1001. Asciminib plus dasatinib and prednisone for Philadelphia chromosome-positive acute leukemia.
作者: Marlise R Luskin.;Mark A Murakami.;Julia Keating.;Yael Flamand.;Eric S Winer.;Jacqueline S Garcia.;Maximilian Stahl.;Richard M Stone.;Martha Wadleigh.;Stella L Jaeckle.;Ella Hagopian.;David M Weinstock.;Jessica Liegel.;Malgorzata McMasters.;Eunice S Wang.;Wendy Stock.;Daniel J DeAngelo.
来源: Blood. 2025年145卷6期577-589页
Dasatinib is an effective treatment for Philadelphia chromosome-positive (Ph+) acute leukemia, but some patients develop resistance. Combination treatment with dasatinib and asciminib, an allosteric inhibitor of BCR::ABL1, may deepen responses and prevent the emergence of dasatinib-resistant clones. In this phase 1 study (NCT03595017), 24 adults with Ph+ acute lymphoblastic leukemia (ALL; n = 22; p190, n = 16; p210, n = 6) and chronic myeloid leukemia in lymphoid blast crisis (n = 2) were treated with escalating daily doses of asciminib in combination with dasatinib 140 mg daily plus prednisone 60 mg/m2 daily to determine the maximum tolerated dose. After a 28-day induction, dasatinib and asciminib were continued indefinitely or until hematopoietic stem cell transplant. The median age was 64.5 years (range, 33-85; 50% aged ≥65 years). The recommended phase 2 dose of asciminib was 80 mg daily in combination with dasatinib and prednisone. The dose limiting toxicity at 160 mg daily was asymptomatic grade 3 pancreatic enzyme elevation without symptomatic pancreatitis. There were no vaso-occlusive events. Among patients with de novo ALL, the complete hematologic remission rates at days 28 and 84 were 84% and 100%, respectively. At day 84, 100% of patients achieved complete cytogenetic remission, 89% achieved measurable residual disease negativity (<0.01%) by multicolor flow cytometry, and 74% and 26% achieved BCR::ABL1 reverse transcription quantitative polymerase chain reaction <0.1% and <0.01%, respectively. Dual BCR::ABL1 inhibition with dasatinib and asciminib is safe with encouraging activity in patients with de novo Ph+ ALL. This trial was registered at www.clinicaltrials.gov as #NCT02081378.
1002. How I treat blastic plasmacytoid dendritic cell neoplasm.
Historically, treatment options for blastic plasmacytoid dendritic cell neoplasm (BPDCN) were limited to conventional chemotherapy, adopted from regimens used to treat acute myeloid or acute lymphoblastic leukemias, or lymphomas. Nowadays, a novel therapy targeting CD123 is available to treat BPDCN. Yet, regardless of treatment choice, achieving a first complete remission represents the main goal of therapy, because it represents the best opportunity to prolong survival in BPDCN, if offered an allogeneic hematopoietic cell transplant (allo-HCT) as consolidative therapy. Although no specific conditioning regimen is considered standard of care in allo-HCT-eligible patients, recent data from 2 large registries reported a survival advantage when offering total body irradiation-based myeloablative conditioning (MAC) regimens. Unfortunately, applicability of MAC regimens is not feasible in patients who are older/unfit, which represents a considerable proportion of patients presenting worldwide. In such cases, reduced intensity conditioning regimens represent the next best option. Autologous HCT could be considered in patients who are older/unfit who did not have bone marrow involvement at initial presentation and at time of the procedure, albeit data supporting this option are less abundant. Future research is needed to decipher the interplay between clinical, genetic, and molecular features of the disease to personalize treatment accordingly, by enhancing efficacy and avoiding unnecessary toxicities.
1003. How I diagnose and treat cardiorespiratory complications of transfusion.
Cardiorespiratory complications after blood transfusion are the leading cause of transfusion-related morbidity and mortality worldwide. Transfusion-related acute lung injury (TRALI) and transfusion-associated circulatory overload (TACO) are the 2 most frequently reported cardiorespiratory complications, both of which have clear pathophysiology-based treatment algorithms. In the past decades, translational research has increased understanding of mechanisms in place, including patient and transfusion risk factors. This has led to updated international definitions, biomarker-based diagnostics, interventions, and risk mitigation measures. Preventive measures have led to a significant reduction in TRALI, and TACO prevention is increasingly highlighted within hemovigilance. In this article, clinical case scenarios illustrate the challenges of diagnosing, treating, and finally classifying cardiorespiratory complications of transfusion. A background on current definitions, diagnostics, and pathophysiological mechanisms will be given, as well as how to deal with cases in which TRALI and TACO are both present. Hemovigilance systems worldwide are essential to provide insight into the incidence of transfusion complications. Furthermore, these systems provide a basis to discover new patient and transfusion risk factors and to better balance the down- and upside of a transfusion for a patient. Finally, we discuss the future challenges and research priorities in the field of cardiorespiratory transfusion-related complications.
1004. Safety and efficacy of standard-of-care ciltacabtagene autoleucel for relapsed/refractory multiple myeloma.
作者: 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.
来源: Blood. 2025年145卷1期85-97页
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.
1013. High-dose IV ascorbic acid therapy for patients with CCUS with TET2 mutations.
作者: Zhuoer Xie.;Jenna Fernandez.;Terra Lasho.;Christy Finke.;Michelle Amundson.;Kristen B McCullough.;Betsy R LaPlant.;Abhishek A Mangaonkar.;Naseema Gangat.;Kaaren K Reichard.;Michelle Elliott.;Thomas E Witzig.;Mrinal M Patnaik.
来源: Blood. 2024年144卷23期2456-2461页
This phase 2 trial assessed high-dose IV ascorbic acid in TET2 mutant clonal cytopenia. Eight of 10 patients were eligible for response assessment, with no responses at week 20 by International Working Group Myelodysplasia Syndromes/Neoplasms criteria. This trial was registered at www.clinicaltrials.gov as #NCT03418038.
1014. How I individualize selection of JAK inhibitors for patients with myelofibrosis.
The advent of Janus kinase inhibitors (JAKis) inaugurated a novel era in the treatment of myelofibrosis (MF), a myeloproliferative neoplasm with heterogeneous clinical manifestations. Four JAKis have been approved for intermediate or high-risk MF, in the United States. Regulatory approval of the first JAK1/2 inhibitor, ruxolitinib, in 2011, transformed the landscape of MF by markedly controlling splenomegaly and constitutional symptoms, improving patients' quality of life, and prolonging survival. Fedratinib, the second approved JAKi, is preferred in the second-line setting. Ruxolitinib and fedratinib can cause myelosuppression and are recommended for patients with the myeloproliferative phenotype. The approval of 2 less-myelosuppressive JAKis, pacritinib and momelotinib, provided essential treatment options for patients with severe thrombocytopenia and anemia, respectively. Momelotinib and pacritinib are potent activin A receptor, type 1 inhibitors with consequent significant benefits for patients with anemia. Transfusion independence was achieved with momelotinib in patients who were severely anemic, and the association of transfusion independence with prolonged overall survival was demonstrated. Judicious treatment decisions regarding JAKis can be made with in-depth understanding of the pivotal clinical trials that evaluated JAKis and their therapeutic attributes and should be guided by the dominant clinical manifestations and the type/degree of cytopenia(s) (myeloproliferative/cytopenic phenotypes). This article reviews our clinical approach to treatment with JAKis and their sequencing in patients with MF by presenting 3 clinical vignettes.
1015. Classification and risk stratification in T-lineage acute lymphoblastic leukemia.
Cure rates for patients with acute lymphoblastic leukemia (ALL) have improved markedly in recent decades, in part because of risk stratification incorporating leukemia genomics, response to treatment, and clinical features to be able to determine at diagnosis which patients are more likely to relapse or have refractory disease. Although risk stratification is well developed for patients with B-lineage ALL, it remains challenging for those with T-lineage ALL (T-ALL). Prognostic factors validated across clinical trials and real-world data in T-ALL include age, central nervous system involvement, and measurable residual disease (MRD) response. Immunophenotype, including early T-cell precursor ALL, is widely used to classify T-ALL but is not consistently associated with outcome in multivariable risk models. Historically, few genetic alterations have been consistently associated with outcome, but recent comprehensive, large-scale genomic profiling has identified multiple genetic subtypes and alterations associated with outcome independent of MRD. This review highlights ongoing efforts to identify reliable prognostic biomarkers and underscores the potential of genomics-based classification to guide future T-ALL treatment strategies.
1016. Ex vivo venetoclax sensitivity predicts clinical response in acute myeloid leukemia in the prospective VenEx trial.
作者: Sari Kytölä.;Ida Vänttinen.;Tanja Ruokoranta.;Anu Partanen.;Annasofia Holopainen.;Joseph Saad.;Milla E L Kuusisto.;Sirpa Koskela.;Riikka Räty.;Maija Itälä-Remes.;Imre Västrik.;Minna Suvela.;Alun Parsons.;Kimmo Porkka.;Krister Wennerberg.;Caroline A Heckman.;Tero Jalkanen.;Teppo Huttunen.;Pia Ettala.;Johanna Rimpiläinen.;Timo Siitonen.;Marja Pyörälä.;Heikki Kuusanmäki.;Mika Kontro.
来源: Blood. 2025年145卷4期409-421页
The B-cell lymphoma 2 inhibitor venetoclax has shown promise for treating acute myeloid leukemia (AML). However, identifying patients likely to respond remains a challenge, especially for those with relapsed/refractory (R/R) disease. We evaluated the utility of ex vivo venetoclax sensitivity testing to predict treatment responses to venetoclax-azacitidine in a prospective, multicenter, phase 2 trial. The trial recruited 104 participants with previously untreated (n = 48), R/R (n = 39), or previously treated secondary AML (sAML) (n = 17). The primary end point was complete remission or complete remission with incomplete hematologic recovery (CR/CRi) rate in ex vivo sensitive trial participants during the first 3 therapy cycles. The key secondary end points included the correlations between ex vivo drug sensitivity, responses, and survival. Venetoclax sensitivity was successfully assessed in 102 of 104 participants, with results available within a median of 3 days from sampling. In previously untreated AML, ex vivo sensitivity corresponded to an 85% (34/40) CR/CRi rate, with a median overall survival (OS) of 28.7 months, compared with 5.5 months for ex vivo resistant patients (P = .002). For R/R/sAML, ex vivo sensitivity resulted in a 62% CR/CRi rate (21/34) and median OS of 9.7 vs 3.3 months for ex vivo resistant patients (P < .001). In univariate and multivariate analysis, ex vivo venetoclax sensitivity was the strongest predictor for a favorable treatment response and survival. This trial demonstrates the feasibility of integrating ex vivo drug testing into clinical practice to identify patients with AML, particularly in the R/R setting, who benefit from venetoclax. This trial was registered at www.clinicaltrials.gov as #NCT04267081.
1017. Platelet generation in vivo and in vitro.
Platelets play crucial roles in hemostasis, thrombosis, and immunity, but our understanding of their complex biogenesis (thrombopoiesis) is currently incomplete. Deeper insight into the mechanisms of platelet biogenesis inside and outside the body is fundamental for managing hematological disorders and for the development of novel cell-based therapies. In this article, we address the current understanding of in vivo thrombopoiesis, including mechanisms of platelet generation from megakaryocytes (proplatelet formation, cytoplasmic fragmentation, and membrane budding) and their physiological location. Progress has been made in replicating these processes in vitro for potential therapeutic application, notably in platelet transfusion and bioengineering of platelets for novel targeted therapies. The current platelet-generating systems and their limitations, particularly yield, scalability, and functionality, are discussed. Finally, we highlight the current controversies and challenges in the field that need to be addressed to achieve a full understanding of these processes, in vivo and in vitro.
1018. Complement biosensors identify a classical pathway stimulus in complement-mediated thrombotic microangiopathy.
作者: Michael A Cole.;Nikhil Ranjan.;Gloria F Gerber.;Xiang-Zuo Pan.;Daniel Flores-Guerrero.;George McNamara.;Shruti Chaturvedi.;C John Sperati.;Keith R McCrae.;Robert A Brodsky.
来源: Blood. 2024年144卷24期2528-2545页
Complement-mediated thrombotic microangiopathy (CM-TMA) or hemolytic uremic syndrome, previously identified as atypical hemolytic uremic syndrome, is a TMA characterized by germ line variants or acquired antibodies to complement proteins and regulators. Building upon our prior experience with the modified Ham (mHam) assay for ex vivo diagnosis of complementopathies, we have developed an array of cell-based complement "biosensors" by selective removal of complement regulatory proteins (CD55 and CD59, CD46, or a combination thereof) in an autonomously bioluminescent HEK293 cell line. These biosensors can be used as a sensitive method for diagnosing CM-TMA and monitoring therapeutic complement blockade. Using specific complement pathway inhibitors, this model identifies immunoglobulin M (IgM)-driven classical pathway stimulus during both acute disease and in many patients during clinical remission. This provides a potential explanation for ∼50% of patients with CM-TMA who lack an alternative pathway "driving" variant and suggests at least a subset of CM-TMA is characterized by a breakdown of IgM immunologic tolerance.
1019. How I treat older patients with relapsed/refractory diffuse large B-cell lymphoma.
Diffuse large B-cell lymphoma (DLBCL) is an aggressive, yet curable, malignancy, but older patients are at higher risk of relapsed disease because they may not be eligible for full-intensity frontline chemoimmunotherapy or have comorbidities that limit standard treatments. Recent years have brought more treatment options than ever for this patient population, but it remains challenging to determine which can be safely and effectively offered to older patients. Formal determinations of fitness including geriatric assessments remain critical, but there is less guidance on how to best use this tool in the relapsed setting. Chimeric antigen receptor T-cell therapy is accessible to older patients, provided they can be supported through the intensive road to this treatment. If relapse occurs despite this or alternative therapies are preferred, many novel therapeutic options and combinations exist with some potential modifications for older adults, such as bispecific antibodies, tafasitamab and lenalidomide, polatuzumab-containing regimens, or loncastuximab tesirine. This article provides a summary of our approach to the management of this diverse population of older patients with relapsed or refractory DLBCL.
1020. How I treat sickle cell disease with gene therapy.
In 2023, 2 different gene therapies were approved for individuals with severe sickle cell disease (SCD). The small number of patients treated on the pivotal clinical trials that led to these approvals have experienced dramatic short-term reductions in the occurrence of painful vaso-occlusive crises, but the long-term safety and efficacy of these genetic therapies are yet to be ascertained. Several challenges and treatment-related concerns have emerged in regard to administering these therapies in clinical practice. This article discusses the selection and preparation of individuals with SCD who wish to receive autologous gene therapy, as well as the salient features of the care needed to support them through a long and arduous treatment process. I specifically focus on postinfusion care, as it relates to immune monitoring and infection prevention. Compared with allogeneic hematopoietic cell transplantation, delivering autologous gene therapy to an individual with SCD has distinct nuances that require awareness and special interventions. Using clinical vignettes derived from real-life patients, I provide perspectives on the complex decision-making process for gene therapy for SCD based on currently available data and make recommendations for evaluating and supporting these patients.
|