Pub Date : 2025-01-02DOI: 10.1182/blood.2024026463
Hervé Dombret
{"title":"Introduction to a How I Treat series on acute lymphoblastic leukemia.","authors":"Hervé Dombret","doi":"10.1182/blood.2024026463","DOIUrl":"https://doi.org/10.1182/blood.2024026463","url":null,"abstract":"","PeriodicalId":9102,"journal":{"name":"Blood","volume":"145 1","pages":"1-2"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2023023154
Jack Bartram, Philip Ancliff, Ajay Vora
Abstract: Infant acute lymphoblastic leukemia (ALL) is an aggressive malignancy that has historically been associated with a very poor prognosis. Despite large cooperative international trials and incremental increases in intensity of therapy, there has been no significant improvement in outcome over the last 3 decades. Using representative cases, we highlight the key differences between KMT2A-rearranged and KMT2A-germ line infant ALL, and how advances in molecular diagnostics are unpicking KMT2A-germ line genetics and guiding treatment reduction. We focus on KM2TA-rearranged infant B-cell ALL for which the last few years have seen the emergence of novel therapies that both are more effective and less toxic than conventional chemotherapy. Of these, there is promising early data on the efficacy and tolerability of the bispecific T-cell engager monoclonal antibody, blinatumomab, as well as the use of autologous and allogeneic chimeric antigen receptor T-cell therapy. We discuss how we can improve risk stratification and incorporate these new agents to replace the most toxic elements of currently deployed intensive chemotherapy schedules with their associated unacceptable toxicity.
婴儿急性淋巴细胞白血病(ALL)是一种侵袭性恶性肿瘤,预后一直很差。尽管进行了大规模的国际合作试验并逐步提高了治疗强度,但在过去的 30 年中,预后并没有明显改善。通过代表性病例,我们强调了 KMT2A 重组婴儿 ALL 与 KMT2A 基因婴儿 ALL 之间的主要区别,以及分子诊断的进步如何揭示 KMT2A 基因遗传学并指导减少治疗。我们的重点是KM2TA基因重排的婴儿B细胞ALL,过去几年中出现了比传统化疗更有效、毒性更低的新型疗法。其中,双特异性 T 细胞吞噬单克隆抗体 blinatumomab 的疗效和耐受性以及自体和异体嵌合抗原受体 T 细胞疗法的使用都取得了令人鼓舞的早期数据。我们将讨论如何改进风险分层,并结合这些新药来取代目前使用的强化化疗方案中毒性最强的部分及其相关的不可接受的毒性。
{"title":"How I treat infant acute lymphoblastic leukemia.","authors":"Jack Bartram, Philip Ancliff, Ajay Vora","doi":"10.1182/blood.2023023154","DOIUrl":"10.1182/blood.2023023154","url":null,"abstract":"<p><strong>Abstract: </strong>Infant acute lymphoblastic leukemia (ALL) is an aggressive malignancy that has historically been associated with a very poor prognosis. Despite large cooperative international trials and incremental increases in intensity of therapy, there has been no significant improvement in outcome over the last 3 decades. Using representative cases, we highlight the key differences between KMT2A-rearranged and KMT2A-germ line infant ALL, and how advances in molecular diagnostics are unpicking KMT2A-germ line genetics and guiding treatment reduction. We focus on KM2TA-rearranged infant B-cell ALL for which the last few years have seen the emergence of novel therapies that both are more effective and less toxic than conventional chemotherapy. Of these, there is promising early data on the efficacy and tolerability of the bispecific T-cell engager monoclonal antibody, blinatumomab, as well as the use of autologous and allogeneic chimeric antigen receptor T-cell therapy. We discuss how we can improve risk stratification and incorporate these new agents to replace the most toxic elements of currently deployed intensive chemotherapy schedules with their associated unacceptable toxicity.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":"35-42"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141436692","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2023022893
Renske M. van ’t Oever, E. J. T. Verweij, Masja de Haas
Alloimmunization during pregnancy occurs when a mother produces antibodies against fetal antigens, leading to complications like hemolytic disease of the fetus and newborn (HDFN) and fetal and neonatal alloimmune thrombocytopenia (FNAIT). HDFN involves destruction of fetal red blood cells, potentially causing severe anemia, hydrops fetalis, and fetal death. FNAIT affects fetal platelets and possibly endothelial cells, resulting in risk of intracranial hemorrhage and brain damage. Traditional invasive methods for fetal antigen genotyping, like amniocentesis, carried miscarriage risks. The discovery of cell-free fetal DNA (cff-DNA) in maternal plasma enabled safe, noninvasive prenatal testing (NIPT). Initially used for Rhesus antigen D blood group typing, NIPT now covers various blood group antigens. Advances in technology have further enhanced the accuracy of NIPT. Despite challenges such as low cff-DNA fractions and complex genetic variations, NIPT has become essential in managing alloimmunized pregnancies. In NIPT it is important to prevent both false-positive results and false-negative results. Particularly in the coming decades, more possibilities for personalized antenatal treatment for HDFN and FNAIT cases will become apparent and accurate NIPT blood group antigen typing results are crucial for guiding clinical decisions. In this paper we describe this journey and provide practical tools for the clinic.
{"title":"How I use noninvasive prenatal testing for red blood cell and platelet antigens","authors":"Renske M. van ’t Oever, E. J. T. Verweij, Masja de Haas","doi":"10.1182/blood.2023022893","DOIUrl":"https://doi.org/10.1182/blood.2023022893","url":null,"abstract":"Alloimmunization during pregnancy occurs when a mother produces antibodies against fetal antigens, leading to complications like hemolytic disease of the fetus and newborn (HDFN) and fetal and neonatal alloimmune thrombocytopenia (FNAIT). HDFN involves destruction of fetal red blood cells, potentially causing severe anemia, hydrops fetalis, and fetal death. FNAIT affects fetal platelets and possibly endothelial cells, resulting in risk of intracranial hemorrhage and brain damage. Traditional invasive methods for fetal antigen genotyping, like amniocentesis, carried miscarriage risks. The discovery of cell-free fetal DNA (cff-DNA) in maternal plasma enabled safe, noninvasive prenatal testing (NIPT). Initially used for Rhesus antigen D blood group typing, NIPT now covers various blood group antigens. Advances in technology have further enhanced the accuracy of NIPT. Despite challenges such as low cff-DNA fractions and complex genetic variations, NIPT has become essential in managing alloimmunized pregnancies. In NIPT it is important to prevent both false-positive results and false-negative results. Particularly in the coming decades, more possibilities for personalized antenatal treatment for HDFN and FNAIT cases will become apparent and accurate NIPT blood group antigen typing results are crucial for guiding clinical decisions. In this paper we describe this journey and provide practical tools for the clinic.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"39 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142939879","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2024027053
Amar H Kelkar, Gregory A Abel
{"title":"Decoding cost-effectiveness of PNH therapies.","authors":"Amar H Kelkar, Gregory A Abel","doi":"10.1182/blood.2024027053","DOIUrl":"https://doi.org/10.1182/blood.2024027053","url":null,"abstract":"","PeriodicalId":9102,"journal":{"name":"Blood","volume":"145 1","pages":"8-10"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913266","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2024024517
Adam J Lamble, Alexandra E Kovach, Nirali N Shah
Abstract: Despite significant advancements in single-antigen targeted therapies for B-cell acute lymphoblastic leukemia (B-ALL), nonresponse and relapse persist as major challenges. Antigen escape after blinatumomab or CD19-directed chimeric antigen receptor (CAR) T cells (CD19-CAR), as CD19-negative B-ALL or lineage switch (LS) to acute myeloid leukemia, present diagnostic and treatment complexities. Given the poor outcomes for patients experiencing a postinfusion relapse, particularly those with loss of the target antigen, a strategic approach to diagnosis and treatment is imperative. In this discussion, we outline a systematic approach to managing postimmunotherapy events, categorized by CD19-positive relapse, CD19-negative relapse, and LS. We explore treatment modalities including CD19-CAR reinfusions, humanized CAR constructs, combinatorial strategies, and alternative antigen-targeted therapies, such as blinatumomab and inotuzumab. Challenges in diagnosis, particularly with antigen-escape, are addressed, highlighting the role of next-generation sequencing and multiparameter flow cytometry for myeloid marker monitoring.
尽管B细胞急性淋巴细胞白血病(B-ALL)的单一抗原靶向疗法取得了重大进展,但无应答和复发仍是主要挑战。在使用 blinatumomab 或 CD19 引导的嵌合抗原受体 T 细胞(CD19-CAR)后出现的抗原逃逸,以及 CD19 阴性 B-ALL 或向急性髓性白血病的血系转换(LS),给诊断和治疗带来了复杂性。鉴于输液后复发的患者,尤其是靶抗原缺失的患者疗效不佳,因此必须采取战略性的诊断和治疗方法。在本讨论中,我们概述了处理免疫治疗后事件的系统方法,按 CD19 阳性复发、CD19 阴性复发和 LS 分类。我们探讨的治疗模式包括 CD19-CAR 再灌注、人源化 CAR 构建物、组合策略和替代抗原靶向疗法,如 blinatumomab 和 inotuzumab。报告还探讨了诊断中的挑战,特别是抗原逃避,强调了下一代测序和多参数流式细胞术在骨髓标志物监测中的作用。
{"title":"How I treat postimmunotherapy relapsed B-ALL.","authors":"Adam J Lamble, Alexandra E Kovach, Nirali N Shah","doi":"10.1182/blood.2024024517","DOIUrl":"10.1182/blood.2024024517","url":null,"abstract":"<p><strong>Abstract: </strong>Despite significant advancements in single-antigen targeted therapies for B-cell acute lymphoblastic leukemia (B-ALL), nonresponse and relapse persist as major challenges. Antigen escape after blinatumomab or CD19-directed chimeric antigen receptor (CAR) T cells (CD19-CAR), as CD19-negative B-ALL or lineage switch (LS) to acute myeloid leukemia, present diagnostic and treatment complexities. Given the poor outcomes for patients experiencing a postinfusion relapse, particularly those with loss of the target antigen, a strategic approach to diagnosis and treatment is imperative. In this discussion, we outline a systematic approach to managing postimmunotherapy events, categorized by CD19-positive relapse, CD19-negative relapse, and LS. We explore treatment modalities including CD19-CAR reinfusions, humanized CAR constructs, combinatorial strategies, and alternative antigen-targeted therapies, such as blinatumomab and inotuzumab. Challenges in diagnosis, particularly with antigen-escape, are addressed, highlighting the role of next-generation sequencing and multiparameter flow cytometry for myeloid marker monitoring.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":"64-74"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141757122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2024025945
Surbhi Sidana, Krina K Patel, Lauren C Peres, Radhika Bansal, Mehmet H Kocoglu, Leyla Shune, Shebli Atrash, Kinaya Smith, Shonali Midha, Christopher Ferreri, Binod Dhakal, Danai Dima, Patrick Costello, Charlotte Wagner, Ran Reshef, Hitomi Hosoya, Lekha Mikkilineni, Djordje Atanackovic, Saurabh Chhabra, Ricardo Parrondo, Omar Nadeem, Hashim Mann, Nilesh Kalariya, Vanna Hovanky, Gabriel De Avila, Ciara L Freeman, Frederick L Locke, Melissa Alsina, Sandy Wong, Megan Herr, Myo Htut, Joseph McGuirk, Douglas W Sborov, Jack Khouri, Thomas Martin, Murali Janakiram, Yi Lin, Doris K Hansen
Abstract: Ciltacabtagene autoleucel (cilta-cel) was approved in 2022 for patients with relapsed/refractory multiple myeloma (RRMM). We report outcomes with cilta-cel in the standard-of-care setting. Patients with RRMM who underwent leukapheresis for cilta-cel manufacturing between 1 March 2022 and 31 December 2022 at 16 US academic medical centers were included. Overall, 255 patients underwent leukapheresis and 236 (92.5%) received cilta-cel, of which 54% would not have met CARTITUDE-1 eligibility criteria. In treated patients (N = 236), cytokine release syndrome was seen in 75% (grade ≥3, 5%), immune effector cell-associated neurotoxicity syndrome in 14% (grade ≥3, 4%), and delayed neurotoxicity in 10%. Overall and complete response rates were as follows: all patients who received cilta-cel (N = 236), 89% and 70%; patients receiving conforming cilta-cel (n = 191), 94% and 74%; and conforming cilta-cel with fludarabine/cyclophosphamide lymphodepletion (n = 152), 95% and 76%, respectively. Nonrelapse mortality was 10%, most commonly from infection. After a median follow-up of 13 months from cilta-cel, the median progression-free survival (PFS) was not reached, with 12-month estimate being 68% (95% confidence interval, 62-74). High ferritin levels, high-risk cytogenetics, and extramedullary disease were independently associated with inferior PFS, with a signal for prior B-cell maturation antigen-targeted therapy (P = .08). Second primary malignancies excluding nonmelanoma skin cancers were seen in 5.5% and myeloid malignancies/acute leukemia in 1.7%. We observed a favorable efficacy profile of standard-of-care cilta-cel in RRMM, despite more than half the patients not meeting the CARTITUDE-1 eligibility criteria.
{"title":"Safety and efficacy of standard-of-care ciltacabtagene autoleucel for relapsed/refractory multiple myeloma.","authors":"Surbhi Sidana, Krina K Patel, Lauren C Peres, Radhika Bansal, Mehmet H Kocoglu, Leyla Shune, Shebli Atrash, Kinaya Smith, Shonali Midha, Christopher Ferreri, Binod Dhakal, Danai Dima, Patrick Costello, Charlotte Wagner, Ran Reshef, Hitomi Hosoya, Lekha Mikkilineni, Djordje Atanackovic, Saurabh Chhabra, Ricardo Parrondo, Omar Nadeem, Hashim Mann, Nilesh Kalariya, Vanna Hovanky, Gabriel De Avila, Ciara L Freeman, Frederick L Locke, Melissa Alsina, Sandy Wong, Megan Herr, Myo Htut, Joseph McGuirk, Douglas W Sborov, Jack Khouri, Thomas Martin, Murali Janakiram, Yi Lin, Doris K Hansen","doi":"10.1182/blood.2024025945","DOIUrl":"10.1182/blood.2024025945","url":null,"abstract":"<p><strong>Abstract: </strong>Ciltacabtagene autoleucel (cilta-cel) was approved in 2022 for patients with relapsed/refractory multiple myeloma (RRMM). We report outcomes with cilta-cel in the standard-of-care setting. Patients with RRMM who underwent leukapheresis for cilta-cel manufacturing between 1 March 2022 and 31 December 2022 at 16 US academic medical centers were included. Overall, 255 patients underwent leukapheresis and 236 (92.5%) received cilta-cel, of which 54% would not have met CARTITUDE-1 eligibility criteria. In treated patients (N = 236), cytokine release syndrome was seen in 75% (grade ≥3, 5%), immune effector cell-associated neurotoxicity syndrome in 14% (grade ≥3, 4%), and delayed neurotoxicity in 10%. Overall and complete response rates were as follows: all patients who received cilta-cel (N = 236), 89% and 70%; patients receiving conforming cilta-cel (n = 191), 94% and 74%; and conforming cilta-cel with fludarabine/cyclophosphamide lymphodepletion (n = 152), 95% and 76%, respectively. Nonrelapse mortality was 10%, most commonly from infection. After a median follow-up of 13 months from cilta-cel, the median progression-free survival (PFS) was not reached, with 12-month estimate being 68% (95% confidence interval, 62-74). High ferritin levels, high-risk cytogenetics, and extramedullary disease were independently associated with inferior PFS, with a signal for prior B-cell maturation antigen-targeted therapy (P = .08). Second primary malignancies excluding nonmelanoma skin cancers were seen in 5.5% and myeloid malignancies/acute leukemia in 1.7%. We observed a favorable efficacy profile of standard-of-care cilta-cel in RRMM, despite more than half the patients not meeting the CARTITUDE-1 eligibility criteria.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":"85-97"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370959","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2024026887
Juan Carlos Balandrán, Iannis Aifantis
{"title":"Vitamin C deprivation puts stem cells to sleep.","authors":"Juan Carlos Balandrán, Iannis Aifantis","doi":"10.1182/blood.2024026887","DOIUrl":"https://doi.org/10.1182/blood.2024026887","url":null,"abstract":"","PeriodicalId":9102,"journal":{"name":"Blood","volume":"145 1","pages":"7-8"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913565","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-02DOI: 10.1182/blood.2024025846
Carlo Castruccio Castracani, Laura Breda, Tyler E Papp, Amaliris Guerra, Enrico Radaelli, Charles-Antoine Assenmacher, Giovanni Finesso, Barbara L Mui, Ying K Tam, Simona Fontana, Chiara Riganti, Veronica Fiorito, Sara Petrillo, Emanuela Tolosano, Hamideh Parhiz, Stefano Rivella
Abstract: X-linked sideroblastic anemia (XLSA) is a congenital anemia caused by mutations in ALAS2, a gene responsible for heme synthesis. Treatments are limited to pyridoxine supplements and blood transfusions, offering no definitive cure except for allogeneic hematopoietic stem cell transplantation, only accessible to a subset of patients. The absence of a suitable animal model has hindered the development of gene therapy research for this disease. We engineered a conditional Alas2-knockout (KO) mouse model using tamoxifen administration or treatment with lipid nanoparticles carrying Cre-mRNA and conjugated to an anti-CD117 antibody. Alas2-KOBM animals displayed a severe anemic phenotype characterized by ineffective erythropoiesis (IE), leading to low numbers of red blood cells, hemoglobin, and hematocrit. In particular, erythropoiesis in these animals showed expansion of polychromatic erythroid cells, characterized by reduced oxidative phosphorylation, mitochondria's function, and activity of key tricarboxylic acid cycle enzymes. In contrast, glycolysis was increased in the unsuccessful attempt to extend cell survival despite mitochondrial dysfunction. The IE was associated with marked splenomegaly and low hepcidin levels, leading to iron accumulation in the liver, spleen, and bone marrow and the formation of ring sideroblasts. To investigate the potential of a gene therapy approach for XLSA, we developed a lentiviral vector (X-ALAS2-LV) to direct ALAS2 expression in erythroid cells. Infusion of bone marrow (BM) cells with 0.6 to 1.4 copies of the X-ALAS2-LV in Alas2-KOBM mice improved complete blood cell levels, tissue iron accumulation, and survival rates. These findings suggest our vector could be curative in patients with XLSA.
{"title":"An erythroid-specific lentiviral vector improves anemia and iron metabolism in a new model of XLSA.","authors":"Carlo Castruccio Castracani, Laura Breda, Tyler E Papp, Amaliris Guerra, Enrico Radaelli, Charles-Antoine Assenmacher, Giovanni Finesso, Barbara L Mui, Ying K Tam, Simona Fontana, Chiara Riganti, Veronica Fiorito, Sara Petrillo, Emanuela Tolosano, Hamideh Parhiz, Stefano Rivella","doi":"10.1182/blood.2024025846","DOIUrl":"10.1182/blood.2024025846","url":null,"abstract":"<p><strong>Abstract: </strong>X-linked sideroblastic anemia (XLSA) is a congenital anemia caused by mutations in ALAS2, a gene responsible for heme synthesis. Treatments are limited to pyridoxine supplements and blood transfusions, offering no definitive cure except for allogeneic hematopoietic stem cell transplantation, only accessible to a subset of patients. The absence of a suitable animal model has hindered the development of gene therapy research for this disease. We engineered a conditional Alas2-knockout (KO) mouse model using tamoxifen administration or treatment with lipid nanoparticles carrying Cre-mRNA and conjugated to an anti-CD117 antibody. Alas2-KOBM animals displayed a severe anemic phenotype characterized by ineffective erythropoiesis (IE), leading to low numbers of red blood cells, hemoglobin, and hematocrit. In particular, erythropoiesis in these animals showed expansion of polychromatic erythroid cells, characterized by reduced oxidative phosphorylation, mitochondria's function, and activity of key tricarboxylic acid cycle enzymes. In contrast, glycolysis was increased in the unsuccessful attempt to extend cell survival despite mitochondrial dysfunction. The IE was associated with marked splenomegaly and low hepcidin levels, leading to iron accumulation in the liver, spleen, and bone marrow and the formation of ring sideroblasts. To investigate the potential of a gene therapy approach for XLSA, we developed a lentiviral vector (X-ALAS2-LV) to direct ALAS2 expression in erythroid cells. Infusion of bone marrow (BM) cells with 0.6 to 1.4 copies of the X-ALAS2-LV in Alas2-KOBM mice improved complete blood cell levels, tissue iron accumulation, and survival rates. These findings suggest our vector could be curative in patients with XLSA.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":"98-113"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142799452","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}