Pub Date : 2025-01-08DOI: 10.1182/blood.2024026310
Timothy P. Hughes, Agnes S. M. Yong, David M. Ross
One of the most remarkable achievements of the tyrosine kinase inhibitor (TKI) era has been the capacity to induce deep molecular remissions that are sustainable off therapy in patients with chronic myeloid leukemia (CML), that is, treatment-free remission (TFR). TFR was first described in a handful of patients within 3 to 4 years of imatinib approval. In 2004, TFR was tested in a small French pilot study, followed soon after by the French STIM and Australasian TWISTER studies. These early trials demonstrated that TFR was achievable but also showed that rapid relapse was equally likely. Perhaps the most critical observation was that relapsing patients could be rapidly and safely returned to deep molecular remission after restarting therapy, minimizing the risk associated with TFR attempts. Consensus criteria for TFR eligibility were established soon after those studies were reported. Over the past decade, TFR criteria have been broadened, key predictive markers of success identified, and overall safety of TFR in the wider clinical community confirmed. Despite this progress, TFR is still only achieved in a fraction of patients with CML globally. Over the next decade, the focus will be making TFR the mainstream pathway for as many patients as possible as well as scaling back the duration of therapy required. More potent, better targeted TKIs, and immune modulation will likely have a significant impact. Predictive assays should enable most patients who attempt TFR to do so with a high probability of success. Ultimately TFR should be seen as the first step on an ambitious pathway toward cure for patients with CML.
{"title":"The evolution of treatment-free remission","authors":"Timothy P. Hughes, Agnes S. M. Yong, David M. Ross","doi":"10.1182/blood.2024026310","DOIUrl":"https://doi.org/10.1182/blood.2024026310","url":null,"abstract":"One of the most remarkable achievements of the tyrosine kinase inhibitor (TKI) era has been the capacity to induce deep molecular remissions that are sustainable off therapy in patients with chronic myeloid leukemia (CML), that is, treatment-free remission (TFR). TFR was first described in a handful of patients within 3 to 4 years of imatinib approval. In 2004, TFR was tested in a small French pilot study, followed soon after by the French STIM and Australasian TWISTER studies. These early trials demonstrated that TFR was achievable but also showed that rapid relapse was equally likely. Perhaps the most critical observation was that relapsing patients could be rapidly and safely returned to deep molecular remission after restarting therapy, minimizing the risk associated with TFR attempts. Consensus criteria for TFR eligibility were established soon after those studies were reported. Over the past decade, TFR criteria have been broadened, key predictive markers of success identified, and overall safety of TFR in the wider clinical community confirmed. Despite this progress, TFR is still only achieved in a fraction of patients with CML globally. Over the next decade, the focus will be making TFR the mainstream pathway for as many patients as possible as well as scaling back the duration of therapy required. More potent, better targeted TKIs, and immune modulation will likely have a significant impact. Predictive assays should enable most patients who attempt TFR to do so with a high probability of success. Ultimately TFR should be seen as the first step on an ambitious pathway toward cure for patients with CML.","PeriodicalId":9102,"journal":{"name":"Blood","volume":"93 1","pages":""},"PeriodicalIF":20.3,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142987893","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 : 2025-01-08DOI: 10.1182/blood.2024026515
Brian J Druker
No abstract provided.
{"title":"Introduction to a review series on reflections on a quarter century of TKIs in CML.","authors":"Brian J Druker","doi":"10.1182/blood.2024026515","DOIUrl":"https://doi.org/10.1182/blood.2024026515","url":null,"abstract":"<p><p>No abstract provided.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":21.0,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142944659","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 : 2025-01-07DOI: 10.1182/blood.2024025154
Mark J Levis, Mehdi Hamadani, Brent R Logan, Richard J Jones, Anurag K Singh, Mark R Litzow, John R Wingard, Esperanza B Papadopoulos, Alexander E Perl, Robert J Soiffer, Celalettin Ustun, Masumi Ueda Oshima, Geoffrey L Uy, Edmund K Waller, Sumithira Vasu, Melhem M Solh, Asmita Mishra, Lori Muffly, Hee-Je Kim, Matthias Stelljes, Yuho Najima, Masahiro Onozawa, Kirsty J Thomson, Arnon Nagler, Andrew H Wei, Guido Marcucci, Caroline Chen, Nahla Hasabou, Matt Rosales, Jason E Hill, Stanley C Gill, Rishita Nuthethi, Denise King, Adam M Mendizabal, Steven M Devine, Mary M Horowitz, Yi-Bin Chen
BMT CTN 1506 ("MORPHO"; NCT02997202) was a randomized phase 3 study of gilteritinib compared to placebo as maintenance therapy after hematopoietic stem cell transplantation (HCT) for patients with FLT3-ITD-mutated acute myeloid leukemia (AML). A key secondary endpoint was to determine the impact on survival of pre- and/or post-HCT measurable residual disease (MRD), as determined using a highly sensitive assay for FLT3-ITD mutations. Generally, gilteritinib maintenance therapy was associated with improved relapse-free survival (RFS) for participants with detectable peri-HCT MRD, whereas no benefit was evident for those lacking detectable MRD. We conducted a post-hoc analysis of the data and found that the level of MRD detected with this approach correlated remarkably with RFS and relapse risk, and that MRD detectable at any level negatively impacted RFS. In the placebo arm, 42.2% of participants with detectable FLT3-ITD MRD relapsed compared to 13.4% of those without detectable MRD. We found that 14.8% of participants had multiple FLT3-ITD clones detected as MRD and had worse survival irrespective of treatment arm. Finally, we examined the kinetics of FLT3-ITD clonal relapse or eradication and found that participants on the placebo arm with detectable MRD relapsed rapidly after HCT, often within a few weeks. MRD-positive participants on the gilteritinib arm relapsed either with FLT3 wild type clones (as assessed by capillary electrophoresis), after cessation of gilteritinib with persistent MRD, or on progression of multi-clonal disease. These data demonstrate the potential of using FLT3-ITD MRD to guide therapy with gilteritinib for this subtype of AML.
{"title":"Measurable residual disease and posttransplantation gilteritinib maintenance for patients with FLT3-ITD-mutated AML.","authors":"Mark J Levis, Mehdi Hamadani, Brent R Logan, Richard J Jones, Anurag K Singh, Mark R Litzow, John R Wingard, Esperanza B Papadopoulos, Alexander E Perl, Robert J Soiffer, Celalettin Ustun, Masumi Ueda Oshima, Geoffrey L Uy, Edmund K Waller, Sumithira Vasu, Melhem M Solh, Asmita Mishra, Lori Muffly, Hee-Je Kim, Matthias Stelljes, Yuho Najima, Masahiro Onozawa, Kirsty J Thomson, Arnon Nagler, Andrew H Wei, Guido Marcucci, Caroline Chen, Nahla Hasabou, Matt Rosales, Jason E Hill, Stanley C Gill, Rishita Nuthethi, Denise King, Adam M Mendizabal, Steven M Devine, Mary M Horowitz, Yi-Bin Chen","doi":"10.1182/blood.2024025154","DOIUrl":"https://doi.org/10.1182/blood.2024025154","url":null,"abstract":"<p><p>BMT CTN 1506 (\"MORPHO\"; NCT02997202) was a randomized phase 3 study of gilteritinib compared to placebo as maintenance therapy after hematopoietic stem cell transplantation (HCT) for patients with FLT3-ITD-mutated acute myeloid leukemia (AML). A key secondary endpoint was to determine the impact on survival of pre- and/or post-HCT measurable residual disease (MRD), as determined using a highly sensitive assay for FLT3-ITD mutations. Generally, gilteritinib maintenance therapy was associated with improved relapse-free survival (RFS) for participants with detectable peri-HCT MRD, whereas no benefit was evident for those lacking detectable MRD. We conducted a post-hoc analysis of the data and found that the level of MRD detected with this approach correlated remarkably with RFS and relapse risk, and that MRD detectable at any level negatively impacted RFS. In the placebo arm, 42.2% of participants with detectable FLT3-ITD MRD relapsed compared to 13.4% of those without detectable MRD. We found that 14.8% of participants had multiple FLT3-ITD clones detected as MRD and had worse survival irrespective of treatment arm. Finally, we examined the kinetics of FLT3-ITD clonal relapse or eradication and found that participants on the placebo arm with detectable MRD relapsed rapidly after HCT, often within a few weeks. MRD-positive participants on the gilteritinib arm relapsed either with FLT3 wild type clones (as assessed by capillary electrophoresis), after cessation of gilteritinib with persistent MRD, or on progression of multi-clonal disease. These data demonstrate the potential of using FLT3-ITD MRD to guide therapy with gilteritinib for this subtype of AML.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":21.0,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142944581","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 : 2025-01-07DOI: 10.1182/blood.2024024846
Sivasubramanian Baskar, Haiyong Peng, Erika M Gaglione, Elizabeth J Carstens, Margaret A Lindorfer, Inhye E Ahn, Sarah E M Herman, Martin Skarzynski, Jing Chang, Keyvan Keyvanfar, Vicent Butera, Amy Z Blackburn, Berengere Vire, Irina Maric, Maryalice Stetler-Stevenson, Constance M Yuan, Michael A Eckhaus, Susan Soto, Mohammed Z H Farooqui, Ronald P Taylor, Christoph Rader, Adrian Wiestner
Monoclonal antibodies (mAbs) improve survival of patients with mature B-cell malignancies. Fcγ-receptor dependent effector mechanisms kill tumor cells but can promote antigen loss through trogocytosis, contributing to treatment failures. Cell-bound mAbs trigger the complement cascade to deposit C3 activation fragments and lyse cells. Within 24 hours after ofatumumab administration to patients with chronic lymphocytic leukemia (CLL), circulating tumor cells had lost CD20 and were opsonized with C3d. We hypothesized that C3d provides a target to eliminate residual CD20 negative tumor cells. To test this hypothesis, we generated C8xi, a mouse/human chimeric IgG1 that reacts with human but not mouse C3d. C8xi was effective in a patient-derived xenograft model against CD20 negative, C3d opsonized CLL cells from patients treated with ofatumumab. We also generated rabbit mAbs, two of which were chosen because they bound mouse and human C3d with low nanomolar affinity but were minimally cross-reactive with full-length C3. Anti-C3d rabbit/human chimeric IgG1 in combination with ofatumumab or rituximab prolonged survival of xenografted mice that model three different types of non-Hodgkin lymphoma (NHL). For example, in a diffuse large B-cell lymphoma model (SU-DHL-6), median survival with single-agent CD20 mAb was 114 days but was not reached for mAb combination treatment (P=.008). In another NHL model (SU-DHL-4), single-agent and combination mAb therapy eradicated lymphoma in most mice. In long-term survivors from both cohorts, there was no evidence of adverse effects. We propose that C3d mAbs combined with complement fixing CD20 mAbs can deliver a one-two punch and increase efficacy of mAb-based therapy.
{"title":"Potentiating CD20 monoclonal antibody therapy by targeting complement C3 fragment covalently deposited on lymphoma cells.","authors":"Sivasubramanian Baskar, Haiyong Peng, Erika M Gaglione, Elizabeth J Carstens, Margaret A Lindorfer, Inhye E Ahn, Sarah E M Herman, Martin Skarzynski, Jing Chang, Keyvan Keyvanfar, Vicent Butera, Amy Z Blackburn, Berengere Vire, Irina Maric, Maryalice Stetler-Stevenson, Constance M Yuan, Michael A Eckhaus, Susan Soto, Mohammed Z H Farooqui, Ronald P Taylor, Christoph Rader, Adrian Wiestner","doi":"10.1182/blood.2024024846","DOIUrl":"https://doi.org/10.1182/blood.2024024846","url":null,"abstract":"<p><p>Monoclonal antibodies (mAbs) improve survival of patients with mature B-cell malignancies. Fcγ-receptor dependent effector mechanisms kill tumor cells but can promote antigen loss through trogocytosis, contributing to treatment failures. Cell-bound mAbs trigger the complement cascade to deposit C3 activation fragments and lyse cells. Within 24 hours after ofatumumab administration to patients with chronic lymphocytic leukemia (CLL), circulating tumor cells had lost CD20 and were opsonized with C3d. We hypothesized that C3d provides a target to eliminate residual CD20 negative tumor cells. To test this hypothesis, we generated C8xi, a mouse/human chimeric IgG1 that reacts with human but not mouse C3d. C8xi was effective in a patient-derived xenograft model against CD20 negative, C3d opsonized CLL cells from patients treated with ofatumumab. We also generated rabbit mAbs, two of which were chosen because they bound mouse and human C3d with low nanomolar affinity but were minimally cross-reactive with full-length C3. Anti-C3d rabbit/human chimeric IgG1 in combination with ofatumumab or rituximab prolonged survival of xenografted mice that model three different types of non-Hodgkin lymphoma (NHL). For example, in a diffuse large B-cell lymphoma model (SU-DHL-6), median survival with single-agent CD20 mAb was 114 days but was not reached for mAb combination treatment (P=.008). In another NHL model (SU-DHL-4), single-agent and combination mAb therapy eradicated lymphoma in most mice. In long-term survivors from both cohorts, there was no evidence of adverse effects. We propose that C3d mAbs combined with complement fixing CD20 mAbs can deliver a one-two punch and increase efficacy of mAb-based therapy.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":21.0,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142944603","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}
The maintenance of cellular redox balance is crucial for cell survival and homeostasis and is disrupted with aging. Selenoproteins, comprising essential antioxidant enzymes, raise intriguing questions about their involvement in hematopoietic aging and potential reversibility. Motivated by our observation of mRNA downregulation of key antioxidant selenoproteins in aged human hematopoietic stem cells (HSCs) and previous findings of increased lipid peroxidation in aged hematopoiesis, we employed tRNASec gene (Trsp) knockout (KO) mouse model to simulate disrupted selenoprotein synthesis. This revealed insights into the protective roles of selenoproteins in preserving HSC stemness and B-lineage maturation, despite negligible effects on myeloid cells. Notably, Trsp KO exhibited B lymphocytopenia and reduced HSCs' self-renewal capacity, recapitulating certain aspects of aged phenotypes, along with the upregulation of aging-related genes in both HSCs and pre-B cells. While Trsp KO activated an antioxidant response transcription factor NRF2, we delineated a lineage-dependent phenotype driven by lipid peroxidation, which was exacerbated with aging yet ameliorated by ferroptosis inhibitors such as vitamin E. Interestingly, the myeloid genes were ectopically expressed in pre-B cells of Trsp KO mice, and KO pro-B/pre-B cells displayed differentiation potential toward functional CD11b+ fraction in the transplant model, suggesting that disrupted selenoprotein synthesis induces the potential of B-to-myeloid switch. Given the similarities between the KO model and aged wild-type mice, including ferroptosis vulnerability, impaired HSC self-renewal and B-lineage maturation, and characteristic lineage switch, our findings underscore the critical role of selenoprotein-mediated redox regulation in maintaining balanced hematopoiesis and suggest the preventive potential of selenoproteins against aging-related alterations.
{"title":"Selenoprotein-Mediated Redox Regulation Shapes the Cell Fate of HSCs and Mature Lineages.","authors":"Yumi Aoyama, Hiromi Yamazaki, Koutarou Nishimua, Masaki Nomura, Tsukasa Shigehiro, Takafumi Suzuki, Weijia Zang, Yota Tatara, Hiromi Ito, Yasutaka Hayashi, Yui Koike, Miki Fukumoto, Atsushi Tanaka, Yifan Zhang, Wataru Saika, Chihiro Hasegawa, Shuya Kasai, Yingyi Kong, Yohei Minakuchi, Ken Itoh, Masayuki Yamamoto, Shinya Toyokuni, Atsushi Toyoda, Tomokatsu Ikawa, Akifumi Takaori-Kondo, Daichi Inoue","doi":"10.1182/blood.2024025402","DOIUrl":"https://doi.org/10.1182/blood.2024025402","url":null,"abstract":"<p><p>The maintenance of cellular redox balance is crucial for cell survival and homeostasis and is disrupted with aging. Selenoproteins, comprising essential antioxidant enzymes, raise intriguing questions about their involvement in hematopoietic aging and potential reversibility. Motivated by our observation of mRNA downregulation of key antioxidant selenoproteins in aged human hematopoietic stem cells (HSCs) and previous findings of increased lipid peroxidation in aged hematopoiesis, we employed tRNASec gene (Trsp) knockout (KO) mouse model to simulate disrupted selenoprotein synthesis. This revealed insights into the protective roles of selenoproteins in preserving HSC stemness and B-lineage maturation, despite negligible effects on myeloid cells. Notably, Trsp KO exhibited B lymphocytopenia and reduced HSCs' self-renewal capacity, recapitulating certain aspects of aged phenotypes, along with the upregulation of aging-related genes in both HSCs and pre-B cells. While Trsp KO activated an antioxidant response transcription factor NRF2, we delineated a lineage-dependent phenotype driven by lipid peroxidation, which was exacerbated with aging yet ameliorated by ferroptosis inhibitors such as vitamin E. Interestingly, the myeloid genes were ectopically expressed in pre-B cells of Trsp KO mice, and KO pro-B/pre-B cells displayed differentiation potential toward functional CD11b+ fraction in the transplant model, suggesting that disrupted selenoprotein synthesis induces the potential of B-to-myeloid switch. Given the similarities between the KO model and aged wild-type mice, including ferroptosis vulnerability, impaired HSC self-renewal and B-lineage maturation, and characteristic lineage switch, our findings underscore the critical role of selenoprotein-mediated redox regulation in maintaining balanced hematopoiesis and suggest the preventive potential of selenoproteins against aging-related alterations.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":21.0,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142944611","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 : 2025-01-07DOI: 10.1182/blood.2024026482
Yang-Yang Ding, Jonathan H Sussman, Kellyn Madden, Joseph P Loftus, Robert K Chen, Catherine D Falkenstein, Diego Alberto Barcenas Lopez, David A Hottman, Benjamin Mathier, Wenbao Yu, Jason Xu, Changya Chen, Chia-Hui Chen, Bing He, Shovik Bandyopadhyay, Zhan Zhang, DongGeun Lee, Hong Wang, Junmin Peng, Chi V Dang, Kai Tan, Sarah K Tasian
Philadelphia chromosome-like B-cell acute lymphoblastic leukemia (Ph-like ALL) is driven by genetic alterations that induce constitutive kinase signaling and is associated with chemoresistance and high relapse risk in children and adults. Preclinical studies in the most common CRLF2-rearranged/JAK pathway-activated Ph-like ALL subtype have shown variable responses to JAK inhibitor-based therapies, suggesting incomplete oncogene addiction and highlighting a need to elucidate alternative biologic dependencies and therapeutic vulnerabilities, while the ABL-class Ph-like ALL subtype appears preferentially sensitive to SRC/ABL- or PDGFRB-targeting inhibitors. Which patients may be responsive versus resistant to tyrosine kinase inhibitor (TKI)-based precision medicine approaches remains a critical knowledge gap. Using bulk and single-cell multiomics analyses, we profiled residual cells from CRLF2-rearranged or ABL1-rearranged Ph-like ALL patient-derived xenograft models treated in vivo with targeted inhibitors to identify TKI-resistant subpopulations and potential mechanisms of therapeutic escape. We detected a specific MYC dependency in Ph-like ALL cells and defined a new leukemia cell subpopulation with senescence-associated stem cell-like features regulated by AP-1 transcription factors. This dormant ALL subpopulation was effectively eradicated by dual pharmacologic inhibition of BCL-2 and JAK/STAT or SRC/ABL pathways, a clinically-relevant therapeutic strategy. Single cell-derived molecular signatures of this senescence and stem/progenitor-like subpopulation further predicted poor clinical outcomes associated with other high-risk genetic subtypes of childhood B-ALL and thus may have broader prognostic applicability beyond Ph-like ALL.
{"title":"Targeting Senescent Stemlike Subpopulations in Philadelphia Chromosome-Like Acute Lymphoblastic Leukemia.","authors":"Yang-Yang Ding, Jonathan H Sussman, Kellyn Madden, Joseph P Loftus, Robert K Chen, Catherine D Falkenstein, Diego Alberto Barcenas Lopez, David A Hottman, Benjamin Mathier, Wenbao Yu, Jason Xu, Changya Chen, Chia-Hui Chen, Bing He, Shovik Bandyopadhyay, Zhan Zhang, DongGeun Lee, Hong Wang, Junmin Peng, Chi V Dang, Kai Tan, Sarah K Tasian","doi":"10.1182/blood.2024026482","DOIUrl":"https://doi.org/10.1182/blood.2024026482","url":null,"abstract":"<p><p>Philadelphia chromosome-like B-cell acute lymphoblastic leukemia (Ph-like ALL) is driven by genetic alterations that induce constitutive kinase signaling and is associated with chemoresistance and high relapse risk in children and adults. Preclinical studies in the most common CRLF2-rearranged/JAK pathway-activated Ph-like ALL subtype have shown variable responses to JAK inhibitor-based therapies, suggesting incomplete oncogene addiction and highlighting a need to elucidate alternative biologic dependencies and therapeutic vulnerabilities, while the ABL-class Ph-like ALL subtype appears preferentially sensitive to SRC/ABL- or PDGFRB-targeting inhibitors. Which patients may be responsive versus resistant to tyrosine kinase inhibitor (TKI)-based precision medicine approaches remains a critical knowledge gap. Using bulk and single-cell multiomics analyses, we profiled residual cells from CRLF2-rearranged or ABL1-rearranged Ph-like ALL patient-derived xenograft models treated in vivo with targeted inhibitors to identify TKI-resistant subpopulations and potential mechanisms of therapeutic escape. We detected a specific MYC dependency in Ph-like ALL cells and defined a new leukemia cell subpopulation with senescence-associated stem cell-like features regulated by AP-1 transcription factors. This dormant ALL subpopulation was effectively eradicated by dual pharmacologic inhibition of BCL-2 and JAK/STAT or SRC/ABL pathways, a clinically-relevant therapeutic strategy. Single cell-derived molecular signatures of this senescence and stem/progenitor-like subpopulation further predicted poor clinical outcomes associated with other high-risk genetic subtypes of childhood B-ALL and thus may have broader prognostic applicability beyond Ph-like ALL.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":21.0,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142944625","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 : 2025-01-06DOI: 10.1182/blood.2024026211
Shruti Gupta, Sarah Allison Kaunfer, Kevin L Chen, Julie-Alexia Dias, Anitha Vijayan, Arun Rajasekaran, Jason Prosek, Huong L Truong, Anthony Wood, Claude Bassil, Amanda DeMauro Renaghan, Chintan V Shah, Jingjing Zhang, Ilya Glezerman, Christopher Carlos, Katherine Kelly, Christopher J Passero, Jan Drappatz, Ala Abudayyeh, Daniel Shin, C John Sperati, Bradley J Yelvington, Swetha Rani Kanduri, Javier A Neyra, Daniel Edmonston, Anushree C Shirali, Anip Bansal, Abdallah Geara, Zain Mithani, Susan L Ziolkowski, Arash Rashidi, Jonathan Jakubowski, Ashwini Pujari, David A Bond, Emily K Dotson, Sarah A Wall, John T Patton, Jason N Barreto, Sandra M Herrmann, Mohammad Salman Sheikh, Rachid C Baz, Jae Hui Lee, Nicholas Lucchesi, Michael Kolman, Muhammad Ahsan Rasheed, Afsheen Afzal, Dasol Kang, Amrita Mahesh, Raymond Hsu, Anthony Nicolaysen, Kibrewessen Tefera, Claire Schretlen, Ryan M Miller, Juan Carlos Q Velez, Alexander H Flannery, Abinet M Aklilu, Shuchi Anand, Soniya Chandrasekhara, Vicki Donley, Ashka Patel, Jian Ni, Shobana Krishnamurthy, Rafia Ali, Osman A Yilmam, Sophia L Wells, Jessica L Ortega, Olivia L Green-Lingren, Rebecca Karp Leaf, Meghan E Sise, Lakshmi Nayak, Ann S LaCasce, Nelson Leung, David E Leaf
High-dose methotrexate (MTX) results in high rates of acute kidney injury (AKI), neutropenia, and hepatotoxicity. Glucarpidase is a recombinant enzyme that cleaves MTX, but clinical data supporting its use are scarce. We examined the association between glucarpidase administration and outcomes in adults with MTX-AKI from 28 cancer centers across the U.S using a sequential target trial emulation framework. The primary end point was kidney recovery at hospital discharge, defined as survival to discharge with serum creatinine <1.5-fold baseline and without dialysis-dependence. Key secondary end points were time-to-kidney recovery, neutropenia and transaminitis on day 7, and time-to-death. Using multivariable logistic and Cox regression models, we compared outcomes in patients who received glucarpidase within 4 days following MTX initiation with those in patients who did not. Among 708 patients with MTX-AKI, 209 (29.5%) received glucarpidase. Overall, 183 (25.8%) had a primary end point event. Glucarpidase receipt was associated with a 2.70-fold higher adjusted odds of kidney recovery (95% CI, 1.69-4.31) compared to no glucarpidase receipt. Patients treated with glucarpidase also had faster time-to-kidney recovery (adjusted hazard ratio [aHR], 1.88, 95% CI, 1.18-3.33) and lower risks of grade ≥2 neutropenia (adjusted odds ratio [aOR], 0.50, 95% CI, 0.28-0.91) and grade ≥2 transaminitis (aOR, 0.50, 95% CI, 0.28-0.91) on day 7. There was no difference in time-to-death (aHR, 0.76; 95 CI, 0.49-1.18). These data suggest glucarpidase may improve both renal and extrarenal outcomes in patients with MTX-AKI.
{"title":"Glucarpidase for Treatment of High-Dose Methotrexate Toxicity.","authors":"Shruti Gupta, Sarah Allison Kaunfer, Kevin L Chen, Julie-Alexia Dias, Anitha Vijayan, Arun Rajasekaran, Jason Prosek, Huong L Truong, Anthony Wood, Claude Bassil, Amanda DeMauro Renaghan, Chintan V Shah, Jingjing Zhang, Ilya Glezerman, Christopher Carlos, Katherine Kelly, Christopher J Passero, Jan Drappatz, Ala Abudayyeh, Daniel Shin, C John Sperati, Bradley J Yelvington, Swetha Rani Kanduri, Javier A Neyra, Daniel Edmonston, Anushree C Shirali, Anip Bansal, Abdallah Geara, Zain Mithani, Susan L Ziolkowski, Arash Rashidi, Jonathan Jakubowski, Ashwini Pujari, David A Bond, Emily K Dotson, Sarah A Wall, John T Patton, Jason N Barreto, Sandra M Herrmann, Mohammad Salman Sheikh, Rachid C Baz, Jae Hui Lee, Nicholas Lucchesi, Michael Kolman, Muhammad Ahsan Rasheed, Afsheen Afzal, Dasol Kang, Amrita Mahesh, Raymond Hsu, Anthony Nicolaysen, Kibrewessen Tefera, Claire Schretlen, Ryan M Miller, Juan Carlos Q Velez, Alexander H Flannery, Abinet M Aklilu, Shuchi Anand, Soniya Chandrasekhara, Vicki Donley, Ashka Patel, Jian Ni, Shobana Krishnamurthy, Rafia Ali, Osman A Yilmam, Sophia L Wells, Jessica L Ortega, Olivia L Green-Lingren, Rebecca Karp Leaf, Meghan E Sise, Lakshmi Nayak, Ann S LaCasce, Nelson Leung, David E Leaf","doi":"10.1182/blood.2024026211","DOIUrl":"10.1182/blood.2024026211","url":null,"abstract":"<p><p>High-dose methotrexate (MTX) results in high rates of acute kidney injury (AKI), neutropenia, and hepatotoxicity. Glucarpidase is a recombinant enzyme that cleaves MTX, but clinical data supporting its use are scarce. We examined the association between glucarpidase administration and outcomes in adults with MTX-AKI from 28 cancer centers across the U.S using a sequential target trial emulation framework. The primary end point was kidney recovery at hospital discharge, defined as survival to discharge with serum creatinine <1.5-fold baseline and without dialysis-dependence. Key secondary end points were time-to-kidney recovery, neutropenia and transaminitis on day 7, and time-to-death. Using multivariable logistic and Cox regression models, we compared outcomes in patients who received glucarpidase within 4 days following MTX initiation with those in patients who did not. Among 708 patients with MTX-AKI, 209 (29.5%) received glucarpidase. Overall, 183 (25.8%) had a primary end point event. Glucarpidase receipt was associated with a 2.70-fold higher adjusted odds of kidney recovery (95% CI, 1.69-4.31) compared to no glucarpidase receipt. Patients treated with glucarpidase also had faster time-to-kidney recovery (adjusted hazard ratio [aHR], 1.88, 95% CI, 1.18-3.33) and lower risks of grade ≥2 neutropenia (adjusted odds ratio [aOR], 0.50, 95% CI, 0.28-0.91) and grade ≥2 transaminitis (aOR, 0.50, 95% CI, 0.28-0.91) on day 7. There was no difference in time-to-death (aHR, 0.76; 95 CI, 0.49-1.18). These data suggest glucarpidase may improve both renal and extrarenal outcomes in patients with MTX-AKI.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":""},"PeriodicalIF":21.0,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142930697","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.2024026814
Susan Harding, Joseph Mikhael
{"title":"Down with dex!","authors":"Susan Harding, Joseph Mikhael","doi":"10.1182/blood.2024026814","DOIUrl":"https://doi.org/10.1182/blood.2024026814","url":null,"abstract":"","PeriodicalId":9102,"journal":{"name":"Blood","volume":"145 1","pages":"3-4"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913526","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.2023023156
Nicola Gökbuget, Björn Steffen
Abstract: Despite advancements in new treatments, management of older patients with acute lymphoblastic leukemia (ALL) remains an unmet medical need. With increasing age, patients with ALL have a significantly lower complete remission rate, higher early mortality and relapse rate, and poorer survival than younger patients. This is attributed to a higher prevalence of adverse prognostic factors among older individuals and reduced tolerance to chemotherapy. Progress has been made in tailoring moderately intensive chemotherapy protocols for Philadelphia chromosome (Ph)/BCR::ABL-negative ALL in older patients, and recent phase 2 studies have explored integrating immunotherapy into initial treatment with very promising results. However, establishing new standard regimens for this age group remains and improving general management strategy is a pending task.
{"title":"How I treat older patients with Ph/BCR-ABL-negative acute lymphoblastic leukemia.","authors":"Nicola Gökbuget, Björn Steffen","doi":"10.1182/blood.2023023156","DOIUrl":"10.1182/blood.2023023156","url":null,"abstract":"<p><strong>Abstract: </strong>Despite advancements in new treatments, management of older patients with acute lymphoblastic leukemia (ALL) remains an unmet medical need. With increasing age, patients with ALL have a significantly lower complete remission rate, higher early mortality and relapse rate, and poorer survival than younger patients. This is attributed to a higher prevalence of adverse prognostic factors among older individuals and reduced tolerance to chemotherapy. Progress has been made in tailoring moderately intensive chemotherapy protocols for Philadelphia chromosome (Ph)/BCR::ABL-negative ALL in older patients, and recent phase 2 studies have explored integrating immunotherapy into initial treatment with very promising results. However, establishing new standard regimens for this age group remains and improving general management strategy is a pending task.</p>","PeriodicalId":9102,"journal":{"name":"Blood","volume":" ","pages":"53-63"},"PeriodicalIF":21.0,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142406068","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}