrespective of location.</p><p>Coming back to the Lucas lab study and why it has made such a big impact in the field, they undertook a skeleton-wide imaging stud
{"title":"Different roles for different bones—Location matters for blood production","authors":"David G. Kent","doi":"10.1002/hem3.127","DOIUrl":"https://doi.org/10.1002/hem3.127","url":null,"abstract":"<p>Laboratories studying hematopoietic stem cell (HSC) biology from across the world all have their own way of doing things, and this extends not just to the various cell surface marker combinations used to isolate HSCs but also to the more mundane aspects of cell isolation and preparation. At the very beginning of an HSC experiment, all researchers are faced with the question of which bones to obtain their bone marrow sample from. In the mouse, where options are more numerous, researchers commonly obtain marrow samples from the tibia, femur, hip, sternum, or spine. Some researchers then proceed to isolate the marrow by flushing, others by centrifuging bones, and still others by crushing. Yet, the vast majority of studies simply report that mouse bone marrow was isolated, and where it is specified, it is rarely looked at as a major experimental variable (e.g., a study with bone marrow from the tibia and femur is not viewed differently to one that also obtains marrow from spine and hip). Does it really matter which bone (or which part of which bone) cells are isolated from? Recent evidence suggests that the answer is a resounding “yes.”</p><p>While numerous anecdotes have circulated around various conference circuits and between highly specialized HSC labs, it is rare to see published studies that go into the fine detail of differences in the anatomical location of bone marrow blood cells. That said, studies have been cropping up in various guises over the years, and a major landmark study came out this year from Daniel Lucas' group<span><sup>1</sup></span> that added fuel to the fire and appears to cement the unique roles of different bones in blood cell production. One of the earliest studies was from Brian Lord in the 1970s where colony-forming cells were isolated from either the central marrow shaft (dubbed “axial”) or the edges of the bone (named “marginal”) and distinct numbers and types of colonies resided in each cell preparation.<span><sup>2</sup></span> This study preceded a large number of studies that focused on different types of bone and bone matrix, different proposed HSC niches, and different neighboring cell types (reviewed in Comazzetto et al.<span><sup>3</sup></span>).</p><p>Another provocative study emerged from David Bryder's lab in 2015<span><sup>4</sup></span> that challenged the notion of transplantable HSCs being equally distributed across the skeleton of a repopulated mouse. Following 16 weeks of transplantation and monitoring of overall chimerism in the blood, each of the right and left legs (tibia, femur, and hip) were assessed for donor chimerism, and the differences between bones were substantial, with some animals showing chimerism nearly exclusively in a single bone. Experimentally, this again challenges researchers to not consider that all marrow is equal irrespective of location.</p><p>Coming back to the Lucas lab study and why it has made such a big impact in the field, they undertook a skeleton-wide imaging stud","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.127","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141730110","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}
Mian S, Ariza-McNaughton L, Anjos-Afonso F, et al. Influence of donor–recipient sex on engraftment of normal and leukemia stem cells in xenotransplantation. HemaSphere. 2024;8:e80.
In the author listing of the manuscript, the last name of an author was misspelled. The correct spelling is Remisha Gurung. This has been corrected.
We apologize for this error.
Mian S、Ariza-McNaughton L、Anjos-Afonso F 等:异种移植中供体与受体性别对正常干细胞和白血病干细胞移植的影响。HemaSphere.2024;8:e80.In the author listing of the manuscript, the last name of an author was misspelled.正确的拼写是 Remisha Gurung。我们对此错误深表歉意。
{"title":"Correction to “Influence of donor–recipient sex on engraftment of normal and leukemia stem cells in xenotransplantation”","authors":"","doi":"10.1002/hem3.119","DOIUrl":"https://doi.org/10.1002/hem3.119","url":null,"abstract":"<p>Mian S, Ariza-McNaughton L, Anjos-Afonso F, et al. Influence of donor–recipient sex on engraftment of normal and leukemia stem cells in xenotransplantation. <i>HemaSphere</i>. 2024;8:e80.</p><p>In the author listing of the manuscript, the last name of an author was misspelled. The correct spelling is Remisha Gurung. This has been corrected.</p><p>We apologize for this error.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.119","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141730111","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>The quest to unlock the secrets of eternal youth or extending life span have been described throughout history. This includes ancient texts describing the “fountain of youth” and more recently within the popular fiction series “Harry Potter” where the philosopher's stone provides an “elixir of life.” In reality, our longevity is in part due to the presence of an effective immune system coupled with our ability to pre-emptively manipulate this using vaccinations. Indeed, it could be argued that vaccinations remain one of the most successful health interventions in human history, consigning many of the debilitating illness such as smallpox to the annals of history. Nevertheless, there remains an ongoing need for the rapid development and deployment of new vaccines to protect ourselves against new and emerging threats such as COVID-19. However, whilst we can design new vaccines based on an exquisite understanding of the “enemy,” this needs to be coupled with an individual's ability to mount an effective immune response—which, unfortunately, declines as we age.<span><sup>1</sup></span></p><p>It is now established that the hematopoietic stem cell (HSC) population changes over time.<span><sup>2</sup></span> In youth, the HSCs population has a balanced output of lymphoid and myeloid cells (bal-HSCs), but then changes towards myeloid-biased HSCs (my-HSCs) in older individuals. This in turn results in decreased lymphopoiesis, increased myelopoiesis as well as proinflammation, myeloid-related malignancies and a reduced adaptive immune response in older individuals.<span><sup>3</sup></span> An elegant new study published in Nature by the Weissman lab<span><sup>4</sup></span> now provides a tantalising new approach to improve our immune response. They demonstrate that an antibody depletion-based strategy targeting my-HSCs, can push the immune system in favor of a more balanced HSCs, and in essence, reversing time to rejuvenate an old immune system to a more youthful age.</p><p>Previous studies which have characterised the HSC population (Lin<sup>−</sup>, SCA1<sup>+</sup> KIT<sup>+</sup> FLT3<sup>−</sup> CD34<sup>−</sup>), found that my-HSCs have higher expression of CD150 (<i>Slamf1</i>) compared to bal-HSCs.<span><sup>3</sup></span> In this new study, Ross et al.<span><sup>4</sup></span> sought to extend this by using transcriptional datasets to find the best set of cell surface markers that identify my-HSCs for antibody targeting. They found that the most highly enriched cell-surface markers in my-HSCs were CD41 (<i>Itga2b</i>), CD62p (<i>Selp</i>), and NEO1 (<i>Neo1</i>). These were then validated using flow cytometry on CD150<sup>high</sup> HSCs (my-HSCs) vs. CD150<sup>low</sup> HSCs (bal-HSCs) and found that in older mice, the proportion of HSCs that were NEO1+, CD41+, and CD62p+ increased, consistent with the observed increase in my-HSCs associated with aging. These markers were also largely limited to HSCs except for CD41 that was also found on meg
对于捐献者来说,在捐献前使用抗体预处理来清除我的造血干细胞,不仅能提高捐献者造血干细胞库的质量,还能让老年人群成为潜在的捐献者。汉森-科萨希(Hansen J. Kosasih)和查尔斯-德-博克(Charles E. de Bock)共同构思并撰写了这篇文章。两位作者均同意最终版本。作者声明没有利益冲突。
{"title":"Less is more: Depleting myeloid-biased HSCs to restore immune function","authors":"Hansen J. Kosasih, Charles E. de Bock","doi":"10.1002/hem3.125","DOIUrl":"https://doi.org/10.1002/hem3.125","url":null,"abstract":"<p>The quest to unlock the secrets of eternal youth or extending life span have been described throughout history. This includes ancient texts describing the “fountain of youth” and more recently within the popular fiction series “Harry Potter” where the philosopher's stone provides an “elixir of life.” In reality, our longevity is in part due to the presence of an effective immune system coupled with our ability to pre-emptively manipulate this using vaccinations. Indeed, it could be argued that vaccinations remain one of the most successful health interventions in human history, consigning many of the debilitating illness such as smallpox to the annals of history. Nevertheless, there remains an ongoing need for the rapid development and deployment of new vaccines to protect ourselves against new and emerging threats such as COVID-19. However, whilst we can design new vaccines based on an exquisite understanding of the “enemy,” this needs to be coupled with an individual's ability to mount an effective immune response—which, unfortunately, declines as we age.<span><sup>1</sup></span></p><p>It is now established that the hematopoietic stem cell (HSC) population changes over time.<span><sup>2</sup></span> In youth, the HSCs population has a balanced output of lymphoid and myeloid cells (bal-HSCs), but then changes towards myeloid-biased HSCs (my-HSCs) in older individuals. This in turn results in decreased lymphopoiesis, increased myelopoiesis as well as proinflammation, myeloid-related malignancies and a reduced adaptive immune response in older individuals.<span><sup>3</sup></span> An elegant new study published in Nature by the Weissman lab<span><sup>4</sup></span> now provides a tantalising new approach to improve our immune response. They demonstrate that an antibody depletion-based strategy targeting my-HSCs, can push the immune system in favor of a more balanced HSCs, and in essence, reversing time to rejuvenate an old immune system to a more youthful age.</p><p>Previous studies which have characterised the HSC population (Lin<sup>−</sup>, SCA1<sup>+</sup> KIT<sup>+</sup> FLT3<sup>−</sup> CD34<sup>−</sup>), found that my-HSCs have higher expression of CD150 (<i>Slamf1</i>) compared to bal-HSCs.<span><sup>3</sup></span> In this new study, Ross et al.<span><sup>4</sup></span> sought to extend this by using transcriptional datasets to find the best set of cell surface markers that identify my-HSCs for antibody targeting. They found that the most highly enriched cell-surface markers in my-HSCs were CD41 (<i>Itga2b</i>), CD62p (<i>Selp</i>), and NEO1 (<i>Neo1</i>). These were then validated using flow cytometry on CD150<sup>high</sup> HSCs (my-HSCs) vs. CD150<sup>low</sup> HSCs (bal-HSCs) and found that in older mice, the proportion of HSCs that were NEO1+, CD41+, and CD62p+ increased, consistent with the observed increase in my-HSCs associated with aging. These markers were also largely limited to HSCs except for CD41 that was also found on meg","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.125","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141730160","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}
Judith M. Boer, Uri Ilan, Aurélie Boeree, Karin P. S. Langenberg, Jan Koster, Marco J. Koudijs, Jayne Y. Hehir-Kwa, Stefan Nierkens, Corinne Rossi, Jan J. Molenaar, Bianca F. Goemans, Monique L. den Boer, C. Michel Zwaan
Over the past 10 years, institutional and national molecular tumor boards have been implemented for relapsed or refractory pediatric cancer to prioritize targeted drugs for individualized treatment based on actionable oncogenic lesions, including the Dutch iTHER platform. Hematological malignancies form a minority in precision medicine studies. Here, we report on 56 iTHER leukemia/lymphoma patients for which we considered cell surface markers and oncogenic aberrations as actionable events, supplemented with ex vivo drug sensitivity for six patients. Prior to iTHER registration, 34% of the patients had received allogeneic hematopoietic cell transplantation (HCT) and 18% CAR-T therapy. For 51 patients (91%), a sample with sufficient tumor percentage (≥20%) required for comprehensive diagnostic testing was obtained. Up to 10 oncogenic actionable events were prioritized in 49/51 patients, and immunotherapy targets were identified in all profiled patients. Targeted treatment(s) based on the iTHER advice was given to 24 of 51 patients (47%), including immunotherapy in 17 patients, a targeted drug matching an oncogenic aberration in 12 patients, and a drug based on ex vivo drug sensitivity in one patient, resulting in objective responses and a bridge to HCT in the majority of the patients. In conclusion, comprehensive profiling of relapsed/refractory hematological malignancies showed multiple oncogenic and immunotherapy targets for a precision medicine approach, which requires multidisciplinary expertise to prioritize the best treatment options for this rare, heavily pretreated pediatric population.
{"title":"Oncogenic and immunological targets for matched therapy of pediatric blood cancer patients: Dutch iTHER study experience","authors":"Judith M. Boer, Uri Ilan, Aurélie Boeree, Karin P. S. Langenberg, Jan Koster, Marco J. Koudijs, Jayne Y. Hehir-Kwa, Stefan Nierkens, Corinne Rossi, Jan J. Molenaar, Bianca F. Goemans, Monique L. den Boer, C. Michel Zwaan","doi":"10.1002/hem3.122","DOIUrl":"10.1002/hem3.122","url":null,"abstract":"<p>Over the past 10 years, institutional and national molecular tumor boards have been implemented for relapsed or refractory pediatric cancer to prioritize targeted drugs for individualized treatment based on actionable oncogenic lesions, including the Dutch iTHER platform. Hematological malignancies form a minority in precision medicine studies. Here, we report on 56 iTHER leukemia/lymphoma patients for which we considered cell surface markers and oncogenic aberrations as actionable events, supplemented with ex vivo drug sensitivity for six patients. Prior to iTHER registration, 34% of the patients had received allogeneic hematopoietic cell transplantation (HCT) and 18% CAR-T therapy. For 51 patients (91%), a sample with sufficient tumor percentage (≥20%) required for comprehensive diagnostic testing was obtained. Up to 10 oncogenic actionable events were prioritized in 49/51 patients, and immunotherapy targets were identified in all profiled patients. Targeted treatment(s) based on the iTHER advice was given to 24 of 51 patients (47%), including immunotherapy in 17 patients, a targeted drug matching an oncogenic aberration in 12 patients, and a drug based on ex vivo drug sensitivity in one patient, resulting in objective responses and a bridge to HCT in the majority of the patients. In conclusion, comprehensive profiling of relapsed/refractory hematological malignancies showed multiple oncogenic and immunotherapy targets for a precision medicine approach, which requires multidisciplinary expertise to prioritize the best treatment options for this rare, heavily pretreated pediatric population.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247331/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619862","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}
Frank G. Rücker, Andrea Corbacioglu, Julia Krzykalla, Sibylle Cocciardi, Claudia Lengerke, Ulrich Germing, Gerald Wulf, Maisun A. Samra, Lino L. Teichmann, Michael Lübbert, Michael W. M. Kühn, Martin Bentz, Jörg Westermann, Lars Bullinger, Verena I. Gaidzik, Annika Meid, Sophia Aicher, Frank Stegelmann, Daniela Weber, Anika Schrade, Felicitas Thol, Michael Heuser, Arnold Ganser, Axel Benner, Hartmut Döhner, Konstanze Döhner, for the German-Austrian Acute Myeloid Leukemia Study Group (AMLSG)
<p>The transcription factor CCAAT/enhancer binding protein alpha (CEBPA) is a key regulator of myelopoiesis and granulocyte differentiation.<span><sup>1, 2</sup></span> The intronless <i>CEBPA</i> gene on chromosome 19q13.1 encodes two DNA-binding protein isoforms: a full-length 42-kDa protein (p42) and a shorter 30-kDa isoform (p30), initiated from two distinct start sites.<span><sup>2</sup></span> The p42 isoform contains two N-terminal transactivation domains (TAD1, TAD2), whereas the p30 isoform lacks TAD1. Both isoforms contain the highly conserved C-terminal basic DNA-binding domain and the leucine zipper (bZIP) involved in DNA binding and protein dimerization. In younger adult patients, mutations of <i>CEBPA</i> (<i>CEBPA</i><sup>mut</sup>) are present in 5%–10% of newly diagnosed acute myeloid leukemia (AML); the frequency in older patients is considerably lower.<span><sup>2-5</sup></span> There are two mutational patterns: the first one clusters at the N-terminus involving the two TADs, typically frame-shift mutations; the second one at the C-terminus affecting bZIP, typically in-frame mutations. Out-of-frame TAD mutations result in the truncated p30 isoform that has been shown to act as a dominant negative of the p42 isoform and to be associated with increased proliferation and minimal differentiation of myeloid progenitors.<span><sup>2, 6</sup></span> Depending on the position, in-frame bZIP mutations cause a p42 isoform defective either in DNA binding or homo- and heterodimerization.<span><sup>3, 7</sup></span></p><p>Approximately half of the <i>CEBPA</i><sup>mut</sup> AML exhibit biallelic mutations (<i>CEBPA</i><sup>bi</sup>), typically consisting of one TAD and one bZIP mutation on separate alleles.<span><sup>3, 4</sup></span> Based on specific genetic features and its prognostic impact, <i>CEBPA</i><sup>bi</sup> was defined as a distinct entity within the 2016 WHO classification and was categorized as favorable in the risk stratification of the 2017 European LeukemiaNet (ELN) recommendations. Recent studies in pediatric and adult AML have demonstrated <i>CEBPA</i><sup>bZIP</sup> mutations, and in particular, in-frame mutations (<i>CEBPA</i><sup>bZIP_inf</sup>), to be associated with a unique gene-expression profile and favorable outcome, regardless of the mono- or biallelic status.<span><sup>8-10</sup></span> Based on these data, the former entity of AML with <i>CEBPA</i><sup>bi</sup> was expanded by single mutations in bZIP (smbZIP-<i>CEBPA</i>) in the current 2022 WHO classification and replaced by AML with <i>CEBPA</i><sup>bZIP_inf</sup> (irrespective of the allelic status) within the 2022 International Consensus Classification (ICC) of myeloid neoplasm and acute leukemias.<span><sup>11, 12</sup></span> Furthermore, <i>CEBPA</i><sup>bZIP_inf</sup> (irrespective of the allelic status) is now categorized as favorable in the 2022 ELN risk stratification.<span><sup>13</sup></span></p><p>To evaluate the prognostic impact of <i>CEBPA
smCEBPA)和突变类型:(1) dmCEBPA 伴有 bZIP 的框内插入/缺失(dmCEBPAbZIP_InDel_inf,n = 220),(2) bZIP 的移帧插入/缺失或无义突变(dmCEBPAbZIP_InDel_fs,n = 13),(3) bZIP 的错义突变(dmCEBPAbZIP_ms、n = 22),(4)其他(dmCEBPAother,n = 8),(5)smCEBPAbZIP_InDel_inf(n = 46),(6)smCEBPAbZIP_InDel_fs(n = 32),(7)smCEBPAbZIP_ms(n = 11),或(8)smCEBPAother(n = 176)。这八组患者在一些临床和并发遗传特征以及达到 CR1 和预后方面存在显著差异(佐证资料 S1:表 1)。为了评估八种 CEBPA 突变类型对预后的影响,我们对 EFS 和 OS 进行了条件推理树模型。在八个同样相关的组别中,dmCEBPAbZIP_InDel_inf 和 smCEBPAbZIP_InDel_inf 患者因事件(难治性疾病、复发和/或死亡)累积发生率显著较低而在第一个树中与其他组别区分开来。没有观察到进一步的分区(佐证资料 S1:图 1)。假设预后影响主要来自突变类型而非等位基因状态,那么随后的条件推断树模型则通过拆分CEBPA突变类型和等位基因状态来计算EFS和OS。在这两个模型中,CEBPAbZIP_InDel_inf 组再次与第一个树中的其他三组显著分离,而没有进一步分离,这证实了无论等位基因状态如何,CEBPAbZIP_InDel_inf 组的预后都显著良好(图 1A 和佐证资料 S1:图 2)。根据这些发现,随后将患者分为 CEBPAbZIP_InDel_inf(n = 266)、CEBPAbZIP_InDel_fs(n = 45)、CEBPAbZIP_ms(n = 33)或 CEBPAother(n = 184)四组,与等位基因状态无关。这四组患者在一些临床和并发遗传特征以及 CR1 的达标率方面存在明显差异。最明显的是,CEBPAbZIP_InDel_inf 患者更年轻(中位年龄:49 vs. 66 vs. 60 vs. 61; p <.001),达到 CR1 的比例更高(91.4% vs. 81.8% vs. 83.3% vs. 76.2%; p <.001)(佐证资料 S1:表 2)。无论等位基因状态如何,CEBPAbZIP_InDel_inf 患者的 EFS 显著改善(中位[95% CI] 49.8 个月 [16.9-82.7] vs. CEBPAbZIP_InDel_fs 11.5 [8.3-14.6] vs. CEBPAbZIP_ms 12.6 [6.2-19.1] vs. CEBPAother 14.6 [7.7-21.5];P <;.001)和 OS(CEBPAbZIP_InDel_inf 患者的中位数[95% CI] NA [NA-NA] vs. CEBPAbZIP_InDel_fs 患者的 25.7 个月 [10.2-41.3] vs. CEBPAbZIP_ms 患者的 54.3 [14.6-NA] vs. CEBPAother 患者的 45.5 [13.1-77.9] ;P <;.001)(图 1B、C)。值得注意的是,在一项敏感性分析中,将 CR1 患者的生存时间以 HCT 日期为截止日期,结果几乎相同(佐证资料 S1:图 3)。根据性别、急性髓细胞性白血病类型、FLT3-ITD、NPM1突变状态、白细胞(WBC)计数(log10转化)、骨髓细胞数和年龄(包括CR1期HCT作为时间依赖变量)调整的EFS和OS多变量Cox模型(佐证资料S1:附录)显示,年龄增加和WBC升高是不利因素,而CEBPAbZIP_InDel_inf、NPM1突变和CR1期HCT则是有利因素(表1)。最近的一项荟萃分析对来自 6 个不同研究组的 1010 例成人 CEBPA 突变 AML 进行了更详细的分析,通过评估其临床和遗传特征及其对预后的影响,确定了 CEBPA 突变亚组的特征。作者的研究结果与我们的数据一致,CEBPAbZIP_InDel_inf 突变代表了具有不同疾病生物学特性和临床结局的 AML 亚组。尽管存在一定的局限性,尤其是回顾性研究跨越了近三十年,不同的治疗方法也在不断演变,但这两项研究都独立证实了框架内 bZIP 错义突变(目前在 ICC 和 ELN 风险分类中被归入具有 CEBPAbZIP_inf 的急性髓细胞性白血病类别)的不利影响。bZIP错义突变的影响较小,这可能是因为发现bZIP错义突变的定位方式不同,因此具有不同的功能性后果。CEBPAbZIP_InDel_inf 主要影响 bZIP 的分叉区,而 CEBPAbZIP_ms 则聚集在基本区(佐证资料 S1:图 4)。这些不同的突变模式意味着 CEBPAbZIP_ms 影响 DNA 结合,而 CEBPAbZIP_InDel_inf 影响二聚化。小鼠数据显示,bZIP 基本区的错义突变会导致骨髓增生性疾病转变为明显的急性髓细胞性白血病7,而当移植携带 bZIP 最常见的框架内插入突变(K313dup,K-等位基因)的转基因细胞时,会出现急性髓细胞性白血病,K/L 组合会导致最具侵袭性的急性髓细胞性白血病。 15 在这项对 528 例新诊断的接受强化治疗的 CEBPAmut AML 成人患者进行的回顾性、探索性分析中,我们进一步完善了不同 CEBPA 突变类型,尤其是位于 bZIP 结构域的 CEBPA 突变对预后的影响。我们的研究表明,无论等位基因状态如何,有利影响仅限于 CEBPA bZIP InDel 框架内突变,而 CEBPA bZIP 错义突变与不良预后相关。我们的数据以及 Georgi 等人的数据提供了新颖且与临床相关的结果,有助于进一步完善当前 ICC 和 WHO 分类中的 CEBPAmut AML,以及 ELN 建议的风险分层。Frank G. Rücker、Andrea Corbacioglu、Sibylle Cocciardi、Verena I. Gaidzik、Annika Meid 和 Sophia Aicher 进行了实验并验证了数据。Frank G. Rücker、Julia Krzykalla、Daniela Weber 和 Axel Benner 进行了统计分析。Frank G. Rücker、Andrea Corbacioglu、Julia Krzykalla、Daniela Weber、Axel Benner、Hartmut Döhner 和 Konstanze Döhner 对结果进行了分析。Claudia Lengerke、Ulrich Germing、Gerald Wulf、Maisun A. Samra、Lino L. Teichmann、Michael Lübbert、Michael W. M. Kühn、Martin Bentz、Jörg Westermann、Lars Bullinger、
{"title":"Refinement of the prognostic impact of somatic CEBPA bZIP domain mutations in acute myeloid leukemia: Results of the AML Study Group (AMLSG)","authors":"Frank G. Rücker, Andrea Corbacioglu, Julia Krzykalla, Sibylle Cocciardi, Claudia Lengerke, Ulrich Germing, Gerald Wulf, Maisun A. Samra, Lino L. Teichmann, Michael Lübbert, Michael W. M. Kühn, Martin Bentz, Jörg Westermann, Lars Bullinger, Verena I. Gaidzik, Annika Meid, Sophia Aicher, Frank Stegelmann, Daniela Weber, Anika Schrade, Felicitas Thol, Michael Heuser, Arnold Ganser, Axel Benner, Hartmut Döhner, Konstanze Döhner, for the German-Austrian Acute Myeloid Leukemia Study Group (AMLSG)","doi":"10.1002/hem3.123","DOIUrl":"10.1002/hem3.123","url":null,"abstract":"<p>The transcription factor CCAAT/enhancer binding protein alpha (CEBPA) is a key regulator of myelopoiesis and granulocyte differentiation.<span><sup>1, 2</sup></span> The intronless <i>CEBPA</i> gene on chromosome 19q13.1 encodes two DNA-binding protein isoforms: a full-length 42-kDa protein (p42) and a shorter 30-kDa isoform (p30), initiated from two distinct start sites.<span><sup>2</sup></span> The p42 isoform contains two N-terminal transactivation domains (TAD1, TAD2), whereas the p30 isoform lacks TAD1. Both isoforms contain the highly conserved C-terminal basic DNA-binding domain and the leucine zipper (bZIP) involved in DNA binding and protein dimerization. In younger adult patients, mutations of <i>CEBPA</i> (<i>CEBPA</i><sup>mut</sup>) are present in 5%–10% of newly diagnosed acute myeloid leukemia (AML); the frequency in older patients is considerably lower.<span><sup>2-5</sup></span> There are two mutational patterns: the first one clusters at the N-terminus involving the two TADs, typically frame-shift mutations; the second one at the C-terminus affecting bZIP, typically in-frame mutations. Out-of-frame TAD mutations result in the truncated p30 isoform that has been shown to act as a dominant negative of the p42 isoform and to be associated with increased proliferation and minimal differentiation of myeloid progenitors.<span><sup>2, 6</sup></span> Depending on the position, in-frame bZIP mutations cause a p42 isoform defective either in DNA binding or homo- and heterodimerization.<span><sup>3, 7</sup></span></p><p>Approximately half of the <i>CEBPA</i><sup>mut</sup> AML exhibit biallelic mutations (<i>CEBPA</i><sup>bi</sup>), typically consisting of one TAD and one bZIP mutation on separate alleles.<span><sup>3, 4</sup></span> Based on specific genetic features and its prognostic impact, <i>CEBPA</i><sup>bi</sup> was defined as a distinct entity within the 2016 WHO classification and was categorized as favorable in the risk stratification of the 2017 European LeukemiaNet (ELN) recommendations. Recent studies in pediatric and adult AML have demonstrated <i>CEBPA</i><sup>bZIP</sup> mutations, and in particular, in-frame mutations (<i>CEBPA</i><sup>bZIP_inf</sup>), to be associated with a unique gene-expression profile and favorable outcome, regardless of the mono- or biallelic status.<span><sup>8-10</sup></span> Based on these data, the former entity of AML with <i>CEBPA</i><sup>bi</sup> was expanded by single mutations in bZIP (smbZIP-<i>CEBPA</i>) in the current 2022 WHO classification and replaced by AML with <i>CEBPA</i><sup>bZIP_inf</sup> (irrespective of the allelic status) within the 2022 International Consensus Classification (ICC) of myeloid neoplasm and acute leukemias.<span><sup>11, 12</sup></span> Furthermore, <i>CEBPA</i><sup>bZIP_inf</sup> (irrespective of the allelic status) is now categorized as favorable in the 2022 ELN risk stratification.<span><sup>13</sup></span></p><p>To evaluate the prognostic impact of <i>CEBPA","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247273/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619864","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}
Achille Iolascon, Immacolata Andolfo, Roberta Russo, Mayka Sanchez, Fabiana Busti, Dorine Swinkels, Patricia Aguilar Martinez, Rayan Bou-Fakhredin, Martina U. Muckenthaler, Sule Unal, Graça Porto, Tomas Ganz, Antonis Kattamis, Lucia De Franceschi, Maria Domenica Cappellini, Malcolm G. Munro, Ali Taher, from EHA-SWG Red Cell and Iron
Iron is an essential nutrient and a constituent of ferroproteins and enzymes crucial for human life. Generally, nonmenstruating individuals preserve iron very efficiently, losing less than 0.1% of their body iron content each day, an amount that is replaced through dietary iron absorption. Most of the iron is in the hemoglobin (Hb) of red blood cells (RBCs); thus, blood loss is the most common cause of acute iron depletion and anemia worldwide, and reduced hemoglobin synthesis and anemia are the most common consequences of low plasma iron concentrations. The term iron deficiency (ID) refers to the reduction of total body iron stores due to impaired nutrition, reduced absorption secondary to gastrointestinal conditions, increased blood loss, and increased needs as in pregnancy. Iron deficiency anemia (IDA) is defined as low Hb or hematocrit associated with microcytic and hypochromic erythrocytes and low RBC count due to iron deficiency. IDA most commonly affects women of reproductive age, the developing fetus, children, patients with chronic and inflammatory diseases, and the elderly. IDA is the most frequent hematological disorder in children, with an incidence in industrialized countries of 20.1% between 0 and 4 years of age and 5.9% between 5 and 14 years (39% and 48.1% in developing countries). The diagnosis, management, and treatment of patients with ID and IDA change depending on age and gender and during pregnancy. We herein summarize what is known about the diagnosis, treatment, and prevention of ID and IDA and formulate a specific set of recommendations on this topic.
铁是人体必需的营养素,是铁蛋白和酶的组成成分,对人体生命至关重要。一般来说,非月经期的人保存铁的效率非常高,每天损失的铁含量不到体内铁含量的 0.1%,而这一数量可通过饮食中铁的吸收得到补充。大部分铁都存在于红细胞(RBC)的血红蛋白(Hb)中;因此,失血是全球急性铁耗竭和贫血最常见的原因,而血红蛋白合成减少和贫血则是血浆铁浓度过低最常见的后果。缺铁(ID)是指由于营养受损、胃肠道疾病导致吸收减少、失血增加以及妊娠期需求增加而导致体内总储铁减少。缺铁性贫血(IDA)是指由于缺铁导致的低血红蛋白或低血细胞比容,伴有小红细胞和低色素性红细胞以及低红细胞计数。IDA 最常见于育龄妇女、发育中的胎儿、儿童、慢性病和炎症患者以及老年人。IDA 是儿童最常见的血液病,在工业化国家,0 至 4 岁儿童的发病率为 20.1%,5 至 14 岁儿童的发病率为 5.9%(发展中国家的发病率分别为 39% 和 48.1%)。ID 和 IDA 患者的诊断、管理和治疗因年龄、性别和孕期而异。在此,我们总结了有关 ID 和 IDA 诊断、治疗和预防的已知知识,并就这一主题制定了一套具体的建议。
{"title":"Recommendations for diagnosis, treatment, and prevention of iron deficiency and iron deficiency anemia","authors":"Achille Iolascon, Immacolata Andolfo, Roberta Russo, Mayka Sanchez, Fabiana Busti, Dorine Swinkels, Patricia Aguilar Martinez, Rayan Bou-Fakhredin, Martina U. Muckenthaler, Sule Unal, Graça Porto, Tomas Ganz, Antonis Kattamis, Lucia De Franceschi, Maria Domenica Cappellini, Malcolm G. Munro, Ali Taher, from EHA-SWG Red Cell and Iron","doi":"10.1002/hem3.108","DOIUrl":"10.1002/hem3.108","url":null,"abstract":"<p>Iron is an essential nutrient and a constituent of ferroproteins and enzymes crucial for human life. Generally, nonmenstruating individuals preserve iron very efficiently, losing less than 0.1% of their body iron content each day, an amount that is replaced through dietary iron absorption. Most of the iron is in the hemoglobin (Hb) of red blood cells (RBCs); thus, blood loss is the most common cause of acute iron depletion and anemia worldwide, and reduced hemoglobin synthesis and anemia are the most common consequences of low plasma iron concentrations. The term iron deficiency (ID) refers to the reduction of total body iron stores due to impaired nutrition, reduced absorption secondary to gastrointestinal conditions, increased blood loss, and increased needs as in pregnancy. Iron deficiency anemia (IDA) is defined as low Hb or hematocrit associated with microcytic and hypochromic erythrocytes and low RBC count due to iron deficiency. IDA most commonly affects women of reproductive age, the developing fetus, children, patients with chronic and inflammatory diseases, and the elderly. IDA is the most frequent hematological disorder in children, with an incidence in industrialized countries of 20.1% between 0 and 4 years of age and 5.9% between 5 and 14 years (39% and 48.1% in developing countries). The diagnosis, management, and treatment of patients with ID and IDA change depending on age and gender and during pregnancy. We herein summarize what is known about the diagnosis, treatment, and prevention of ID and IDA and formulate a specific set of recommendations on this topic.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247274/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619863","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}
Enrico Attardi, Lucia Tiberi, Giorgio Mattiuz, Daniela Formicola, Elia Dirupo, Marco G. Raddi, Angela Consagra, Debora Vergani, Rosangela Artuso, Valeria Santini
Relevance of germline (GL) predisposition in myelodysplastic syndromes (MDSs) was stressed in both 2022 WHO and International Consensus classifications, but its incidence is probably underestimated, especially in young adult patients. We selected a cohort of 31 consecutive de novo MDS patients with unusual young age (<60 years). We performed exome sequencing (ES) on DNA extracted from noninvasive sources (peripheral blood and saliva), filtering for a panel of 344 genes specifically tailored for detecting GL variants related to clonal and nonclonal cytopenia. We observed at least one high- or low-confidence GL MDS variant in 7/31 (22.6%) and 9/31 (29.0%) of cases, respectively. Four of 31 patients (12.9%) confirmed having established MDS/AML predisposing disorders. We found heterozygous variants in genes involved in DNA repair/cancer predisposition (ATM, ATR, FANCM, PARN, BRCA1, BRCA2, CHEK2, MSH2) in 9/31 (29.0%) cases and variants affecting ribosome biogenesis (SBDS), hematopoietic stem cell (GATA2), and megakaryocyte (ANKRD26) differentiation in single cases. Two cases had variants in RBBP6, a gene previously described exclusively in familial myeloproliferative neoplasms. Lastly, four cases had variants in genes related to inherited anemias (CUBN and PIEZO1 genes). Our results showed that “young” MDS patients aged 40–60 years carried reported and unreported GL variants with an unexpectedly high proportion, and these events co-occurred with somatic mutations recurrent in myeloid neoplasms. We explored the “no man's land” of the young adult MDS cases adopting a practical and scalable diagnostic tool, capable to detect GL variants avoiding invasive methods.
{"title":"Prospective genetic germline evaluation in a consecutive group of adult patients aged <60 years with myelodysplastic syndromes","authors":"Enrico Attardi, Lucia Tiberi, Giorgio Mattiuz, Daniela Formicola, Elia Dirupo, Marco G. Raddi, Angela Consagra, Debora Vergani, Rosangela Artuso, Valeria Santini","doi":"10.1002/hem3.112","DOIUrl":"10.1002/hem3.112","url":null,"abstract":"<p>Relevance of germline (GL) predisposition in myelodysplastic syndromes (MDSs) was stressed in both 2022 WHO and International Consensus classifications, but its incidence is probably underestimated, especially in young adult patients. We selected a cohort of 31 consecutive de novo MDS patients with unusual young age (<60 years). We performed exome sequencing (ES) on DNA extracted from noninvasive sources (peripheral blood and saliva), filtering for a panel of 344 genes specifically tailored for detecting GL variants related to clonal and nonclonal cytopenia. We observed at least one high- or low-confidence GL MDS variant in 7/31 (22.6%) and 9/31 (29.0%) of cases, respectively. Four of 31 patients (12.9%) confirmed having established MDS/AML predisposing disorders. We found heterozygous variants in genes involved in DNA repair/cancer predisposition (<i>ATM, ATR, FANCM, PARN, BRCA1, BRCA2, CHEK2, MSH2</i>) in 9/31 (29.0%) cases and variants affecting ribosome biogenesis (<i>SBDS</i>), hematopoietic stem cell (<i>GATA2</i>), and megakaryocyte (<i>ANKRD26</i>) differentiation in single cases. Two cases had variants in <i>RBBP6</i>, a gene previously described exclusively in familial myeloproliferative neoplasms. Lastly, four cases had variants in genes related to inherited anemias (<i>CUBN</i> and <i>PIEZO1</i> genes). Our results showed that “young” MDS patients aged 40–60 years carried reported and unreported GL variants with an unexpectedly high proportion, and these events co-occurred with somatic mutations recurrent in myeloid neoplasms. We explored the “no man's land” of the young adult MDS cases adopting a practical and scalable diagnostic tool, capable to detect GL variants avoiding invasive methods.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11250510/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141626550","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}
Riccardo Bomben, Antonella Zucchetto, Roberta Laureana, Annalisa Chiarenza, Jacopo Olivieri, Erika Tissino, Francesca M. Rossi, Filippo Vit, Tamara Bittolo, Robel Papotti, Federico Pozzo, Annalisa Gaglio, Massimo Degan, Jerry Polesel, Roberto Marasca, Andrea Visentin, Riccardo Moia, Idanna Innocenti, Candida Vitale, Roberta Murru, Marzia Varettoni, Agostino Tafuri, Francesco Zaja, Massimiliano Postorino, Enrica A. Martino, Adalgisa Condoluci, Davide Rossi, Antonio Cuneo, Francesco Di Raimondo, Paolo Sportoletti, Ilaria Del Giudice, Robin Foà, Francesca R. Mauro, Marta Coscia, Luca Laurenti, Gianluca Gaidano, Livio Trentin, Maria I. Del Principe, Massimo Gentile, Valter Gattei
<p>Chronic lymphocytic leukemia (CLL) is a malignancy of mature clonal B lymphocytes that accumulate in blood, bone marrow, and lymphoid tissues.<span><sup>1</sup></span> One of the most important key players in the pathobiology and progression of CLL is the B-cell receptor (BCR) whose activation supports growth and survival of CLL cells.<span><sup>2</sup></span> For this reason, the use of Bruton's tyrosine kinase inhibitors (BTKi) including ibrutinib emerged as one of the most effective treatment options for both naïve (TN) and relapsed/refractory (RR) CLL.<span><sup>2</sup></span> Treatment with BTKi often results in an increase blood lymphocytosis driven by the release of cells from lymph nodes due to impaired interaction with the microenvironment.<span><sup>2-4</sup></span> One of the key molecules of these interactions is the integrin VLA-4 which mediates both cell–cell and cell–matrix interactions playing a crucial role in the retention of CLL cells in tissue-sites thus protecting them from proapoptotic signal.<span><sup>3, 5-7</sup></span> In keeping with these observations, high expression of the VLA-4 integrin alpha chain CD49d (≥30% positive cells), or expression of CD49d according to a bimodal pattern (i.e., concurrent CD49d-positive and CD49d-negative subpopulations, irrespective of the 30% cutoff) identifies CLL cases with reduced recirculation lymphocytosis, inferior nodal response, and shorter progression-free survival (PFS) in the ibrutinib setting.<span><sup>4, 5</sup></span></p><p>In addition to CD49d evaluation, a 4-factor model has been proposed to identify patients at high risk of treatment failure and death during ibrutinib therapy, afterward validated in real-world, which included the TN/RR status, the levels of β2-microglobulin (β2M) and lactate dehydrogenase (LDH) serum concentration, and the TP53 disruption status.<span><sup>8, 9</sup></span> In this context, as well as in other clinical studies on CLL, the TP53 disruption category includes CLL cases with either concurrent TP53 mutation and deletion, or only one of the lesions.<span><sup>1, 8, 9</sup></span> Recently, the prognostic impact of TP53 disruption in ibrutinib-treated CLL has been refined by demonstrating that only cases with the concomitant presence of TP53 deletion and mutations, did not gain maximum benefit from this therapy.<span><sup>10, 11</sup></span></p><p>The aim of this study is to integrate these observations in a comprehensive scoring system for a better management of ibrutinib-treated patients.</p><p>The study is a retrospective/multicenter analysis of 401 CLL patients treated with ibrutinib in the current clinical practice (12/2013–03/2022; approvals IRB-05-2010/IRB-05-2015; CRO Aviano). All CLL cases were characterized for CD49d expression, as reported previously.<span><sup>4-7, 12</sup></span> TP53 disruption was simultaneously evaluated by FISH (17p deletion, del17p) and next-generation sequencing (<i>TP53</i> mutations), as reported.<span><s
{"title":"CD49d expression is included in a revised 4-factor model predicting outcome in patients with chronic lymphocytic leukemia treated with ibrutinib: A multicenter real-world experience","authors":"Riccardo Bomben, Antonella Zucchetto, Roberta Laureana, Annalisa Chiarenza, Jacopo Olivieri, Erika Tissino, Francesca M. Rossi, Filippo Vit, Tamara Bittolo, Robel Papotti, Federico Pozzo, Annalisa Gaglio, Massimo Degan, Jerry Polesel, Roberto Marasca, Andrea Visentin, Riccardo Moia, Idanna Innocenti, Candida Vitale, Roberta Murru, Marzia Varettoni, Agostino Tafuri, Francesco Zaja, Massimiliano Postorino, Enrica A. Martino, Adalgisa Condoluci, Davide Rossi, Antonio Cuneo, Francesco Di Raimondo, Paolo Sportoletti, Ilaria Del Giudice, Robin Foà, Francesca R. Mauro, Marta Coscia, Luca Laurenti, Gianluca Gaidano, Livio Trentin, Maria I. Del Principe, Massimo Gentile, Valter Gattei","doi":"10.1002/hem3.128","DOIUrl":"10.1002/hem3.128","url":null,"abstract":"<p>Chronic lymphocytic leukemia (CLL) is a malignancy of mature clonal B lymphocytes that accumulate in blood, bone marrow, and lymphoid tissues.<span><sup>1</sup></span> One of the most important key players in the pathobiology and progression of CLL is the B-cell receptor (BCR) whose activation supports growth and survival of CLL cells.<span><sup>2</sup></span> For this reason, the use of Bruton's tyrosine kinase inhibitors (BTKi) including ibrutinib emerged as one of the most effective treatment options for both naïve (TN) and relapsed/refractory (RR) CLL.<span><sup>2</sup></span> Treatment with BTKi often results in an increase blood lymphocytosis driven by the release of cells from lymph nodes due to impaired interaction with the microenvironment.<span><sup>2-4</sup></span> One of the key molecules of these interactions is the integrin VLA-4 which mediates both cell–cell and cell–matrix interactions playing a crucial role in the retention of CLL cells in tissue-sites thus protecting them from proapoptotic signal.<span><sup>3, 5-7</sup></span> In keeping with these observations, high expression of the VLA-4 integrin alpha chain CD49d (≥30% positive cells), or expression of CD49d according to a bimodal pattern (i.e., concurrent CD49d-positive and CD49d-negative subpopulations, irrespective of the 30% cutoff) identifies CLL cases with reduced recirculation lymphocytosis, inferior nodal response, and shorter progression-free survival (PFS) in the ibrutinib setting.<span><sup>4, 5</sup></span></p><p>In addition to CD49d evaluation, a 4-factor model has been proposed to identify patients at high risk of treatment failure and death during ibrutinib therapy, afterward validated in real-world, which included the TN/RR status, the levels of β2-microglobulin (β2M) and lactate dehydrogenase (LDH) serum concentration, and the TP53 disruption status.<span><sup>8, 9</sup></span> In this context, as well as in other clinical studies on CLL, the TP53 disruption category includes CLL cases with either concurrent TP53 mutation and deletion, or only one of the lesions.<span><sup>1, 8, 9</sup></span> Recently, the prognostic impact of TP53 disruption in ibrutinib-treated CLL has been refined by demonstrating that only cases with the concomitant presence of TP53 deletion and mutations, did not gain maximum benefit from this therapy.<span><sup>10, 11</sup></span></p><p>The aim of this study is to integrate these observations in a comprehensive scoring system for a better management of ibrutinib-treated patients.</p><p>The study is a retrospective/multicenter analysis of 401 CLL patients treated with ibrutinib in the current clinical practice (12/2013–03/2022; approvals IRB-05-2010/IRB-05-2015; CRO Aviano). All CLL cases were characterized for CD49d expression, as reported previously.<span><sup>4-7, 12</sup></span> TP53 disruption was simultaneously evaluated by FISH (17p deletion, del17p) and next-generation sequencing (<i>TP53</i> mutations), as reported.<span><s","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247362/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619861","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}
Nina Mogensen, Ulrika Kreicbergs, Birgitte K. Albertsen, Päivi M. Lähteenmäki, Mats Heyman, Arja Harila
This study aimed to assess how parents perceived treatment-related side effects during acute lymphoblastic leukemia (ALL) treatment. Parents of children 1–17.9 years at diagnosis in Sweden, Finland, and Denmark who were alive and in first remission ≥6 months after end of ALL treatment were asked to respond on specific items regarding how their child was affected by side effects related to vincristine (VCR), corticosteroids, peg-asparaginase (ASP), and maintenance therapy, as well as overall impact of these treatments, complications in general, and their perception of impact on their child in comparison with other children with ALL. Parents of 307 children responded. More than a third reported that their child had been affected to a high extent by VCR (39.7%) and corticosteroids (35.8%), with walking difficulties, muscular weakness, pain, changes in appetite, and mood swings as the most common and severe symptoms. Reporting of these toxicities was lacking from the NOPHO ALL2008 database, except for peripheral paralysis (12.1%). For distinct toxicities reported in the NOPHO ALL2008 database, for example, thrombosis and pancreatitis, parent reports were similar to the database. Although a high overall negative impact during treatment was reported, parents generally rated the impact on their child as less, or similar, to other children with ALL. Parents perceived VCR and corticosteroid therapy, in particular, to have a negative impact on their child during ALL treatment, which was not captured in the NOPHO ALL2008 toxicity reporting. Our results highlight the importance of including patient/parent-reported outcomes in toxicity reporting.
{"title":"Parents' perception of treatment-related toxicity in children treated according to the NOPHO ALL2008 protocol for acute lymphoblastic leukemia","authors":"Nina Mogensen, Ulrika Kreicbergs, Birgitte K. Albertsen, Päivi M. Lähteenmäki, Mats Heyman, Arja Harila","doi":"10.1002/hem3.124","DOIUrl":"https://doi.org/10.1002/hem3.124","url":null,"abstract":"<p>This study aimed to assess how parents perceived treatment-related side effects during acute lymphoblastic leukemia (ALL) treatment. Parents of children 1–17.9 years at diagnosis in Sweden, Finland, and Denmark who were alive and in first remission ≥6 months after end of ALL treatment were asked to respond on specific items regarding how their child was affected by side effects related to vincristine (VCR), corticosteroids, peg-asparaginase (ASP), and maintenance therapy, as well as overall impact of these treatments, complications in general, and their perception of impact on their child in comparison with other children with ALL. Parents of 307 children responded. More than a third reported that their child had been affected to a high extent by VCR (39.7%) and corticosteroids (35.8%), with walking difficulties, muscular weakness, pain, changes in appetite, and mood swings as the most common and severe symptoms. Reporting of these toxicities was lacking from the NOPHO ALL2008 database, except for peripheral paralysis (12.1%). For distinct toxicities reported in the NOPHO ALL2008 database, for example, thrombosis and pancreatitis, parent reports were similar to the database. Although a high overall negative impact during treatment was reported, parents generally rated the impact on their child as less, or similar, to other children with ALL. Parents perceived VCR and corticosteroid therapy, in particular, to have a negative impact on their child during ALL treatment, which was not captured in the NOPHO ALL2008 toxicity reporting. Our results highlight the importance of including patient/parent-reported outcomes in toxicity reporting.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"8 7","pages":""},"PeriodicalIF":7.6,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.124","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141597148","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}