Pub Date : 2026-01-09DOI: 10.1182/bloodadvances.2025018073
Hyatt Balke-Want, Philipp Gödel, Christoph Schmid, Francis Ayuketang Ayuk, Birte Friedrichs, Pearl van Heteren, Silke Holtkamp, Gregor Zadoyan, Corinne Brillant, Joana Costa, Linda Hanssens, Tatjana Holzer, Christian Wöhle, Stefanie Biedermann, Iris Bürger, Rimas J Orentas, Toon Overstijns, Christoph Scheid, Udo Holtick, Stefan Miltenyi, Michael J Hallek, Peter Borchmann, Nadine Kutsch
Emerging long-term data indicates relapse rates of over 50% after CD19 redirected chimeric antigen receptor (CAR) T cell therapy in relapsed or refractory (r/r) B-cell non-Hodgkin lymphoma (B-NHL). To reduce selective pressure on the CD19 antigen we conducted a first-in-human phase I clinical trial of zamtocabtagene autoleucel (zamto-cel) - a non-cryopreserved tandem CD20-CD19-directed CAR-T cell therapy. Two predefined dose levels (DL1=1x106 and DL2=2.5x106 CAR+ T cells/kg body) were applied. The primary endpoint (EP) was the maximum tolerated dose (MTD). Secondary EPs included adverse events (AEs), best overall response (BOR) and biomarker assessments. A total of 12 patients, 6 per dose level were treated. No DLT and no cytokine release syndrome (CRS) or immune effector cell-associated neurotoxicity syndrome (ICANS) grade ≥3 were observed. Thus, MTD was not reached. The BOR by investigator assessment was 75% with 5/12 patients (42%) achieving complete remission (CR) until month 12 with no relapse in clinical evaluation up to 5 years after infusion. CR was associated with higher mean Cmax and detection of zamto-cel beyond month 6. Additional product characterization revealed increased expression of CD27 and CD127 along with increased expansion of CAR+ TCM cells in patients with CR, thus facilitating persistence and improved outcomes in r/r B-NHL treated with zamto-cel. Based on the promising risk-to-benefit ratio, evaluation of zamto-cel at DL2 is ongoing in pivotal Phase II clinical trials for patients with r/r aggressive B-NHL. This trial was registered at www.clinicaltrials.gov as #NCT03870945.
{"title":"Zamtocabtagene Autoleucel in Relapsed/refractory B-NHL: 5-year Follow Up of a CD20/19 tandem CAR T Cell Phase 1 Trial.","authors":"Hyatt Balke-Want, Philipp Gödel, Christoph Schmid, Francis Ayuketang Ayuk, Birte Friedrichs, Pearl van Heteren, Silke Holtkamp, Gregor Zadoyan, Corinne Brillant, Joana Costa, Linda Hanssens, Tatjana Holzer, Christian Wöhle, Stefanie Biedermann, Iris Bürger, Rimas J Orentas, Toon Overstijns, Christoph Scheid, Udo Holtick, Stefan Miltenyi, Michael J Hallek, Peter Borchmann, Nadine Kutsch","doi":"10.1182/bloodadvances.2025018073","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018073","url":null,"abstract":"<p><p>Emerging long-term data indicates relapse rates of over 50% after CD19 redirected chimeric antigen receptor (CAR) T cell therapy in relapsed or refractory (r/r) B-cell non-Hodgkin lymphoma (B-NHL). To reduce selective pressure on the CD19 antigen we conducted a first-in-human phase I clinical trial of zamtocabtagene autoleucel (zamto-cel) - a non-cryopreserved tandem CD20-CD19-directed CAR-T cell therapy. Two predefined dose levels (DL1=1x106 and DL2=2.5x106 CAR+ T cells/kg body) were applied. The primary endpoint (EP) was the maximum tolerated dose (MTD). Secondary EPs included adverse events (AEs), best overall response (BOR) and biomarker assessments. A total of 12 patients, 6 per dose level were treated. No DLT and no cytokine release syndrome (CRS) or immune effector cell-associated neurotoxicity syndrome (ICANS) grade ≥3 were observed. Thus, MTD was not reached. The BOR by investigator assessment was 75% with 5/12 patients (42%) achieving complete remission (CR) until month 12 with no relapse in clinical evaluation up to 5 years after infusion. CR was associated with higher mean Cmax and detection of zamto-cel beyond month 6. Additional product characterization revealed increased expression of CD27 and CD127 along with increased expansion of CAR+ TCM cells in patients with CR, thus facilitating persistence and improved outcomes in r/r B-NHL treated with zamto-cel. Based on the promising risk-to-benefit ratio, evaluation of zamto-cel at DL2 is ongoing in pivotal Phase II clinical trials for patients with r/r aggressive B-NHL. This trial was registered at www.clinicaltrials.gov as #NCT03870945.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942289","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 : 2026-01-08DOI: 10.1182/bloodadvances.2025018315
Daniel Wang, Manraj Singh Sra, Satoko Ito, Ding Quan Ng, Samira Glaeser-Khan, Daniel Y Wang, Sam Hirniak, Alfred I Lee, Adam Cuker, George Goshua
Iron deficiency anemia (IDA) affects nearly one-third of women globally, with heavy menstrual bleeding (HMB) being a significant risk factor among those of reproductive age. Despite the substantial burden of HMB-related IDA, patients often face delays in receiving effective treatment. Oral iron is typically used as first-line therapy, despite frequent gastrointestinal side effects and challenges in treatment adherence. This study evaluated the reproductive lifespan cost-effectiveness of first-line intravenous (IV) versus oral iron therapy for women with HMB and IDA. We developed a Markov model simulating a cohort of women from age 18 to 51, comparing first-line treatment with IV iron dextran, IV ferumoxytol, IV iron sucrose, or oral ferrous sulfate. Costs and quality-adjusted life years (QALYs) were estimated from the societal perspective. IV iron dextran was the cost-effective treatment, yielding 19.26 QALYs at a cost of $157,500, compared with 19.10 QALYs at $152,900 for oral ferrous sulfate. The incremental cost-effectiveness ratio for IV iron dextran was $28,600 per QALY. Treatment with IV ferumoxytol and iron sucrose cost $158,300 and $163,500 respectively and did not provide additional QALY benefit compared with IV iron dextran, which remained the cost-effective treatment across a range of scenarios and sensitivity analyses. Our findings indicate that first-line treatment with IV iron dextran is the cost-effective strategy for managing IDA in women with HMB at commonly accepted willingness-to-pay thresholds. These results support expanding access to IV iron as a first-line option and highlight the need to reduce treatment delays and insurance-related barriers.
{"title":"Cost-Effectiveness of First-Line IV Versus Oral Iron for Iron Deficiency Anemia in Women with Heavy Menstrual Bleeding.","authors":"Daniel Wang, Manraj Singh Sra, Satoko Ito, Ding Quan Ng, Samira Glaeser-Khan, Daniel Y Wang, Sam Hirniak, Alfred I Lee, Adam Cuker, George Goshua","doi":"10.1182/bloodadvances.2025018315","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018315","url":null,"abstract":"<p><p>Iron deficiency anemia (IDA) affects nearly one-third of women globally, with heavy menstrual bleeding (HMB) being a significant risk factor among those of reproductive age. Despite the substantial burden of HMB-related IDA, patients often face delays in receiving effective treatment. Oral iron is typically used as first-line therapy, despite frequent gastrointestinal side effects and challenges in treatment adherence. This study evaluated the reproductive lifespan cost-effectiveness of first-line intravenous (IV) versus oral iron therapy for women with HMB and IDA. We developed a Markov model simulating a cohort of women from age 18 to 51, comparing first-line treatment with IV iron dextran, IV ferumoxytol, IV iron sucrose, or oral ferrous sulfate. Costs and quality-adjusted life years (QALYs) were estimated from the societal perspective. IV iron dextran was the cost-effective treatment, yielding 19.26 QALYs at a cost of $157,500, compared with 19.10 QALYs at $152,900 for oral ferrous sulfate. The incremental cost-effectiveness ratio for IV iron dextran was $28,600 per QALY. Treatment with IV ferumoxytol and iron sucrose cost $158,300 and $163,500 respectively and did not provide additional QALY benefit compared with IV iron dextran, which remained the cost-effective treatment across a range of scenarios and sensitivity analyses. Our findings indicate that first-line treatment with IV iron dextran is the cost-effective strategy for managing IDA in women with HMB at commonly accepted willingness-to-pay thresholds. These results support expanding access to IV iron as a first-line option and highlight the need to reduce treatment delays and insurance-related barriers.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932257","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 : 2026-01-07DOI: 10.1182/bloodadvances.2025018546
Gloria Iacoboni, Emil Kyvsgaard, Víctor Navarro, David A Russler-Germain, Katharine Louise Lewis, Alejandro Martin Garcia-Sancho, Itziar Carro, Philip A Thompson, Dylan Therwhanger, David Quintela Vilchez, Matthew Ku, Pilar Gómez-Prieto, Evelyn Valencia-Espinoza, Gala Vega, Ana Jiménez-Ubieto, Mariana Beatriz Bastos Oreiro, Ángel Serna, Simon Husby, Beatriz de la Cruz Benito, Jing Luan, Sergi Camarillas, Lucía Medina, Almudena Cabero Martínez, Raúl Córdoba, Miguel Angel Canales, Juan-Manuel Manuel Sancho, Francesc Bosch, Anna Sureda, Stephen J Schuster, Chan Y Cheah, Martin Hutchings, Pere Barba, Pau Abrisqueta
{"title":"Impact of Prior Bendamustine Exposure on Bispecific Antibody Outcomes in Relapsed/Refractory Follicular Lymphoma.","authors":"Gloria Iacoboni, Emil Kyvsgaard, Víctor Navarro, David A Russler-Germain, Katharine Louise Lewis, Alejandro Martin Garcia-Sancho, Itziar Carro, Philip A Thompson, Dylan Therwhanger, David Quintela Vilchez, Matthew Ku, Pilar Gómez-Prieto, Evelyn Valencia-Espinoza, Gala Vega, Ana Jiménez-Ubieto, Mariana Beatriz Bastos Oreiro, Ángel Serna, Simon Husby, Beatriz de la Cruz Benito, Jing Luan, Sergi Camarillas, Lucía Medina, Almudena Cabero Martínez, Raúl Córdoba, Miguel Angel Canales, Juan-Manuel Manuel Sancho, Francesc Bosch, Anna Sureda, Stephen J Schuster, Chan Y Cheah, Martin Hutchings, Pere Barba, Pau Abrisqueta","doi":"10.1182/bloodadvances.2025018546","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018546","url":null,"abstract":"","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917089","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 : 2026-01-07DOI: 10.1182/bloodadvances.2025018757
Enrica Antonia Martino, Annalisa Pitino, Ernesto Vigna, Raffaella Pasquale, Isacco Ferrarini, Riccardo Moia, Andrea Visentin, Alessandro Sanna, Marina Motta, Massimo Moratti, Paolo Sportoletti, Annalisa Chiarenza, Alessandro Maggi, Valentina Zammit, Michele Merli, Idanna Innocenti, Claudia Giordano, Laura Nocilli, Massimiliano Postorino, Caterina Stelitano, Andrea Ferrario, Anna Maria Frustaci, Marcello Riva, Sara Pepe, Adalberto Ibatici, Stefania Scardino, Paola Anticoli Borza, Laura Ballotta, Salvatrice Mancuso, Francesco Malaspina, Anna Mele, Sara Galimberti, Gioacchino Catania, Annamaria Giordano, Ilaria Angeletti, Luana Schiattone, Elsa Pennese, Rosanna Maria Miccolis, Angelo Fama, Giulio Giordano, Catello Califano, Antonella Bruzzese, Santino Caserta, Giuliana Farina, Pietro Bulian, Giacomo Loseto, Barbara Pocali, Vanessa Innao, Piero Galieni, Vincenzo Fraticelli, Candida Vitale, Azzurra Romeo, Marco Rossi, Ilaria Scortechini, Federico Vozella, Luigi Malandruccolo, Marzia Varettoni, Lucia Morello, Giuseppe Pietrantuono, Esmeralda Conte, Martina Cantelli, Roberta Murru, Daniele Caracciolo, Enrico Derenzini, Valentina Di Martina, Roberto Marasca, Maria Ilaria Del Principe, Amalia Figuera, Francesco Angotzi, Marta Coscia, Nicola Di Renzo, Luca Laurenti, Nicola Amodio, Pellegrino Musto, Francesco Di Raimondo, Arcangelo Liso, Alessandra Tedeschi, Livio Trentin, Gianluca Gaidano, Francesca Romana Mauro, Giovanni Tripepi, Fortunato Morabito, Valter Gattei, Massimo Gentile
Bruton tyrosine kinase inhibitors (BTKis) have dramatically changed the therapeutic landscape of chronic lymphocytic leukemia (CLL), with ibrutinib, first-in-class, demonstrating durable efficacy even in high-risk patients. However, off-target adverse events (AEs) have raised concerns, prompting the development of more selective second-generation BTKi, as zanubrutinib, designed to improve tolerability while maintaining efficacy. Despite encouraging results from clinical trials, real-world data comparing zanubrutinib with ibrutinib remain limited. In this multicenter, retrospective study, we analyzed 934 CLL patients treated outside clinical trials, including 393 receiving zanubrutinib and 541 receiving ibrutinib. We evaluated time to treatment discontinuation (TTD) and time to next treatment or death (TTNTD) in both the overall cohort and a propensity score-matched population. Zanubrutinib-treated patients experienced lower 12-month discontinuation rates (overall:12.6% versus 21.4%; matched:12.4% versus 20.2%) and higher 12-month TTNTD rates (overall:91.9% versus 83.0%; matched:93.2% versus 83.4%). Multivariable analyses confirmed zanubrutinib as an independent predictor of longer TTD and TTNTD, while high-risk features, including age, relapsed/refractory disease, Binet stage C, TP53 disruption, ECOG 2-3, and congestive heart failure, were consistently associated with poorer outcomes. AEs leading to discontinuation, particularly atrial fibrillation, bleeding, and infections, were less frequent with zanubrutinib, reflecting its favorable safety profile. These findings provide real-world evidence that zanubrutinib offers more durable disease control and improved persistence compared with ibrutinib, reinforcing its clinical value as a preferred second-generation BTKi. Nevertheless, the relatively short follow-up for zanubrutinib warrants cautious interpretation of long-term outcomes and underscores the need for ongoing observation to fully characterize its durability and safety.
{"title":"Real-World Safety and Effectiveness of Zanubrutinib versus Ibrutinib in CLL: The CLL-ZANU2024 Italian Cohort.","authors":"Enrica Antonia Martino, Annalisa Pitino, Ernesto Vigna, Raffaella Pasquale, Isacco Ferrarini, Riccardo Moia, Andrea Visentin, Alessandro Sanna, Marina Motta, Massimo Moratti, Paolo Sportoletti, Annalisa Chiarenza, Alessandro Maggi, Valentina Zammit, Michele Merli, Idanna Innocenti, Claudia Giordano, Laura Nocilli, Massimiliano Postorino, Caterina Stelitano, Andrea Ferrario, Anna Maria Frustaci, Marcello Riva, Sara Pepe, Adalberto Ibatici, Stefania Scardino, Paola Anticoli Borza, Laura Ballotta, Salvatrice Mancuso, Francesco Malaspina, Anna Mele, Sara Galimberti, Gioacchino Catania, Annamaria Giordano, Ilaria Angeletti, Luana Schiattone, Elsa Pennese, Rosanna Maria Miccolis, Angelo Fama, Giulio Giordano, Catello Califano, Antonella Bruzzese, Santino Caserta, Giuliana Farina, Pietro Bulian, Giacomo Loseto, Barbara Pocali, Vanessa Innao, Piero Galieni, Vincenzo Fraticelli, Candida Vitale, Azzurra Romeo, Marco Rossi, Ilaria Scortechini, Federico Vozella, Luigi Malandruccolo, Marzia Varettoni, Lucia Morello, Giuseppe Pietrantuono, Esmeralda Conte, Martina Cantelli, Roberta Murru, Daniele Caracciolo, Enrico Derenzini, Valentina Di Martina, Roberto Marasca, Maria Ilaria Del Principe, Amalia Figuera, Francesco Angotzi, Marta Coscia, Nicola Di Renzo, Luca Laurenti, Nicola Amodio, Pellegrino Musto, Francesco Di Raimondo, Arcangelo Liso, Alessandra Tedeschi, Livio Trentin, Gianluca Gaidano, Francesca Romana Mauro, Giovanni Tripepi, Fortunato Morabito, Valter Gattei, Massimo Gentile","doi":"10.1182/bloodadvances.2025018757","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018757","url":null,"abstract":"<p><p>Bruton tyrosine kinase inhibitors (BTKis) have dramatically changed the therapeutic landscape of chronic lymphocytic leukemia (CLL), with ibrutinib, first-in-class, demonstrating durable efficacy even in high-risk patients. However, off-target adverse events (AEs) have raised concerns, prompting the development of more selective second-generation BTKi, as zanubrutinib, designed to improve tolerability while maintaining efficacy. Despite encouraging results from clinical trials, real-world data comparing zanubrutinib with ibrutinib remain limited. In this multicenter, retrospective study, we analyzed 934 CLL patients treated outside clinical trials, including 393 receiving zanubrutinib and 541 receiving ibrutinib. We evaluated time to treatment discontinuation (TTD) and time to next treatment or death (TTNTD) in both the overall cohort and a propensity score-matched population. Zanubrutinib-treated patients experienced lower 12-month discontinuation rates (overall:12.6% versus 21.4%; matched:12.4% versus 20.2%) and higher 12-month TTNTD rates (overall:91.9% versus 83.0%; matched:93.2% versus 83.4%). Multivariable analyses confirmed zanubrutinib as an independent predictor of longer TTD and TTNTD, while high-risk features, including age, relapsed/refractory disease, Binet stage C, TP53 disruption, ECOG 2-3, and congestive heart failure, were consistently associated with poorer outcomes. AEs leading to discontinuation, particularly atrial fibrillation, bleeding, and infections, were less frequent with zanubrutinib, reflecting its favorable safety profile. These findings provide real-world evidence that zanubrutinib offers more durable disease control and improved persistence compared with ibrutinib, reinforcing its clinical value as a preferred second-generation BTKi. Nevertheless, the relatively short follow-up for zanubrutinib warrants cautious interpretation of long-term outcomes and underscores the need for ongoing observation to fully characterize its durability and safety.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145916801","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 : 2026-01-07DOI: 10.1182/bloodadvances.2025018031
Talha Badar, Ludovica Marando, Eric Latorre-Crespo, Terra L Lasho, Francyess Denis Oliva, Chenyu Lin, Benjamin J McCormick, Mobachir El Kettani, Kashish Shah, Yael N Kusne, Omer Jamy, Kendall Diebold, Alexander Coltoff, Christy M Finke, James M Foran, Mohamed A Kharfan-Dabaja, Yao-Shan Fan, Liuyan Jiang, Rong He, Miles Thomas, Anand A Patel, David S Viswanatha, Mithun Vinod Shah, Antoine N Saliba, Abhishek A Mangaonkar, Kristina Kirschner, Aref Al-Kali, Naseema Gangat, Mark R Litzow, Mrinal M Patnaik
TP53 and PPM1D are key regulators of DNA damage response and repair and somatic mutations in these genes often co-occur in hematopoietic cells, expanding under genotoxic stress. Unlike with TP53 mutations, where mechanisms of progression are defined, pathways underlying clonal fitness and transformation in PPM1D mutant cells remain unclear. In collaboration with 5 academic institutions, we analyzed the clinical and molecular landscape of 337 patients with clonal hematopoiesis (CH) and clonal cytopenia of undetermined significance (CCUS) across four genotypes: PPM1Dmt/TP53wt (n= 170 [50%]), PPM1Dmt/TP53mt (n= 25 [7%]), TP53mt/PPM1Dwt (n=17 [5%]), and TP53wt/PPM1Dwt (n= 125 [38%]). All PPM1D variants were truncating, located in exon 6 of the gene, with a median variant allele frequency (VAF) of 6% (0.3-64%). The PPM1Dmt/TP53mt genotype was most frequently encountered in therapy related (t)CH/CCUS (80%, 66.5%, 76.5% and 19%; p= <0.001) and had a shorter time interval to detection from last genotoxic exposure (6.2, 5.9, 11.25, and 24.5 months; p= <0.001), in comparison to PPM1Dmt/TP53wt, TP53mt/PPM1Dwt and TP53wt/PPM1Dwt genotypes, respectively. Acknowledging the short follow-up duration, rates of malignant transformation were lower in PPM1Dmt/TP53wt (2%) and PPM1Dmt/TP53mt (4%) groups, in comparison to PPM1Dwt/TP53wt (12%) and PPM1Dwt/TP53mt (17%) groups (p= <0.001), respectively. In summary, PPM1D mutations are frequently observed in t-CH/CCUS, with low median VAFs and are associated with low rates of progression, even when co-mutated with TP53.
{"title":"Spectrum, prevalence, and clinical correlates of PPM1D mutations in patients with clonal hematopoiesis and clonal cytopenias.","authors":"Talha Badar, Ludovica Marando, Eric Latorre-Crespo, Terra L Lasho, Francyess Denis Oliva, Chenyu Lin, Benjamin J McCormick, Mobachir El Kettani, Kashish Shah, Yael N Kusne, Omer Jamy, Kendall Diebold, Alexander Coltoff, Christy M Finke, James M Foran, Mohamed A Kharfan-Dabaja, Yao-Shan Fan, Liuyan Jiang, Rong He, Miles Thomas, Anand A Patel, David S Viswanatha, Mithun Vinod Shah, Antoine N Saliba, Abhishek A Mangaonkar, Kristina Kirschner, Aref Al-Kali, Naseema Gangat, Mark R Litzow, Mrinal M Patnaik","doi":"10.1182/bloodadvances.2025018031","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018031","url":null,"abstract":"<p><p>TP53 and PPM1D are key regulators of DNA damage response and repair and somatic mutations in these genes often co-occur in hematopoietic cells, expanding under genotoxic stress. Unlike with TP53 mutations, where mechanisms of progression are defined, pathways underlying clonal fitness and transformation in PPM1D mutant cells remain unclear. In collaboration with 5 academic institutions, we analyzed the clinical and molecular landscape of 337 patients with clonal hematopoiesis (CH) and clonal cytopenia of undetermined significance (CCUS) across four genotypes: PPM1Dmt/TP53wt (n= 170 [50%]), PPM1Dmt/TP53mt (n= 25 [7%]), TP53mt/PPM1Dwt (n=17 [5%]), and TP53wt/PPM1Dwt (n= 125 [38%]). All PPM1D variants were truncating, located in exon 6 of the gene, with a median variant allele frequency (VAF) of 6% (0.3-64%). The PPM1Dmt/TP53mt genotype was most frequently encountered in therapy related (t)CH/CCUS (80%, 66.5%, 76.5% and 19%; p= <0.001) and had a shorter time interval to detection from last genotoxic exposure (6.2, 5.9, 11.25, and 24.5 months; p= <0.001), in comparison to PPM1Dmt/TP53wt, TP53mt/PPM1Dwt and TP53wt/PPM1Dwt genotypes, respectively. Acknowledging the short follow-up duration, rates of malignant transformation were lower in PPM1Dmt/TP53wt (2%) and PPM1Dmt/TP53mt (4%) groups, in comparison to PPM1Dwt/TP53wt (12%) and PPM1Dwt/TP53mt (17%) groups (p= <0.001), respectively. In summary, PPM1D mutations are frequently observed in t-CH/CCUS, with low median VAFs and are associated with low rates of progression, even when co-mutated with TP53.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145916833","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 : 2026-01-07DOI: 10.1182/bloodadvances.2025018724
Orly Leiva, Steven Soo, Olivia C Liu, Victor You, Andrew Stephen Palmer, Justin Abraham Kahla, Yasmeen Murtaza, Olatoyosi Odenike, Anand A Patel, Jeanne DeCara, Patrycja M Dubielecka, Max Petersen, Michelle H Lee, Joan How, Gabriela S Hobbs
Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), are chronic myeloid neoplasms associated with increased risk of cardiovascular disease (CVD). Statins are a common group of cholesterol-lower medications recommended for the primary and secondary prevention of CVD, including arterial thrombotic events. Emerging evidence suggests that statins may reduce the risk of developing MPNs and their use may be associated with improved survival. However, statins impact on cardiovascular and hematologic outcomes among patients with MPNs remains uncharacterized. We conducted a multicenter retrospective cohort study of patients with MPNs who had at least one transthoracic echocardiogram (TTE) from 2010 to 2024. Inverse-probability treatment weighting (IPTW) competing-risk regression analysis was performed to assess the association between statin use at time of index TTE on major adverse cardiovascular events (MACE), MPN disease progression, and all-cause death. MPN patients were analyzed as a whole and separately by type (ET or PV and MF). A total of 669 patients were included, 43.9% were on statin use, 50.5% were female, 83.9% were White, 78.8% had JAK2 driver mutation, and 72.9% had class I guideline indication for statin therapy. There were 267 (39.9%) PV, 234 (35.0%) ET, and 168 (25.1%) MF patients. After IPTW, statin use was associated with lower risk of MACE (SHR 0.83, 95% CI 0.70 - 0.98) but not MPN disease progression (SHR 0.96, 95% CI 0.72 - 1.29) or all-cause death (HR 1.04, 95% CI 0.87 - 1.24). Among patients with ET or PV, statin use was associated with lower risk of MACE (SHR 0.78, 95% CI 0.64 - 0.95) but not MPN progression (SHR 1.03, 95% CI 0.74 - 1.44) or all-cause death (HR 0.85, 95% CI 0.68 - 1.06). Among patients with MF, there was no difference in MACE, leukemia progression, or all-cause death. Among patients with MPNs who underwent TTE, statin use was associated with lower risk of MACE, particularly among patients with ET or PV. However, there was no association between statin use and all-cause death or MPN disease progression. Statin therapy is underutilized in this patient population. Further studies are needed to explore the utility of statin therapy in patients with MPN and identify patients who would benefit most from statin therapy.
骨髓增生性肿瘤(mpn),包括真性红细胞增多症(PV)、原发性血小板增多症(ET)和骨髓纤维化(MF),是与心血管疾病(CVD)风险增加相关的慢性髓系肿瘤。他汀类药物是一组常见的降胆固醇药物,推荐用于心血管疾病的一级和二级预防,包括动脉血栓事件。新出现的证据表明,他汀类药物可以降低发生mpn的风险,其使用可能与提高生存率有关。然而,他汀类药物对mpn患者心血管和血液学预后的影响仍未明确。我们进行了一项多中心回顾性队列研究,研究对象为2010年至2024年间至少进行过一次经胸超声心动图(TTE)检查的mpn患者。进行反概率治疗加权(IPTW)竞争风险回归分析,以评估TTE指数时他汀类药物使用与主要不良心血管事件(MACE)、MPN疾病进展和全因死亡之间的关系。对MPN患者进行整体分析,并按类型(ET或PV和MF)分别进行分析。总共纳入669例患者,43.9%的患者使用他汀类药物,50.5%为女性,83.9%为白人,78.8%患有JAK2驱动突变,72.9%患有他汀类药物治疗的I类指南指征。PV 267例(39.9%),ET 234例(35.0%),MF 168例(25.1%)。IPTW后,他汀类药物的使用与较低的MACE风险(SHR 0.83, 95% CI 0.70 - 0.98)相关,但与MPN疾病进展(SHR 0.96, 95% CI 0.72 - 1.29)或全因死亡(HR 1.04, 95% CI 0.87 - 1.24)无关。在ET或PV患者中,他汀类药物与较低的MACE风险相关(SHR 0.78, 95% CI 0.64 - 0.95),但与MPN进展(SHR 1.03, 95% CI 0.74 - 1.44)或全因死亡(HR 0.85, 95% CI 0.68 - 1.06)无关。在MF患者中,MACE、白血病进展或全因死亡没有差异。在接受TTE治疗的mpn患者中,他汀类药物的使用与较低的MACE风险相关,特别是在ET或PV患者中。然而,他汀类药物的使用与全因死亡或MPN疾病进展之间没有关联。他汀类药物治疗在这类患者中未得到充分利用。需要进一步的研究来探索他汀类药物治疗在MPN患者中的效用,并确定从他汀类药物治疗中获益最多的患者。
{"title":"Impact of Statin Use on Cardiovascular and Hematologic Outcomes Among Patients with Myeloproliferative Neoplasms.","authors":"Orly Leiva, Steven Soo, Olivia C Liu, Victor You, Andrew Stephen Palmer, Justin Abraham Kahla, Yasmeen Murtaza, Olatoyosi Odenike, Anand A Patel, Jeanne DeCara, Patrycja M Dubielecka, Max Petersen, Michelle H Lee, Joan How, Gabriela S Hobbs","doi":"10.1182/bloodadvances.2025018724","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018724","url":null,"abstract":"<p><p>Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), are chronic myeloid neoplasms associated with increased risk of cardiovascular disease (CVD). Statins are a common group of cholesterol-lower medications recommended for the primary and secondary prevention of CVD, including arterial thrombotic events. Emerging evidence suggests that statins may reduce the risk of developing MPNs and their use may be associated with improved survival. However, statins impact on cardiovascular and hematologic outcomes among patients with MPNs remains uncharacterized. We conducted a multicenter retrospective cohort study of patients with MPNs who had at least one transthoracic echocardiogram (TTE) from 2010 to 2024. Inverse-probability treatment weighting (IPTW) competing-risk regression analysis was performed to assess the association between statin use at time of index TTE on major adverse cardiovascular events (MACE), MPN disease progression, and all-cause death. MPN patients were analyzed as a whole and separately by type (ET or PV and MF). A total of 669 patients were included, 43.9% were on statin use, 50.5% were female, 83.9% were White, 78.8% had JAK2 driver mutation, and 72.9% had class I guideline indication for statin therapy. There were 267 (39.9%) PV, 234 (35.0%) ET, and 168 (25.1%) MF patients. After IPTW, statin use was associated with lower risk of MACE (SHR 0.83, 95% CI 0.70 - 0.98) but not MPN disease progression (SHR 0.96, 95% CI 0.72 - 1.29) or all-cause death (HR 1.04, 95% CI 0.87 - 1.24). Among patients with ET or PV, statin use was associated with lower risk of MACE (SHR 0.78, 95% CI 0.64 - 0.95) but not MPN progression (SHR 1.03, 95% CI 0.74 - 1.44) or all-cause death (HR 0.85, 95% CI 0.68 - 1.06). Among patients with MF, there was no difference in MACE, leukemia progression, or all-cause death. Among patients with MPNs who underwent TTE, statin use was associated with lower risk of MACE, particularly among patients with ET or PV. However, there was no association between statin use and all-cause death or MPN disease progression. Statin therapy is underutilized in this patient population. Further studies are needed to explore the utility of statin therapy in patients with MPN and identify patients who would benefit most from statin therapy.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917101","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 : 2026-01-07DOI: 10.1182/bloodadvances.2025018264
Johnny Mahlangu, Ana Boban, Maria Bruzelius, Giancarlo Castaman, Kingsley Kevin Hampton, Paul Knoebl, Aurélien Lebreton, Silvia Linari, Francisco Jose Lopez-Jaime, Camila Martins Mazini Tavares Tavares, Mohamed Salim Nekkal, Keiji Nogami, Amalie Rhode Høgh Nielsen, Amy D Shapiro, Roseline D'Oiron
Concizumab is an anti-tissue factor pathway inhibitor (TFPI) monoclonal antibody intended for once-daily subcutaneous prophylactic treatment for people with hemophilia A or B with and without inhibitors. Results from the phase 3 explorer7 study confirmed superiority of concizumab prophylaxis over no prophylaxis in reducing the annualized bleeding rate (ABR) in people with hemophilia A or B with inhibitors (HAwI/HBwI). Male patients aged ≥12 years were randomized 1:2 to no prophylaxis (group 1) or concizumab prophylaxis (group 2), or allocated to concizumab prophylaxis (groups 3 and 4). After ≥24 weeks of treatment, patients in group 1 could switch to concizumab prophylaxis. At the 56-week cut‑off (defined as when all patients in groups 2-4 had completed the visit at 56 weeks or permanently discontinued treatment), bleed-related efficacy, pharmacokinetics and pharmacodynamics, and safety were assessed. Of the 133 patients enrolled (HAwI, n=80; HBwI, n=53), 114 received concizumab prophylaxis (groups 2-4) and 19 were randomized to no prophylaxis (group 1). After ≥24 weeks, 13 patients from group 1 switched to concizumab. Median ABR for treated spontaneous and traumatic bleeding episodes in patients receiving concizumab was 0.8 (interquartile range [IQR] 0.0-3.2) at the 56‑week cut-off, consistent with the low bleeding rates (median ABR 0.0, IQR 0.0-3.3) at the 32‑week cut‑off. Concizumab and free TFPI concentration remained stable over time. No new safety concerns were reported. Longer‑term (≥1 year) efficacy and safety results of concizumab prophylaxis for HAwI/HBwI were consistent with the 32-week cut-off results in explorer7. ClinicalTrials.gov; NCT04083781.
{"title":"Concizumab in hemophilia with inhibitors: Longer-term efficacy and safety results from the phase 3 explorer7 study.","authors":"Johnny Mahlangu, Ana Boban, Maria Bruzelius, Giancarlo Castaman, Kingsley Kevin Hampton, Paul Knoebl, Aurélien Lebreton, Silvia Linari, Francisco Jose Lopez-Jaime, Camila Martins Mazini Tavares Tavares, Mohamed Salim Nekkal, Keiji Nogami, Amalie Rhode Høgh Nielsen, Amy D Shapiro, Roseline D'Oiron","doi":"10.1182/bloodadvances.2025018264","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018264","url":null,"abstract":"<p><p>Concizumab is an anti-tissue factor pathway inhibitor (TFPI) monoclonal antibody intended for once-daily subcutaneous prophylactic treatment for people with hemophilia A or B with and without inhibitors. Results from the phase 3 explorer7 study confirmed superiority of concizumab prophylaxis over no prophylaxis in reducing the annualized bleeding rate (ABR) in people with hemophilia A or B with inhibitors (HAwI/HBwI). Male patients aged ≥12 years were randomized 1:2 to no prophylaxis (group 1) or concizumab prophylaxis (group 2), or allocated to concizumab prophylaxis (groups 3 and 4). After ≥24 weeks of treatment, patients in group 1 could switch to concizumab prophylaxis. At the 56-week cut‑off (defined as when all patients in groups 2-4 had completed the visit at 56 weeks or permanently discontinued treatment), bleed-related efficacy, pharmacokinetics and pharmacodynamics, and safety were assessed. Of the 133 patients enrolled (HAwI, n=80; HBwI, n=53), 114 received concizumab prophylaxis (groups 2-4) and 19 were randomized to no prophylaxis (group 1). After ≥24 weeks, 13 patients from group 1 switched to concizumab. Median ABR for treated spontaneous and traumatic bleeding episodes in patients receiving concizumab was 0.8 (interquartile range [IQR] 0.0-3.2) at the 56‑week cut-off, consistent with the low bleeding rates (median ABR 0.0, IQR 0.0-3.3) at the 32‑week cut‑off. Concizumab and free TFPI concentration remained stable over time. No new safety concerns were reported. Longer‑term (≥1 year) efficacy and safety results of concizumab prophylaxis for HAwI/HBwI were consistent with the 32-week cut-off results in explorer7. ClinicalTrials.gov; NCT04083781.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917071","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 : 2026-01-07DOI: 10.1182/bloodadvances.2025017751
Saurabh Zanwar, Jithma P Abeykoon, Shirley D'Sa, Damien Roos-Weil, Dirk R Larson, Colin L Colby, Eric Durot, Efstathios Kastritis, Encarl Uppal, Oliver Tomkins, Pierre Morel, Patrizia Mondello, Lydia Montes, Jonas Paludo, Sikander Ailawadhi, Shayna Sarosiek, Olabisi Ogunbiyi, Pascalle Cornillet-Lefebvre, S Vincent Rajkumar, Anne Quinquenel, Angela Dispenzieri, Rafael Fonseca, Morie A Gertz, Shaji K Kumar, Meletios Athanasios Dimopoulos, Stephen M Ansell, Steven P Treon, Jorge J Castillo, Prashant Kapoor
Waldenström macroglobulinemia (WM) is characterized by recurrent MYD88 and CXCR4 mutations, whose prognostic value in chemoimmunotherapy-treated patients remains unclear. Moreover, the typically prolonged progression-free survival (PFS) correlates inconsistently with overall survival (OS), underscoring the importance of examining other surrogates. Progression of disease within 24 months (POD24), an established early endpoint, delineates functionally high-risk patients in other indolent lymphomas. This international study evaluated 253 patients receiving frontline fixed-duration bendamustine-rituximab (BR), a common chemoimmunotherapy for WM. At median follow-up of 5.9 years, 5-year PFS and OS were 65% and 87%, respectively; 5-year PFS was similar between MYD88L265P (90%) and MYD88wild-type (WT) subcohorts (64% each, p=0.4). Among 89 patients with known CXCR4 status, the subcohort with CXCR4mutation (28%) had shorter PFS (median, 3.3 versus 8.8 years; HR 2.8, p=0.0036) and OS (HR 2.6, p=0.036) compared to CXCR4WT. POD24 occurred in 11.5% of patients who demonstrated inferior subsequent OS (5-year OS: 71% versus 86%; HR 3.1, p=0.005) and higher mortality (SMR 3.7), unlike the non-POD24 group, whose mortality was comparable to the matched general population (SMR 1.1). In conclusion, BR is effective, irrespective of the MYD88 status, but CXCR4 mutations and POD24 portend worse outcomes. Non-POD24 patients represent a cohort with distinctly favorable outcome.
{"title":"POD24 is a Novel Determinant of Prognosis in Patients with Waldenström Macroglobulinemia.","authors":"Saurabh Zanwar, Jithma P Abeykoon, Shirley D'Sa, Damien Roos-Weil, Dirk R Larson, Colin L Colby, Eric Durot, Efstathios Kastritis, Encarl Uppal, Oliver Tomkins, Pierre Morel, Patrizia Mondello, Lydia Montes, Jonas Paludo, Sikander Ailawadhi, Shayna Sarosiek, Olabisi Ogunbiyi, Pascalle Cornillet-Lefebvre, S Vincent Rajkumar, Anne Quinquenel, Angela Dispenzieri, Rafael Fonseca, Morie A Gertz, Shaji K Kumar, Meletios Athanasios Dimopoulos, Stephen M Ansell, Steven P Treon, Jorge J Castillo, Prashant Kapoor","doi":"10.1182/bloodadvances.2025017751","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025017751","url":null,"abstract":"<p><p>Waldenström macroglobulinemia (WM) is characterized by recurrent MYD88 and CXCR4 mutations, whose prognostic value in chemoimmunotherapy-treated patients remains unclear. Moreover, the typically prolonged progression-free survival (PFS) correlates inconsistently with overall survival (OS), underscoring the importance of examining other surrogates. Progression of disease within 24 months (POD24), an established early endpoint, delineates functionally high-risk patients in other indolent lymphomas. This international study evaluated 253 patients receiving frontline fixed-duration bendamustine-rituximab (BR), a common chemoimmunotherapy for WM. At median follow-up of 5.9 years, 5-year PFS and OS were 65% and 87%, respectively; 5-year PFS was similar between MYD88L265P (90%) and MYD88wild-type (WT) subcohorts (64% each, p=0.4). Among 89 patients with known CXCR4 status, the subcohort with CXCR4mutation (28%) had shorter PFS (median, 3.3 versus 8.8 years; HR 2.8, p=0.0036) and OS (HR 2.6, p=0.036) compared to CXCR4WT. POD24 occurred in 11.5% of patients who demonstrated inferior subsequent OS (5-year OS: 71% versus 86%; HR 3.1, p=0.005) and higher mortality (SMR 3.7), unlike the non-POD24 group, whose mortality was comparable to the matched general population (SMR 1.1). In conclusion, BR is effective, irrespective of the MYD88 status, but CXCR4 mutations and POD24 portend worse outcomes. Non-POD24 patients represent a cohort with distinctly favorable outcome.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145916505","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}
Myeloproliferative neoplasms (MPN) are associated with a high symptom burden and impaired quality of life (QoL), often unaided by available treatments. Physical activity has demonstrated benefits in other cancers; however, its potential has yet to be explored in MPN. This randomized controlled trial aimed to evaluate the feasibility, acceptability, and efficacy of a supervised exercise program in patients with MPN based on longitudinal assessment of symptom burden, QoL, and clinical/inflammatory markers (CRP, ESR, ferritin, LDH, serum cytokines). MPN patients (n=55) were randomized 3:2 to a 12-week home-based exercise intervention (flexibility/resistance/aerobics) supervised by a kinesiologist versus waitlist control. Measures included patient-reported outcome questionnaires, peripheral blood sampling and post-intervention interviews with participants. Forty-seven patients completed the trial (FIT, n=26; control, n=21). Median [range] age was 66 years [27-86]; 60% were female. All feasibility benchmarks were met, with 88% of participants satisfied and 92% with intent to continue an exercise program on a regular basis. A significant reduction in lactate dehydrogenase levels (LDH) was observed in FIT patients compared to controls (-14.5 U/L vs +4.0 U/L; p=0.03). Patients interviewed described positive effects of the intervention on symptoms and would unanimously recommend the program to others with MPN. In this pilot study, supervised exercise was feasible, acceptable, and showed potential benefits on inflammatory/disease markers.
骨髓增生性肿瘤(MPN)与高症状负担和生活质量受损(QoL)相关,通常没有可用的治疗方法。体育锻炼已被证明对其他癌症有益;然而,它在MPN中的潜力还有待探索。本随机对照试验旨在通过对症状负担、生活质量和临床/炎症标志物(CRP、ESR、铁蛋白、LDH、血清细胞因子)的纵向评估,评估MPN患者进行监督运动计划的可行性、可接受性和有效性。MPN患者(n=55)以3:2的比例随机分为两组,一组由运动学家监督进行为期12周的家庭运动干预(柔韧性/阻力/有氧),另一组为候补组。措施包括患者报告的结果问卷,外周血采样和干预后对参与者的访谈。47例患者完成了试验(FIT, n=26; control, n=21)。年龄中位数为66岁[27-86岁];60%是女性。所有的可行性基准都达到了,88%的参与者满意,92%的参与者打算继续定期锻炼计划。与对照组相比,FIT患者乳酸脱氢酶(LDH)水平显著降低(-14.5 U/L vs +4.0 U/L; p=0.03)。接受采访的患者描述了干预对症状的积极影响,并一致向其他MPN患者推荐该方案。在这项初步研究中,有监督的运动是可行的、可接受的,并显示出对炎症/疾病标志物的潜在益处。
{"title":"MPN-FIT: a randomized controlled pilot trial of supervised exercise in myeloproliferative neoplasms.","authors":"Marie Ouellet, Angelo Rizzolo, Caroline Venne, Hanane Moussa, Karine Bilodeau, Luigina Mollica, Geneviève Chabot-Roy, Geneviève Huynh-Trudeau, Sylvie Lesage, Michaël Harnois, Lambert Busque, Shireen Sirhan, Natasha Szuber","doi":"10.1182/bloodadvances.2025018682","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025018682","url":null,"abstract":"<p><p>Myeloproliferative neoplasms (MPN) are associated with a high symptom burden and impaired quality of life (QoL), often unaided by available treatments. Physical activity has demonstrated benefits in other cancers; however, its potential has yet to be explored in MPN. This randomized controlled trial aimed to evaluate the feasibility, acceptability, and efficacy of a supervised exercise program in patients with MPN based on longitudinal assessment of symptom burden, QoL, and clinical/inflammatory markers (CRP, ESR, ferritin, LDH, serum cytokines). MPN patients (n=55) were randomized 3:2 to a 12-week home-based exercise intervention (flexibility/resistance/aerobics) supervised by a kinesiologist versus waitlist control. Measures included patient-reported outcome questionnaires, peripheral blood sampling and post-intervention interviews with participants. Forty-seven patients completed the trial (FIT, n=26; control, n=21). Median [range] age was 66 years [27-86]; 60% were female. All feasibility benchmarks were met, with 88% of participants satisfied and 92% with intent to continue an exercise program on a regular basis. A significant reduction in lactate dehydrogenase levels (LDH) was observed in FIT patients compared to controls (-14.5 U/L vs +4.0 U/L; p=0.03). Patients interviewed described positive effects of the intervention on symptoms and would unanimously recommend the program to others with MPN. In this pilot study, supervised exercise was feasible, acceptable, and showed potential benefits on inflammatory/disease markers.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145916445","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 : 2025-12-24DOI: 10.1182/bloodadvances.2025017795
Shyam Ajay Patel, Steven M Devine, Jan Cerny
{"title":"A Proposal for Expedited Stem Cell Transplantation for TP53-Mutant Myelodysplastic Neoplasms and Acute Myeloid Leukemia.","authors":"Shyam Ajay Patel, Steven M Devine, Jan Cerny","doi":"10.1182/bloodadvances.2025017795","DOIUrl":"https://doi.org/10.1182/bloodadvances.2025017795","url":null,"abstract":"","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":" ","pages":""},"PeriodicalIF":7.1,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145817902","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}