1
|
Bhatt KA, Vaynrub AJ, Cham J, Iyer SG, Izar B. Diagnosis and management of concurrent metastatic melanoma and chronic myelomonocytic leukemia. Melanoma Res 2025; 35:192-196. [PMID: 39903257 DOI: 10.1097/cmr.0000000000001025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
While the association between chronic lymphocytic leukemia (CLL) and a higher incidence of melanoma is well documented, the diagnosis of concurrent high-risk chronic myelomonocytic leukemia (CMML) and metastatic melanoma (MM) has not previously been described. Moreover, the treatment of MM and CMML differ greatly in the mechanism of action of their corresponding antineoplastic therapies: treatment of MM frequently involves immune checkpoint inhibitors (ICI), while patients with CMML receive myelosuppressive agents. Simultaneous management of these malignancies can be nuanced due to the potential impact of one treatment's constituents on the activity of the other and the broad and nonoverlapping array of potential adverse effects of these agents. Here, we describe the clinical course of a patient who was diagnosed with concurrent MM and CMML and our approach to the challenging balance of delivering ICI concurrently with the hypomethylating agent azacitidine and the BCL-2 inhibitor venetoclax.
Collapse
Affiliation(s)
- Kishan A Bhatt
- Columbia University Vagelos College of Physicians and Surgeons
| | - Anna J Vaynrub
- Columbia University Vagelos College of Physicians and Surgeons
| | - Jason Cham
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center
| | - Sunil G Iyer
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center
| | - Benjamin Izar
- Columbia University Vagelos College of Physicians and Surgeons
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center
- Columbia Center for Translational Immunology
- Department of Systems Biology, Program for Mathematical Genomics, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
2
|
Zugasti I, Lopez-Guerra M, Castaño-Díez S, Esteban D, Avendaño A, Pomares H, Perez A, García-Ávila S, Conejo IP, de la Fuente Montes C, Martínez-Roca A, Merchán B, Jiménez-Vicente C, Guijarro F, Álamo JR, Cortes-Bullich A, Torrecillas V, Mont L, Carcelero E, Riu G, Zamora L, Bargay J, Triguero A, Suarez-Lledó M, Salas MQ, López-Cadenas F, Ramos F, Xicoy B, Valcárcel D, Arnan M, Martínez C, Rovira M, Fernández-Avilés F, Díez-Campelo M, Esteve J, Díaz-Beyá M. Hypomethylating agents plus venetoclax for high-risk MDS and CMML as bridge therapy to transplant: a GESMD study. Exp Hematol Oncol 2025; 14:61. [PMID: 40287746 PMCID: PMC12032758 DOI: 10.1186/s40164-025-00652-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND High-risk myelodysplastic syndromes (HR-MDS) and chronic myelomonocytic leukemia (CMML) remain therapeutic challenges with suboptimal outcomes. The only potentially curative treatment is allogeneic stem cell transplantation (allo-SCT). The most frequent pre-allo-SCT treatment is monotherapy with hypomethylating agents (HMA), but approximately 40% of patients cannot proceed to allo-SCT, mainly due to disease progression. Recent evidence suggests that combining HMA with venetoclax (HMA/VEN) could increase HMA efficacy in HR-MDS but it remains unclear if this combination could bridge more patients to allo-SCT. METHODS We retrospectively evaluated HMA/VEN as a bridge to allo-SCT in 30 patients with HR-MDS or CMML eligible for transplant. Eighteen patients were treatment-naïve and 12 were refractory or relapsed (R/R). RESULTS As defined by the IWG 2023 criteria, the overall response rate (ORR) was 90% and the composite complete response rate was 77%. For the R/R patients, ORR was 83%. The allo-SCT rate was 83%, and the allo-SCT rate of those patients treated exclusively with HMA/VEN without further bridge therapies was 76%. One- and two-year post-allo-SCT survival was 75% and two-year cumulative incidence of relapse was 30.5%. Follow-up of measurable residual disease identified some molecular relapses that were controlled with preemptive treatment. CONCLUSIONS Our findings indicate that HMA/VEN combination therapy shows promise as a bridging strategy to allo-SCT in HR-MDS and CMML.
Collapse
Affiliation(s)
- Ines Zugasti
- Hospital Clínic Barcelona, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Monica Lopez-Guerra
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sandra Castaño-Díez
- Hospital Clínic Barcelona, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Daniel Esteban
- Hospital Clínic Barcelona, Barcelona, Spain
- Institut Català d'Oncologia (ICO), Hospital Germans Trias I Pujol, Badalona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Alejandro Avendaño
- Hospital Universitario de Salamanca, Salamanca, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Helena Pomares
- Institut Català d'Oncologia, Hospital Duran I Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Ana Perez
- Hospital Universitario Vall d´Hebrón, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Sara García-Ávila
- Hospital del Mar, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Irene Padilla Conejo
- Hospital Universitario de León, León, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Cristina de la Fuente Montes
- Institut Català d'Oncologia (ICO), Hospital Germans Trias I Pujol, Badalona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Alexandra Martínez-Roca
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Beatriz Merchán
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Carlos Jiménez-Vicente
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Francesca Guijarro
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Jose Ramón Álamo
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Albert Cortes-Bullich
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | | | - Lucia Mont
- Universitat de Barcelona, Barcelona, Spain
| | | | - Gisela Riu
- Hospital Clínic Barcelona, Barcelona, Spain
| | - Lurdes Zamora
- Institut Català d'Oncologia (ICO), Hospital Germans Trias I Pujol, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ana Triguero
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Maria Suarez-Lledó
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Maria Queralt Salas
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Felix López-Cadenas
- Hospital del Mar, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Fernando Ramos
- Hospital Universitario de León, León, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Blanca Xicoy
- Institut Català d'Oncologia (ICO), Hospital Germans Trias I Pujol, Badalona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David Valcárcel
- Hospital Universitario Vall d´Hebrón, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Montserrat Arnan
- Institut Català d'Oncologia, Hospital Duran I Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Carmen Martínez
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Montserrat Rovira
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Francesc Fernández-Avilés
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Maria Díez-Campelo
- Hospital Universitario de Salamanca, Salamanca, Spain
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain
| | - Jordi Esteve
- Hospital Clínic Barcelona, Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Marina Díaz-Beyá
- Hospital Clínic Barcelona, Barcelona, Spain.
- Grupo Español de Síndromes Mielodisplásicos (GESMD), Madrid, Spain.
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain.
- Josep Carreras Leukemia Research Institute, Barcelona, Spain.
| |
Collapse
|
3
|
Tremblay D, Hasserjian RP, Rampal RK. Myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes: a practical guide to diagnosis and management. Leukemia 2025:10.1038/s41375-025-02620-8. [PMID: 40253543 DOI: 10.1038/s41375-025-02620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) overlap syndromes are a rare group of biologically and clinically connected hematologic malignancies that includes chronic myelomonocytic leukemia (CMML), the most common subtype, as well as atypical chronic myeloid leukemia, MDS/MPN with SF3B1 and thrombocytosis, and MDS/MPN, not otherwise specified. Given their rarity and overlapping clinical features, accurate diagnosis and risk stratification presents a significant challenge. Therapeutic approaches are largely borrowed from either MDS or MPN and the only curative option for appropriate patients is allogeneic stem cell transplantation. Recent advances have started to uncover the pathobiologic basis for these diseases, leading to novel clinical trials for MDS/MPN overlap syndromes, in particular CMML. This review is a practical guide for the diagnosis and management of MDS/MPN overlap syndromes and presents novel therapeutics being specifically designed for these diseases to improve their historically poor outcomes.
Collapse
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | | | | |
Collapse
|
4
|
Darmusey L, Bagley AJ, Nguyen TT, Carlson HL, Blaylock H, Shrestha SB, Pang A, Tauchmann S, Taylor SC, Foley AC, Niño KE, Pietras EM, Braun TP, Maxson JE. Dual ASXL1 and CSF3R mutations drive myeloid-biased stem cell expansion and enhance neutrophil differentiation. Blood Adv 2025; 9:1593-1607. [PMID: 39777477 PMCID: PMC11986226 DOI: 10.1182/bloodadvances.2024014362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
ABSTRACT Mutations in the epigenetic regulator Additional Sex Combs-Like 1 (ASXL1) are frequently observed in chronic neutrophilic leukemia (CNL). CNL is a myeloproliferative neoplasm (MPN) driven by activating mutations in the Colony Stimulating Factor 3 Receptor (CSF3R), which cause excessive neutrophil production. Despite the high rates of co-occurrence, the interplay between ASXL1 and CSF3R mutations in hematopoiesis and leukemia remains poorly understood. Here, we present a new mouse model with both Asxl1Y588X and Csf3rT621I mutations, which recapitulates features of human MPNs. Csf3r-mutant mice exhibit an age-associated depletion of hematopoietic stem cells, which is tempered by adding Asxl1Y588X. This combination of mutations causes an expansion of myeloid-biased long-term hematopoietic stem cells. As the mice age, they develop neutrophilia, but leukemia is rare, suggesting additional mutations may be required for transformation. Using models of myeloid differentiation, we find that Asxl1 truncation enhances CSF3RT618I-driven neutrophil differentiation, activating inflammatory pathways associated with mature myeloid cell production. Moreover, cells with both mutations have increased H3K4me1 at neutrophil-associated enhancers. Mutant ASXL1 is known to decrease the genome-wide abundance of the repressive histone mark H2AK119ub. Although we see the expected decrease in H2AK119ub in Asxl1-mutant cells, this effect is reversed when CSF3R is also mutated, suggesting a complex interplay between these mutations in regulating chromatin dynamics during hematopoiesis. Our findings highlight context-dependent effects of ASXL1 mutation in myeloid disorders and provide insights into the mechanisms underlying neutrophil differentiation in ASXL1 and CSF3R dual-mutant MPN.
Collapse
Affiliation(s)
- Lucie Darmusey
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Anna J. Bagley
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Thai T. Nguyen
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Hanqian L. Carlson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Hunter Blaylock
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Shawn B. Shrestha
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Amara Pang
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Samantha Tauchmann
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Sarah C. Taylor
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Amy C. Foley
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Katia E. Niño
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eric M. Pietras
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Theodore P. Braun
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Julia E. Maxson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
5
|
Pratz KW. Advancing the outcomes of AML out of antecedent MPN by targeting mutated IDH1. Br J Haematol 2025; 206:1250-1252. [PMID: 39710967 PMCID: PMC11985367 DOI: 10.1111/bjh.19959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Outcomes of myeloproliferative neoplasms (MPN)-associated acute leukaemias are dismal with conventional therapy. Approximately 20% of MPN-associated acute leukaemias have mutations in isocitrate dehydrogenase (IDH). Olutasidenib, and inhibitor of IDH1, demonstrates important clinical benefits in MPN-associated leukaemia with IDH1 mutation. Commentary on: Botton et al. Olutasidenib demonstrates significant clinical activity in mutated IDH1 acute myeloid leukemia arising from a prior myeloproliferative neoplasm. Br J Haematol 2025; 206:1121-1128.
Collapse
Affiliation(s)
- Keith W. Pratz
- Hospital of University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
6
|
Kazmi F, Shrestha N, Liu TFD, Foord T, Heesen P, Booth S, Dodwell D, Lord S, Yeoh KW, Blagden SP. Next-generation sequencing for guiding matched targeted therapies in people with relapsed or metastatic cancer. Cochrane Database Syst Rev 2025; 3:CD014872. [PMID: 40122129 PMCID: PMC11930395 DOI: 10.1002/14651858.cd014872.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
BACKGROUND Matched targeted therapies (MTT) given alone or in combination with systemic anti-cancer therapies have delivered proven survival benefit for many people with newly diagnosed cancer. However, there is little evidence of their effectiveness in the recurrent or late-stage setting. With this uncertainty, alongside the perception that late-stage cancers are too genetically heterogenous or too mutationally diverse to benefit from matched targeted therapies, next-generation sequencing (NGS) of tumours in people with refractory cancer remains a low priority. As a result, next-generation sequencing testing of recurrent or late-stage disease is discouraged. We lack evidence to support the utility of next generation sequencing in guiding matched targeted therapies in this setting. OBJECTIVES To evaluate the benefits and harms of matched targeted therapies in people with advanced cancers in randomised controlled trials. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, ClinicalTrials.gov, and the World Health Organisation International Clinical Trials Registry Platform (WHO-ICTRP) search portal up to 30th October 2024. We also screened reference lists of included studies and also the publications that cited these studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) that had enroled participants with advanced/refractory solid or haematological cancers who had progressed through at least one line of standard anti-cancer systemic therapy. To be eligible, all participants should have received matched targeted therapy based on next-generation sequencing carried out on their tumour (tumour tissue, blood or bone marrow). DATA COLLECTION AND ANALYSIS We systematically searched medical databases (e.g. MEDLINE, Embase) and trial registers for randomised controlled trials (RCTs). Outcomes of interest were progression-free survival (PFS), overall survival (OS), overall response rates (ORR), serious (grade 3 or 4) adverse events (AEs) and quality of life (QOL). We used a random-effects model to pool outcomes across studies and compared predefined subgroups using interaction tests. Grading of Recommendations Assessment, Development and Evaluation (GRADE) assessment of certainty was used to evaluate the quality of evidence. MAIN RESULTS We identified a total of 37 studies, out of which 35 studies (including 9819 participants) were included in the meta-analysis. All included studies compared a matched targeted therapy intervention to standard-of-care treatment, non-matched targeted therapies or no treatment (best supportive care): Matched targeted therapy versus standard-of-care treatment Matched targeted therapy (MTT) compared with standard systematic therapy probably reduces the risk of disease progression by 34% (hazard ratio (HR) = 0.66, 95% confidence interval (CI) 0.59 to 0.74; 14 studies, 3848 participants; moderate-certainty evidence). However, MTT might have little to no difference in risk of death (HR = 0.85, 95% CI 0.75 to 0.97; 14 studies, 3848 participants; low-certainty evidence) and may increase overall response rates (low-certainty evidence). There was no clear evidence of a difference in severe (grade 3/4) adverse events between matched targeted therapy and standard-of-care treatment (low-certainty evidence). There was limited evidence of a difference in quality of life between groups (very low-certainty of evidence). Matched targeted therapy in combination with standard-of-care treatment versus standard-of-care treatment alone Matched targeted therapy in combination with standard-of-care treatment compared with standard-of-care treatment alone probably reduces the risk of disease progression by 39% (HR = 0.61, 95% CI 0.53-0.70, 14 studies, 2,637 participants; moderate-certainty evidence) and risk of death by 21% (HR = 0.79, 95% CI 0.70 to 0.89; 11 studies, 2575 participants, moderate-certainty evidence). The combination of MTT and standard-of-care treatment may also increase overall response rates (low-certainty evidence). There was limited evidence of a difference in the incidence of severe adverse events (very low-certainty evidence) and quality of life between the groups (very low-certainty of evidence). Matched targeted therapy versus non-matched targeted therapy Matched targeted therapy compared with non-matched targeted therapy probably reduces the risk of disease progression by 24% (HR = 0.76, 95% CI 0.64 to 0.89; 3 studies, 1568 participants; moderate-certainty evidence) and may reduce the risk of death by 25% (HR = 0.75, 95% CI 0.65 to 0.86, 1307 participants; low-certainty evidence). There was little to no effect on overall response rates between MTT and non-MTT. There was no clear evidence of a difference in overall response rates (low-certainty evidence) and severe adverse events between MTT and non-MTT (low-certainty evidence). None of the studies comparing MTT and non-MTT reported quality of life. Matched targeted therapy versus best supportive care Matched targeted therapy compared with the best supportive care (BSC) i.e. no active treatment probably reduces the risk of disease progression by 63% (HR 0.37, 95% CI 0.28 to 0.50; 4 studies, 858 participants; moderate-certainty evidence). There was no clear evidence of a difference in overall survival between groups (HR = 0.88, 95% CI 0.73 to 1.06, 3 studies, 783 participants; low-certainty evidence). There was no clear evidence of a difference in overall response rates (very low-certainty of evidence) and incidence of severe adverse events (very low-certainty of evidence) between the groups. Quality of life was reported in a single study but did not provide composite scores. Risk of bias The overall risk of bias was judged low for eight studies, unclear for two studies, and the remaining 27 studies were high risk. AUTHORS' CONCLUSIONS Matched targeted therapies guided by next-generation sequencing in people with advanced cancer prolongs the time before cancer progresses compared to standard therapies. However, there is limited evidence to suggest that it prolongs overall survival, improves the quality of life or increases adverse events. Importantly, this review supports equitable access to next-generation sequencing technology for all people with advanced cancer and offers them the opportunity to access genotype-matched targeted therapies.
Collapse
Affiliation(s)
- Farasat Kazmi
- Department of Oncology, University of Oxford, Oxford, UK
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Nipun Shrestha
- Health Evidence Synthesis, Recommendations and Impact (HESRI), School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Tik Fung Dave Liu
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | | | | | - Stephen Booth
- Department of Haematology, Royal Berkshire Hospital, Reading, UK
| | - David Dodwell
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Oxford, UK
| | - Kheng-Wei Yeoh
- Radiation Oncology, National Cancer Centre, Singapore, Singapore
| | | |
Collapse
|
7
|
Sammut R, Fenwarth L, Pelissier A, Marceau A, Duployez N, Benachour S, Dadone B, Cluzeau T, Loschi M. Clonal Evolution of Myeloid Malignancies Treated With Microtransplantation: A Single-Centre Experience. J Cell Mol Med 2025; 29:e70520. [PMID: 40126789 PMCID: PMC11932058 DOI: 10.1111/jcmm.70520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 01/31/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Microtransplantation is a cellular therapy used in acute myeloid leukaemia and myelodysplastic syndromes as a maintenance therapy in patients ineligible for a regular allogeneic stem cell transplantation. We performed a monocentric retrospective study of acute myeloid leukaemia, myelodysplastic syndromes, and chronic myelomonocytic leukaemia patients who underwent microtransplantations at Nice University Hospital. We analysed the evolution of the disease mutational status after microtransplantation. We report 18 patients who underwent microtransplantation courses, with a total of 47 microtransplantations performed between February 2020 and June 2022. We observed long-term remissions even in high-risk patients. Founder mutations persisted throughout the follow-up, whereas it was more variable for other nonfounder mutations, with most of the nonfounder mutations variant allele frequency decreasing over time. Safety data were reassuring; no graft versus host disease was recorded, and cytokine release syndromes were manageable. Relapses or progressions were associated with the emergence or increase of a TP53 mutated clone. Microtransplantation is a promising therapy for patients ineligible for regular allogeneic stem transplantation. Further larger and randomised studies are required to establish its place as a maintenance therapy in myeloid malignancies.
Collapse
Affiliation(s)
- R. Sammut
- Département d'HématologieCentre Hospitalier Universitaire de NiceNiceFrance
| | - L. Fenwarth
- Laboratoire d'HématologieCentre Hospitalier Universitaire de LilleFrance
| | - A. Pelissier
- Department of Biosystems Science and EngineeringETH ZurichBaselSwitzerland
| | - A. Marceau
- Laboratoire d'HématologieCentre Hospitalier Universitaire de LilleFrance
| | - N. Duployez
- Laboratoire d'HématologieCentre Hospitalier Universitaire de LilleFrance
| | - S. Benachour
- Département d'HématologieCentre Hospitalier Universitaire de NiceNiceFrance
| | - B. Dadone
- Laboratoire Central d'anatomie et de Cytologie PathologiqueCentre Hospitalier Universitaire de NiceNiceFrance
| | - T. Cluzeau
- Département d'HématologieCentre Hospitalier Universitaire de NiceNiceFrance
- INSERM U1065, Centre de Médecine Moléculaire MéditerranéenUniversité Nice Cote d'AzurNiceFrance
- Université Cote d'AzurNiceFrance
| | - M. Loschi
- Département d'HématologieCentre Hospitalier Universitaire de NiceNiceFrance
- INSERM U1065, Centre de Médecine Moléculaire MéditerranéenUniversité Nice Cote d'AzurNiceFrance
- Université Cote d'AzurNiceFrance
| |
Collapse
|
8
|
Castaño-Díez S, López-Guerra M, Zugasti I, Calvo X, Schulz FI, Avendaño A, Mora E, Falantes J, Azaceta G, Ibáñez M, Chen T, Notario C, Amer N, Palomo L, Pomares H, Vila J, Bernal del Castillo T, Jiménez-Vicente C, Esteban D, Guijarro F, Álamo J, Cortés-Bullich A, Torrecillas-Mayayo V, Triguero A, Mont-de Torres L, Carcelero E, Cardús A, Germing U, Betz B, Rozman M, Arenillas L, Zamora L, Díez-Campelo M, Xicoy B, Esteve J, Díaz-Beyá M. AML typical mutations (CEBPA, FLT3, NPM1) identify a high-risk chronic myelomonocytic leukemia independent of CPSS molecular. Blood Adv 2025; 9:39-53. [PMID: 39388660 PMCID: PMC11732582 DOI: 10.1182/bloodadvances.2024013648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/30/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024] Open
Abstract
ABSTRACT Mutations commonly associated with acute myeloid leukemia (AML), such as CEBPA, FLT3, IDH1/2, and NPM1, are rarely found in chronic myelomonocytic leukemia (CMML), and their prognostic significance in CMML has not been clearly identified. In 127 patients with CMML, we have retrospectively analyzed next-generation sequencing and polymerase chain reaction data from bone marrow samples collected at the time of CMML diagnosis. Seven patients harbored CEBPA mutations, 8 FLT3 mutations, 12 IDH1 mutations, 26 IDH2 mutations, and 11 NPM1 mutations. Patients with CMML harboring CEBPA, FLT3, and/or NPM1 mutations (mutCFN) more frequently had the myeloproliferative subtype, a high prevalence of severe cytopenia, and elevated blast counts. Regardless of their CMML Prognostic Scoring System molecular classification, mutCFN patients with CMML had a poor prognosis, and the multivariate analysis identified mutCFN as an independent marker of overall survival. The genetic profile of these mutCFN patients with CMML closely resembled that of patients with AML, with higher-risk clinical characteristics. Our findings lead us to suggest including the assessment of these mutations in CMML prognostic models and treating these patients with AML-type therapies, including intensive chemotherapy and allogeneic stem cell transplantation, whenever feasible. Furthermore, certain targeted therapies approved for use in AML should be considered.
Collapse
MESH Headings
- Humans
- Nucleophosmin
- Mutation
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/diagnosis
- Leukemia, Myelomonocytic, Chronic/mortality
- fms-Like Tyrosine Kinase 3/genetics
- Male
- Female
- Middle Aged
- Aged
- Nuclear Proteins/genetics
- CCAAT-Enhancer-Binding Proteins/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Prognosis
- Adult
- Aged, 80 and over
- Retrospective Studies
Collapse
Affiliation(s)
- Sandra Castaño-Díez
- Department of Hematopathology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
| | - Mònica López-Guerra
- Department of Hematopathology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Inés Zugasti
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Spain
| | - Xavier Calvo
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital del Mar, Barcelona, Spain
| | - Felicitas Isabel Schulz
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Alejandro Avendaño
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca, Spain
| | - Elvira Mora
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Universitario y Politécnico la Fe, Valencia, Spain
| | - José Falantes
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Gemma Azaceta
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Clínico Universitario Lozano Blesa de Zaragoza, Spain
| | - Mariam Ibáñez
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital General Universitario de Valencia, grupo de Investigación de Hematología y Hemoterapia de la Fundación de Investigación del Hospital General Universitario de Valencia, Spain
| | - Tzu Chen
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital General Universitario Morales Meseguer, Murcia, Spain
| | - Cristina Notario
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Universitario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Neus Amer
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Universitario Son Llàtzer, Palma de Mallorca, Spain
| | - Laura Palomo
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Experimental Hematology Group, Vall d'Hebron Institute of Oncology, University Hospital Vall d'Hebron, Barcelona, Spain. Myelodysplastic Syndromes Research Group, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Helena Pomares
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Institut Català d'Oncologia - Hospital Duran i Reynals, L'Hospitalet de LLobregat, LLobregat, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, Spain
| | - Jordi Vila
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Institut Català d'Oncologia - Hospital Dr. Josep Trueta, Girona, Spain
| | - Teresa Bernal del Castillo
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Carlos Jiménez-Vicente
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Spain
| | - Daniel Esteban
- Department of Hematology, Institut Català d’Oncologia-Hospital Universitari Germans Trias i Pujol, Institut de Recerca contra la Leucèmia Josep Carreras, Badalona, Barcelona, Spain
| | - Francesca Guijarro
- Department of Hematopathology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - José Álamo
- Department of Hematopathology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
| | - Albert Cortés-Bullich
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Spain
| | | | - Ana Triguero
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Spain
| | | | - Ester Carcelero
- Department of Pharmacy, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Aina Cardús
- Department of Hematopathology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Beate Betz
- Institute for Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Maria Rozman
- Department of Hematopathology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
| | - Leonor Arenillas
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital del Mar, Barcelona, Spain
| | - Lurdes Zamora
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Institut Català d’Oncologia-Hospital Universitari Germans Trias i Pujol, Institut de Recerca contra la Leucèmia Josep Carreras, Badalona, Barcelona, Spain
| | - María Díez-Campelo
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca, Spain
| | - Blanca Xicoy
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Institut Català d’Oncologia-Hospital Universitari Germans Trias i Pujol, Institut de Recerca contra la Leucèmia Josep Carreras, Badalona, Barcelona, Spain
| | - Jordi Esteve
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Spain
| | - Marina Díaz-Beyá
- Grupo Español de Síndromes Mielodisplásicos, Madrid, Spain
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Spain
| |
Collapse
|
9
|
Tremblay D, Csizmar C, DiNardo CD, Ball S, Rippel N, Hammond D, Kadia TM, Ravandi F, Chien K, Van Hyfte G, Mazumdar M, Saliba A, Mangaonkar A, Lasho T, Al-Kali A, Kremyanskaya M, Feld J, Silverman LR, Komrokji R, Mascarenhas J, Padron E, Garcia-Manero G, Sallman DA, Patnaik MM, Montalban-Bravo G. Venetoclax in combination with hypomethylating agents in chronic myelomonocytic leukemia: a propensity score matched multicenter cohort study. Leukemia 2025; 39:257-260. [PMID: 39533069 DOI: 10.1038/s41375-024-02466-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | | | | | - Somedeb Ball
- Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Noa Rippel
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Grace Van Hyfte
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhu Mazumdar
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Marina Kremyanskaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Feld
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lewis R Silverman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
10
|
Wu Q, Yu C, Yu F, Guo Y, Sheng Y, Li L, Li Y, Zhang Y, Hu C, Wang J, He TC, Huang Y, Ni H, Huo Z, Wu W, Wang GG, Lyu J, Qian Z. Evi1 governs Kdm6b-mediated histone demethylation to regulate the Laptm4b-driven mTOR pathway in hematopoietic progenitor cells. J Clin Invest 2024; 134:e173403. [PMID: 39680456 PMCID: PMC11645144 DOI: 10.1172/jci173403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/24/2024] [Indexed: 12/18/2024] Open
Abstract
Ecotropic viral integration site 1 (EVI1/MECOM) is frequently upregulated in myeloid malignancies. Here, we present an Evi1-transgenic mouse model with inducible expression in hematopoietic stem/progenitor cells (HSPCs). Upon induction of Evi1 expression, mice displayed anemia, thrombocytopenia, lymphopenia, and erythroid and megakaryocyte dysplasia with a significant expansion of committed myeloid progenitor cells, resembling human myelodysplastic syndrome/myeloproliferative neoplasm-like (MDS/MPN-like) disease. Evi1 overexpression prompted HSPCs to exit quiescence and accelerated their proliferation, leading to expansion of committed myeloid progenitors while inhibiting lymphopoiesis. Analysis of global gene expression and Evi1 binding site profiling in HSPCs revealed that Evi1 directly upregulated lysine demethylase 6b (Kdm6b). Subsequently, Kdm6b-mediated H3K27me3 demethylation resulted in activation of various genes, including Laptm4b. Interestingly, KDM6B and LAPTM4B are positively correlated with EVI1 expression in patients with MDS. The EVI1/KDM6B/H3K27me3/LAPTM4B signaling pathway was also identified in EVI1hi human leukemia cell lines. We found that hyperactivation of the LAPTM4B-driven mTOR pathway was crucial for the growth of EVI1hi leukemia cells. Knockdown of Laptm4b partially rescued Evi1-induced abnormal hematopoiesis in vivo. Thus, our study establishes a mouse model to investigate EVI1hi myeloid malignancies, demonstrating the significance of the EVI1-mediated KDM6B/H3K27me3/LAPTM4B signaling axis in their maintenance.
Collapse
Affiliation(s)
- Qiong Wu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Chunjie Yu
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Fang Yu
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Yiran Guo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Yue Sheng
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Liping Li
- Department of Pathology at Geisinger Medical Center, Danville, Pennsylvania, USA
| | - Yafang Li
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Yutao Zhang
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Chao Hu
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jue Wang
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Tong-chuan He
- Department of Orthopaedic Surgery and Rehabilitation Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yong Huang
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Hongyu Ni
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Wenshu Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gang Greg Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Jianxin Lyu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Laboratory Medicine, Zhejiang Provincial People’s Hospital, Affiliate People’s Hospital of Hangzhou Medical College, and
- Laboratory Medicine of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhijian Qian
- Department of Medicine and Department of Biochemistry and Molecular Biology, UF Health Cancer Center, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
11
|
Arslan Davulcu E, Akı SH. A Case of Chronic Myelomonocytic Leukemia With Recurrent Skin Involvement. Cureus 2024; 16:e75193. [PMID: 39759649 PMCID: PMC11700349 DOI: 10.7759/cureus.75193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Chronic myelomonocytic leukemia is a clonal hematopoietic stem cell disorder with both myelodysplastic and myeloproliferative features, leading to a variable clinical presentation. Some types of skin involvement, such as leukemia cutis and blastic plasmacytoid dendritic cell neoplasia, are associated with poor prognosis. This case study describes a 71-year-old male with high-risk CMML, developing pink-purple skin nodules, which regressed with azacitidine and hydroxyurea treatment. Despite recurrence, disease control was achieved without transformation to acute leukemia. This case highlights the need for vigilant monitoring and adaptable treatment strategies in managing CMML with skin involvement.
Collapse
Affiliation(s)
- Eren Arslan Davulcu
- Hematology Clinic, University of Health Sciences Bakırkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, TUR
| | - Suat Hilal Akı
- Pathology Department, Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Istanbul, TUR
| |
Collapse
|
12
|
Hunter AM, Patnaik MM, Itzykson R, Mesa R, Karanes C, Li Y, de Claro RA, Norsworthy KJ, Theoret M, Pulte E, Padron E. Perspectives on drug development in chronic myelomonocytic leukemia: changing the paradigm. Blood 2024; 144:1987-1992. [PMID: 39509115 DOI: 10.1182/blood.2024025648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/27/2024] [Indexed: 11/15/2024] Open
Abstract
ABSTRACT Drug development for chronic myelomonocytic leukemia (CMML) has failed to parallel the recent success observed in related myeloid neoplasms. To address these shortcomings, the US Food and Drug Administration (FDA) held a "Mini-symposium on CMML: Current State of the Art and Trial Design" in September 2023. This symposium brought together a panel of key FDA regulators and academic experts in CMML drug development to discuss challenges and provide perspectives on future drug development for this disease. The panel explored unique challenges that underlie the lack of therapeutic advances in CMML to date and discussed relevant topics such as clinical trial design, study end points, and key regulatory considerations. This article summarizes the key points of discussion from this symposium to facilitate advancements in the field.
Collapse
Affiliation(s)
- Anthony M Hunter
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Raphael Itzykson
- Université Paris Cité, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, Centre National de la Recherche Scientifique, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ruben Mesa
- Hematology and Medical Oncology, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Chatchada Karanes
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Yanxia Li
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - R Angelo de Claro
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Kelly J Norsworthy
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Marc Theoret
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, MD
| | - Elizabeth Pulte
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Eric Padron
- Hematologic Malignancies. H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
13
|
Montalban-Bravo G, Jabbour E, Chien K, Hammond D, Short N, Ravandi F, Konopleva M, Borthakur G, Daver N, Kanagal-Shammana R, Loghavi S, Qiao W, Huang X, Schneider H, Meyer M, Kantarjian H, Garcia-Manero G. Phase 1 study of azacitidine in combination with quizartinib in patients with FLT3 or CBL mutated MDS and MDS/MPN. Leuk Res 2024; 142:107518. [PMID: 38744144 DOI: 10.1016/j.leukres.2024.107518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
We conducted a phase 1 study evaluating 3 dose levels of quizartinib (30 mg, 40 mg or 60 mg) in combination with azacitidine for HMA-naïve or relapsed/refractory MDS or MDS/MPN with FLT3 or CBL mutations. Overall, 12 patients (HMA naïve: n=9, HMA failure: n=3) were enrolled; 7 (58 %) patients had FLT3 mutations and 5 (42 %) had CBL mutations. The maximum tolerated dose was not reached. Most common grade 3-4 treatment-emergent adverse events were thrombocytopenia (n=5, 42 %), anemia (n=4, 33 %), lung infection (n=2, 17 %), skin infection (n=2, 17 %), hyponatremia (n=2, 17 %) and sepsis (n=2, 17 %). The overall response rate was 83 % with median relapse-free and overall survivals of 15.1 months (95 % CI 0.0-38.4 months) and 17.5 months (95 % CI NC-NC), respectively. FLT3 mutation clearance was observed in 57 % (n=4) patients. These data suggest quizartinib is safe and shows encouraging activity in FLT3-mutated MDS and MDS/MPN. This study is registered at Clinicaltrials.gov as NCT04493138.
Collapse
Affiliation(s)
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Kelly Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Nicholas Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | | | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, USA
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, USA
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, USA
| | - Heather Schneider
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Meghan Meyer
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, USA
| | | |
Collapse
|
14
|
Patnaik MM, Tefferi A. Chronic myelomonocytic leukemia: 2024 update on diagnosis, risk stratification and management. Am J Hematol 2024; 99:1142-1165. [PMID: 38450850 PMCID: PMC11096042 DOI: 10.1002/ajh.27271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
DISEASE OVERVIEW Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms, characterized by prominent monocytosis and an inherent risk for leukemic transformation (~15%-20% over 3-5 years). DIAGNOSIS Newly revised diagnostic criteria include sustained (>3 months) peripheral blood (PB) monocytosis (≥0.5 × 109/L; monocytes ≥10% of leukocyte count), consistent bone marrow (BM) morphology, <20% BM or PB blasts (including promonocytes), and cytogenetic or molecular evidence of clonality. Cytogenetic abnormalities occur in ~30% of patients, while >95% harbor somatic mutations: TET2 (~60%), SRSF2 (~50%), ASXL1 (~40%), RAS pathway (~30%), and others. The presence of ASXL1 and DNMT3A mutations and absence of TET2 mutations negatively impact overall survival (ASXL1WT/TET2MT genotype being favorable). RISK STRATIFICATION Several risk models serve similar purposes in identifying high-risk patients that are considered for allogeneic stem cell transplant (ASCT) earlier than later. Risk factors in the Mayo Molecular Model (MMM) include presence of truncating ASXL1 mutations, absolute monocyte count >10 × 109/L, hemoglobin <10 g/dL, platelet count <100 × 109/L, and the presence of circulating immature myeloid cells; the resulting 4-tiered risk categorization includes high (≥3 risk factors), intermediate-2 (2 risk factors), intermediate-1 (1 risk factor), and low (no risk factors); the corresponding median survivals were 16, 31, 59, and 97 months. CMML is also classified as being "myeloproliferative (MP-CMML)" or "myelodysplastic (MD-CMML)," based on the presence or absence of leukocyte count of ≥13 × 109/L. TREATMENT ASCT is the only treatment modality that secures cure or long-term survival and is appropriate for MMM high/intermediate-2 risk disease. Drug therapy is currently not disease-modifying and includes hydroxyurea and hypomethylating agents; a recent phase-3 study (DACOTA) comparing hydroxyurea and decitabine, in high-risk MP-CMML, showed similar overall survival at 23.1 versus 18.4 months, respectively, despite response rates being higher for decitabine (56% vs. 31%). UNIQUE DISEASE ASSOCIATIONS These include systemic inflammatory autoimmune diseases, leukemia cutis and lysozyme-induced nephropathy; the latter requires close monitoring of renal function during leukocytosis and is a potential indication for cytoreductive therapy.
Collapse
Affiliation(s)
- Mrinal M Patnaik
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Sadigh S, Kim AS. Molecular Pathology of Myeloid Neoplasms: Molecular Pattern Recognition. Clin Lab Med 2024; 44:339-353. [PMID: 38821648 DOI: 10.1016/j.cll.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Despite the apparent complexity of the molecular genetic underpinnings of myeloid neoplasms, most myeloid mutational profiles can be understood within a simple framework. Somatic mutations accumulate in hematopoietic stem cells with aging and toxic insults, termed clonal hematopoiesis. These "old stem cells" mutations, predominantly in the epigenetic and RNA spliceosome pathways, act as "founding" driver mutations leading to a clonal myeloid neoplasm when sufficient in number and clone size. Subsequent mutations can create the genetic flavor of the myeloid neoplasm ("backseat" drivers) due to their enrichment in certain entities or act as progression events ("aggressive" drivers) during clonal evolution.
Collapse
Affiliation(s)
- Sam Sadigh
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Annette S Kim
- Division of Diagnostic Genetics and Genomics, Department of Pathology, University of Michigan/Michigan Medicine, 2800 Plymouth Road, NCRC 36-1221-79, Ann Arbor, MI 48109, USA.
| |
Collapse
|
16
|
Boccia R, Xiao H, von Wilamowitz-Moellendorff C, Raorane R, Deshpande S, Klijn SL, Yucel A. A Systematic Literature Review of Predictors of Erythropoiesis-Stimulating Agent Failure in Lower-Risk Myelodysplastic Syndromes. J Clin Med 2024; 13:2702. [PMID: 38731231 PMCID: PMC11084325 DOI: 10.3390/jcm13092702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/04/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Erythropoiesis-stimulating agents (ESAs) are the first-line treatment option for anemia in patients with lower-risk myelodysplastic syndromes (LR-MDS). A systematic literature review was conducted to identify evidence of the association between prognostic factors and ESA response/failure in LR-MDS. MEDLINE, Embase, and relevant conferences were searched systematically for studies assessing the association between prognostic factors and ESA response/failure in adult patients. Of 1566 citations identified, 38 were included. Patient risk status in studies published from 2000 onwards was commonly assessed using the International Prognostic Scoring System (IPSS) or revised IPSS. ESA response was generally assessed using the International Working Group MDS criteria. Among the included studies, statistically significant relationships were found, in both univariate and multivariate analyses, between ESA response and the following prognostic factors: higher hemoglobin levels, lower serum erythropoietin levels, and transfusion independence. Furthermore, other prognostic factors such as age, bone marrow blasts, serum ferritin level, IPSS risk status, and karyotype status did not demonstrate statistically significant relationships with ESA response. This systematic literature review has confirmed prognostic factors of ESA response/failure. Guidance to correctly identify patients with these characteristics could be helpful for clinicians to provide optimal treatment.
Collapse
Affiliation(s)
- Ralph Boccia
- The Center for Cancer and Blood Disorders, 6410 Rockledge Drive, Suite 660, Bethesda, MD 20817, USA
| | - Hong Xiao
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrence Township, NJ 08648, USA; (H.X.)
| | | | - Renuka Raorane
- Evidera, Ltd., UK Office, The Ark, 201 Talgarth Rd, London W6 8BJ, UK; (C.v.W.-M.); (R.R.)
| | - Sohan Deshpande
- Evidera, Ltd., UK Office, The Ark, 201 Talgarth Rd, London W6 8BJ, UK; (C.v.W.-M.); (R.R.)
| | - Sven L. Klijn
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrence Township, NJ 08648, USA; (H.X.)
| | - Aylin Yucel
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrence Township, NJ 08648, USA; (H.X.)
| |
Collapse
|
17
|
Liu L, Song X, Dong W, Li Z, Guo D. Case report: Safety and efficacy of synergistic treatment using selinexor and azacitidine in patients with atypical chronic myeloid leukemia with resistance to decitabine. Front Oncol 2024; 14:1353818. [PMID: 38384813 PMCID: PMC10879427 DOI: 10.3389/fonc.2024.1353818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Background Atypical chronic myeloid leukemia (aCML) is a BCR::ABL1 negative myelodysplastic/myeloproliferative neoplasm with poor overall survival. Some patients can be treated by allogeneic hematopoietic stem cell transplantation (allo-HSCT) from suitable donors. The effectiveness of decitabine or azacitidine (AZA) has recently been reported; however, their combined efficacy with selinexor has not yet been reported. Case description In this study, we report the case of a patient with aCML who was successfully treated with selinexor combined with AZA. A 67-year-old man with a history of gastric mucosa-associated lymphoid tissue (MALT) lymphoma was admitted to the hospital with fatigue and emaciation. He was diagnosed with aCML and no longer responded to decitabine treatment after undergoing seven cycles. The patient was subsequently administered hydroxyurea (HU), selinexor, and AZA. After four courses of combination therapy, his blood cell counts improved; he no longer required transfusions and was able to discontinue HU. The patient continued receiving selinexor and AZA without severe complications. This case is the first to show that combinatorial selinexor and AZA therapy can effectively treat aCML. Conclusion Our case sheds light on the importance of selinexor and AZA combined therapy in the exploration of new treatment strategies for aCML. Moreover, this treatment approach offers the possibility of bridging with allo-HSCT.
Collapse
Affiliation(s)
- Lu Liu
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Xiaofeng Song
- Department of Hand and Foot Surgery, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Wenhao Dong
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Zhao Li
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Dongmei Guo
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| |
Collapse
|
18
|
Jain T, Tsai HL, Elmariah H, Vachhani P, Karantanos T, Wall SA, Gondek LP, Bashey A, Keyzner A, Tamari R, Grunwald MR, Abedin S, Nadiminti KV, Iqbal M, Gerds AT, Viswabandya A, McCurdy SR, Al Malki MM, Varadhan R, Ali H, Gupta V, Jones RJ, Otoukesh S. Haploidentical donor hematopoietic cell transplantation for myelodysplastic/myeloproliferative overlap neoplasms: results from a North American collaboration. Haematologica 2023; 108:3321-3332. [PMID: 37408464 PMCID: PMC10690921 DOI: 10.3324/haematol.2023.283426] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023] Open
Abstract
Haploidentical donors offer a potentially readily available donor, especially for non-White patients, for hematopoietic cell transplantation (HCT). In this North American collaboration, we retrospectively analyzed outcomes of first HCT using haploidentical donor and post-transplantation cyclophosphamide (PTCy) in myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) overlap neoplasms (MDS/MPN). We included 120 consecutive patients who underwent HCT using a haploidentical donor for MDS/MPN across 15 centers. Median age was 62.5 years and 38% were of non-White/Caucasian ethnicity. The median follow-up was 2.4 years. Graft failure was reported in seven of 120 (6%) patients. At 3 years, nonrelapse mortality (NRM) was 25% (95% confidence interval [CI]: 17-34), relapse 27% (95% CI: 18-36), grade 3-4 acute graftversus- host disease 12% (95% CI: 6-18), chronic graft-versus-host disease requiring systemic immunosuppression 14% (95% CI: 7-20), progression-free survival (PFS) 48% (95% CI: 39-59), and overall survival (OS) 56% (95% CI: 47-67). On multivariable analysis, NRM was statistically significantly associated with advancing age at HCT (per decade increment, subdistribution hazard ratio [sdHR] =3.28; 95% CI: 1.30-8.25); relapse with the presence of mutation in EZH2/RUNX1/SETBP1 (sdHR=2.61; 95% CI: 1.06-6.44); PFS with advancing age at HCT (per decade increment, HR=1.98, 95% CI: 1.13-3.45); and OS with advancing age at HCT (per decade increment, HR=2.01; 95% CI: 1.11-3.63) and splenomegaly at HCT/prior splenectomy (HR=2.20; 95% CI: 1.04-4.65). Haploidentical donors are a viable option for HCT in MDS/MPN, especially for those disproportionately represented in the unrelated donor registry. Hence, donor mismatch should not preclude HCT for patients with MDS/MPN, an otherwise incurable malignancy. In addition to patient age, disease-related factors including splenomegaly and high-risk mutations dominate outcomes following HCT.
Collapse
Affiliation(s)
- Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans street, Baltimore, MD, USA 21287.
| | - Hua-Ling Tsai
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 550 North Broadway, Baltimore, MD, USA 21287
| | - Hany Elmariah
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA 33612
| | - Pankit Vachhani
- Division of Hematology and Oncology, O'Neal Comprehensive Cancer Center, University of Alabama, 1802 6th Ave S, Birmingham, AL, USA 35294
| | - Theodoros Karantanos
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans street, Baltimore, MD, USA 21287
| | - Sarah A Wall
- Division of Hematology, The Ohio State University - James Comprehensive Cancer Center, 1800 Cannon Drive, 11th Floor, Columbus, OH, USA 43210
| | - Lukasz P Gondek
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans street, Baltimore, MD, USA 21287
| | - Asad Bashey
- Blood and Marrow Transplant Program, Northside Hospital, 5670 Peachtree Dunwoody Road, Atlanta, GA, USA 30342
| | - Alla Keyzner
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place P.O. Box 1410, New York, NY, USA 10029
| | - Roni Tamari
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Department of Medicine, Weill Cornell Medical College, 1275 York avenue, New York, NY, USA 10065
| | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, 1021 Morehead Medical Drive, LCI Building 2, Suite 60100, Charlotte, NC, USA 28204
| | - Sameem Abedin
- Division of Hematology/Oncology, Medical College of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI, USA 53226
| | - Kalyan Vg Nadiminti
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI, USA 53792
| | - Madiha Iqbal
- Department of Hematology-Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, USA 32224
| | - Aaron T Gerds
- Department of Hematology and Medical Oncology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, USA 44195
| | - Auro Viswabandya
- Princess Margaret Cancer Centre, University of Toronto, 610 University Ave, Toronto, Canada M5G 2C1
| | - Shannon R McCurdy
- University of Pennsylvania, 3400 Civic center road, 12 South Pavilion, Philadelphia, PA 19104
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, 1500 East Duarte Road, Duarte, CA, USA 91010
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 550 North Broadway, Baltimore, MD, USA 21287
| | - Haris Ali
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, 1500 East Duarte Road, Duarte, CA, USA 91010
| | - Vikas Gupta
- Princess Margaret Cancer Centre, University of Toronto, 610 University Ave, Toronto, Canada M5G 2C1
| | - Richard J Jones
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans street, Baltimore, MD, USA 21287
| | - Salman Otoukesh
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, 1500 East Duarte Road, Duarte, CA, USA 91010
| |
Collapse
|
19
|
Bewersdorf JP, Stahl M, Taylor J, Mi X, Chandhok NS, Watts J, Derkach A, Wysocki M, Lu SX, Bourcier J, Hogg SJ, Rahman J, Chaudhry S, Totiger TM, Abdel-Wahab O, Stein EM. E7820, an anti-cancer sulfonamide, degrades RBM39 in patients with splicing factor mutant myeloid malignancies: a phase II clinical trial. Leukemia 2023; 37:2512-2516. [PMID: 37814121 PMCID: PMC10681888 DOI: 10.1038/s41375-023-02050-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Medicine; Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Justin Taylor
- Leukemia Program, Department of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Xiaoli Mi
- Department of Medicine; Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Namrata Sonia Chandhok
- Leukemia Program, Department of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Justin Watts
- Leukemia Program, Department of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Andriy Derkach
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mateusz Wysocki
- Department of Medicine; Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sydney X Lu
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessie Bourcier
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon J Hogg
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jahan Rahman
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sana Chaudhry
- Leukemia Program, Department of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Tulasigeri M Totiger
- Leukemia Program, Department of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Omar Abdel-Wahab
- Department of Medicine; Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eytan M Stein
- Department of Medicine; Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
20
|
Pleyer L, Vaisband M, Drost M, Pfeilstöcker M, Stauder R, Heibl S, Sill H, Girschikofsky M, Stampfl-Mattersberger M, Pichler A, Hartmann B, Petzer A, Schreder M, Schmitt CA, Vallet S, Melchardt T, Zebisch A, Pichler P, Zaborsky N, Machherndl-Spandl S, Wolf D, Keil F, Hasenauer J, Larcher-Senn J, Greil R. Cox proportional hazards deep neural network identifies peripheral blood complete remission to be at least equivalent to morphologic complete remission in predicting outcomes of patients treated with azacitidine-A prospective cohort study by the AGMT. Am J Hematol 2023; 98:1685-1698. [PMID: 37548390 DOI: 10.1002/ajh.27046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/07/2023] [Accepted: 07/16/2023] [Indexed: 08/08/2023]
Abstract
The current gold standard of response assessment in patients with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML) is morphologic complete remission (CR) and CR with incomplete count recovery (CRi), both of which require an invasive BM evaluation. Outside of clinical trials, BM evaluations are only performed in ~50% of patients during follow-up, pinpointing a clinical need for response endpoints that do not necessitate BM assessments. We define and validate a new response type termed "peripheral blood complete remission" (PB-CR) that can be determined from the differential blood count and clinical parameters without necessitating a BM assessment. We compared the predictive value of PB-CR with morphologic CR/CRi in 1441 non-selected, consecutive patients diagnosed with MDS (n = 522; 36.2%), CMML (n = 132; 9.2%), or AML (n = 787; 54.6%), included within the Austrian Myeloid Registry (aMYELOIDr; NCT04438889). Time-to-event analyses were adjusted for 17 covariates remaining in the final Cox proportional hazards (CPH) model. DeepSurv, a CPH neural network model, and permutation-based feature importance were used to validate results. 1441 patients were included. Adjusted median overall survival for patients achieving PB-CR was 22.8 months (95%CI 18.9-26.2) versus 10.4 months (95%CI 9.7-11.2) for those who did not; HR = 0.366 (95%CI 0.303-0.441; p < .0001). Among patients achieving CR, those additionally achieving PB-CR had a median adjusted OS of 32.6 months (95%CI 26.2-49.2) versus 21.7 months (95%CI 16.9-27.7; HR = 0.400 [95%CI 0.190-0.844; p = .0161]) for those who did not. Our deep neural network analysis-based findings from a large, prospective cohort study indicate that BM evaluations solely for the purpose of identifying CR/CRi can be omitted.
Collapse
Affiliation(s)
- Lisa Pleyer
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Marc Vaisband
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Manuel Drost
- Assign Data Management and Biostatistics GmbH, Innsbruck, Austria
| | - Michael Pfeilstöcker
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 3rd Medical Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University Vienna, Vienna, Austria
| | - Reinhard Stauder
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Sonja Heibl
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 4th Medical Department of Internal Medicine, Hematology, Internistic Oncology and Palliative Medicine, Klinikum Wels-Grieskirchen GmbH, Wels, Austria
| | - Heinz Sill
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Michael Girschikofsky
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 1st Medical Department, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz GmbH Elisabethinen, Linz, Austria
| | - Margarete Stampfl-Mattersberger
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Internal Medicine 2, Wiener Gesundheitsverbund, Klinik Donaustadt, Vienna, Austria
| | - Angelika Pichler
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Internal Medicine, Hematology and Internal Oncology, LKH Hochsteiermark, Leoben, Austria
| | - Bernd Hartmann
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Internal Medicine 2, Landeskrankenhaus Feldkirch, Feldkirch, Austria
| | - Andreas Petzer
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Internal Medicine I: Medical Oncology and Hematology, Ordensklinikum Linz GmbH, Barmherzige Schwestern, Linz, Austria
| | - Martin Schreder
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 1st Department of Internal Medicine, Center for Oncology and Hematology, Wiener Gesundheitsverbund, Klinik Ottakring, Vienna, Austria
| | - Clemens A Schmitt
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Hematology and Internal Oncology, Kepler University Hospital, Johannes Kepler University, Linz, Austria
- Charité-University Medical Center, Molecular Cancer Research Center, Berlin, Germany
| | - Sonia Vallet
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- University Hospital Krems, Department of Internal Medicine 2, Karl Landsteiner Private University of Health Sciences, Krems, Austria
| | - Thomas Melchardt
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Armin Zebisch
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Division of Hematology, Medical University of Graz, Graz, Austria
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Petra Pichler
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Clinical Department for Internal Medicine, University Hospital St Poelten, Karl Landsteiner University of Health Sciences, St Poelten, Austria
| | - Nadja Zaborsky
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria
| | - Sigrid Machherndl-Spandl
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 1st Medical Department, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz GmbH Elisabethinen, Linz, Austria
| | - Dominik Wolf
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Felix Keil
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- 3rd Medical Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University Vienna, Vienna, Austria
| | - Jan Hasenauer
- Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | - Richard Greil
- Austrian Group of Medical Tumor Therapy (AGMT) Study Group, Vienna, Austria
- Salzburg Cancer Research Institute (SCRI) Center for Clinical Cancer and Immunology Trials (CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
21
|
Johnson B, Shuai Y, Schweinsberg J, Curtius K. cloneRate: fast estimation of single-cell clonal dynamics using coalescent theory. Bioinformatics 2023; 39:btad561. [PMID: 37699006 PMCID: PMC10534056 DOI: 10.1093/bioinformatics/btad561] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/25/2023] [Indexed: 09/14/2023] Open
Abstract
MOTIVATION While evolutionary approaches to medicine show promise, measuring evolution itself is difficult due to experimental constraints and the dynamic nature of body systems. In cancer evolution, continuous observation of clonal architecture is impossible, and longitudinal samples from multiple timepoints are rare. Increasingly available DNA sequencing datasets at single-cell resolution enable the reconstruction of past evolution using mutational history, allowing for a better understanding of dynamics prior to detectable disease. There is an unmet need for an accurate, fast, and easy-to-use method to quantify clone growth dynamics from these datasets. RESULTS We derived methods based on coalescent theory for estimating the net growth rate of clones using either reconstructed phylogenies or the number of shared mutations. We applied and validated our analytical methods for estimating the net growth rate of clones, eliminating the need for complex simulations used in previous methods. When applied to hematopoietic data, we show that our estimates may have broad applications to improve mechanistic understanding and prognostic ability. Compared to clones with a single or unknown driver mutation, clones with multiple drivers have significantly increased growth rates (median 0.94 versus 0.25 per year; P = 1.6×10-6). Further, stratifying patients with a myeloproliferative neoplasm (MPN) by the growth rate of their fittest clone shows that higher growth rates are associated with shorter time to MPN diagnosis (median 13.9 versus 26.4 months; P = 0.0026). AVAILABILITY AND IMPLEMENTATION We developed a publicly available R package, cloneRate, to implement our methods (Package website: https://bdj34.github.io/cloneRate/). Source code: https://github.com/bdj34/cloneRate/.
Collapse
Affiliation(s)
- Brian Johnson
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Yubo Shuai
- Department of Mathematics, University of California San Diego, La Jolla, CA 92093, United States
| | - Jason Schweinsberg
- Department of Mathematics, University of California San Diego, La Jolla, CA 92093, United States
| | - Kit Curtius
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States
- VA San Diego Healthcare System, San Diego, CA 92161, United States
| |
Collapse
|
22
|
Cai P, Liu S, Duan L, Huo L, Wu D, Chen S, Yang R, Yang X. Sustained Response to Ruxolitinib of Eosinophilia-Associated Myeloproliferative Neoplasm with Translocation t(8;9)(p21;p24). Acta Haematol 2023; 146:397-400. [PMID: 37562364 DOI: 10.1159/000510281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 07/17/2020] [Indexed: 08/12/2023]
Abstract
The translocation t(8;9) produces the fusion gene PCM1-JAK2, resulting in the continuous activation of the JAK2 tyrosine kinase. Myelodysplastic/myeloproliferative neoplasms are the most common disease with t(8;9)/PCM1-JAK2. Individuals with this abnormality have similar features, and JAK2 kinase inhibitor (ruxolitinib) is an effective treatment of the condition. The long-term remission results of ruxolitinib are varied. It is important to determine the response to ruxolitinib. Here, we describe a patient who has been diagnosed with eosinophilia-associated myeloproliferative neoplasm with t(8;9)(p21;p24). This patient has achieved sustained response for >1 year since the administration of ruxolitinib.
Collapse
Affiliation(s)
- Ping Cai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Suhui Liu
- Department of Hematology, NanYang Central Hospital, Henan, China
| | - Lijuan Duan
- Department of Hematology, NanYang Central Hospital, Henan, China
| | - Li Huo
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ruyu Yang
- Department of Hematology, NanYang Central Hospital, Henan, China
| | - Xiaofei Yang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Short NJ, Muftuoglu M, Ong F, Nasr L, Macaron W, Montalban-Bravo G, Alvarado Y, Basyal M, Daver N, Dinardo CD, Borthakur G, Jain N, Ohanian M, Jabbour E, Issa GC, Qiao W, Huang X, Kanagal-Shamanna R, Patel KP, Bose P, Ravandi F, Delumpa R, Abramova R, Garcia-Manero G, Andreeff M, Cortes J, Kantarjian H. A phase 1/2 study of azacitidine, venetoclax and pevonedistat in newly diagnosed secondary AML and in MDS or CMML after failure of hypomethylating agents. J Hematol Oncol 2023; 16:73. [PMID: 37422688 PMCID: PMC10329789 DOI: 10.1186/s13045-023-01476-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Pevonedistat is a first-in-class, small molecular inhibitor of NEDD8-activating enzyme that has clinical activity in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). Preclinical data suggest synergy of pevonedistat with azacitidine and venetoclax. METHODS This single-center, phase 1/2 study evaluated the combination of azacitidine, venetoclax and pevonedistat in older adults with newly diagnosed secondary AML or with MDS or chronic myelomonocytic leukemia (CMML) after failure of hypomethylating agents. Patients received azacitidine 75 mg/m2 IV on days 1-7, venetoclax at maximum dose of 200-400 mg orally on days 1-21 (AML cohort) or days 1-14 (MDS/CMML cohort) and pevonedistat 20 mg/m2 IV on days 1, 3 and 5 for up to 24 cycles. The primary endpoints for the phase 2 portion of the study were the CR/CRi rate in the AML cohort and the overall response rate (CR + mCR + PR + HI) in the MDS/CMML cohort. FINDINGS Forty patients were enrolled (32 with AML and 8 with MDS/CMML). In the AML cohort, the median age was 74 years (range 61-86 years), and 27 patients (84%) had at least one adverse risk cyto-molecular feature, including 15 (47%) with a TP53 mutation or MECOM rearrangement; seventeen patients (53%) had received prior therapy for a preceding myeloid disorder. The CR/CRi rate was 66% (CR 50%; CRi 16%), and the median overall survival (OS) was 8.1 months. In the MDS/CMML cohort, 7 patients (87%) were high or very high risk by the IPSS-R. The overall response rate was 75% (CR 13%; mCR with or without HI 50%; HI 13%). The most common grade 3-4 adverse events were infection in 16 patients (35%), febrile neutropenia in 10 patients (25%) and hypophosphatemia in 9 patients (23%). In an exploratory analysis, early upregulation of NOXA expression was observed, with subsequent decrease in MCL-1 and FLIP, findings consistent with preclinical mechanistic studies of pevonedistat. Upregulation of CD36 was observed, which may have contributed to therapeutic resistance. CONCLUSIONS The triplet combination of azacitidine, venetoclax and pevonedistat shows encouraging activity in this very poor-risk population of patients with AML, MDS or CMML. Trial registration ClinicalTrials.gov (NCT03862157).
Collapse
Affiliation(s)
- Nicholas J Short
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| | - Muharrem Muftuoglu
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Faustine Ong
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Lewis Nasr
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Walid Macaron
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Guillermo Montalban-Bravo
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Yesid Alvarado
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mahesh Basyal
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Naval Daver
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Courtney D Dinardo
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Gautam Borthakur
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Nitin Jain
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Maro Ohanian
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Elias Jabbour
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Ghayas C Issa
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Farhad Ravandi
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Ricardo Delumpa
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Regina Abramova
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Michael Andreeff
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Jorge Cortes
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Hagop Kantarjian
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| |
Collapse
|
24
|
Shomali W, Colucci P, George TI, Kiladjian JJ, Langford C, Patel JL, Reiter A, Vannucchi AM, Gotlib J. Comprehensive response criteria for myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions: a proposal from the MLN International Working Group. Leukemia 2023; 37:981-987. [PMID: 37076693 PMCID: PMC10169632 DOI: 10.1038/s41375-023-01859-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 04/21/2023]
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tracy I George
- ARUP Laboratories and University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Cheryl Langford
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Jay L Patel
- ARUP Laboratories and University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
25
|
Jansko-Gadermeir B, Leisch M, Gassner FJ, Zaborsky N, Dillinger T, Hutter S, Risch A, Melchardt T, Egle A, Drost M, Larcher-Senn J, Greil R, Pleyer L. Myeloid NGS Analyses of Paired Samples from Bone Marrow and Peripheral Blood Yield Concordant Results: A Prospective Cohort Analysis of the AGMT Study Group. Cancers (Basel) 2023; 15:2305. [PMID: 37190237 PMCID: PMC10136651 DOI: 10.3390/cancers15082305] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Background: Next generation sequencing (NGS) has become indispensable for diagnosis, risk stratification, prognostication, and monitoring of response in patients with myeloid neoplasias. Guidelines require bone marrow evaluations for the above, which are often not performed outside of clinical trials, indicating a need for surrogate samples. Methods: Myeloid NGS analyses (40 genes and 29 fusion drivers) of 240 consecutive, non-selected, prospectively collected, paired bone marrow/peripheral blood samples were compared. Findings: Very strong correlation (r = 0.91, p < 0.0001), high concordance (99.6%), sensitivity (98.8%), specificity (99.9%), positive predictive value (99.8%), and negative predictive value (99.6%) between NGS analyses of paired samples was observed. A total of 9/1321 (0.68%) detected mutations were discordant, 8 of which had a variant allele frequency (VAF) ≤ 3.7%. VAFs between peripheral blood and bone marrow samples were very strongly correlated in the total cohort (r = 0.93, p = 0.0001) and in subgroups without circulating blasts (r = 0.92, p < 0.0001) or with neutropenia (r = 0.88, p < 0.0001). There was a weak correlation between the VAF of a detected mutation and the blast count in either the peripheral blood (r = 0.19) or the bone marrow (r = 0.11). Interpretation: Peripheral blood samples can be used to molecularly classify and monitor myeloid neoplasms via NGS without loss of sensitivity/specificity, even in the absence of circulating blasts or in neutropenic patients.
Collapse
Affiliation(s)
- Bettina Jansko-Gadermeir
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
- Laboratory for Molecular Cytology (MZL), 5020 Salzburg, Austria
- Department of Biosciences and Medical Biology, Allergy-Cancer-BioNano Research Centre, University of Salzburg, 5020 Salzburg, Austria
| | - Michael Leisch
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Austrian Group for Medical Tumor Therapy (AGMT) Study Group, 1180 Vienna, Austria
| | - Franz J. Gassner
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
| | - Nadja Zaborsky
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
| | - Thomas Dillinger
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Laboratory for Molecular Cytology (MZL), 5020 Salzburg, Austria
| | - Sonja Hutter
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Laboratory for Molecular Cytology (MZL), 5020 Salzburg, Austria
| | - Angela Risch
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Department of Biosciences and Medical Biology, Allergy-Cancer-BioNano Research Centre, University of Salzburg, 5020 Salzburg, Austria
| | - Thomas Melchardt
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Austrian Group for Medical Tumor Therapy (AGMT) Study Group, 1180 Vienna, Austria
| | - Alexander Egle
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
- Austrian Group for Medical Tumor Therapy (AGMT) Study Group, 1180 Vienna, Austria
| | - Manuel Drost
- Assign Data Management and Biostatistics GmbH, 6020 Innsbruck, Austria
| | | | - Richard Greil
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
- Laboratory for Molecular Cytology (MZL), 5020 Salzburg, Austria
- Austrian Group for Medical Tumor Therapy (AGMT) Study Group, 1180 Vienna, Austria
| | - Lisa Pleyer
- Salzburg Cancer Research Institute (SCRI), Center for Clinical Cancer and Immunology Trials (CCCIT), 5020 Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
- Laboratory for Molecular Cytology (MZL), 5020 Salzburg, Austria
- Austrian Group for Medical Tumor Therapy (AGMT) Study Group, 1180 Vienna, Austria
| |
Collapse
|
26
|
Sun Y, Wang Q, Zhang X, Zhang Z, Wang Q, Cen J, Zhu M, Pan J, Liu D, Wu D, Cai Y, Chen S. Molecular genetics and management of world health organization defined atypical chronic myeloid leukemia. Ann Hematol 2023; 102:777-785. [PMID: 36735076 DOI: 10.1007/s00277-023-05106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023]
Abstract
Atypical chronic myeloid leukemia (CML) is a rare BCR::ABL1-negative hematopoietic stem cell disease characterized by granulocytic proliferation and granulocytic dysplasia. Due to both the challenging diagnosis and the rarity of atypical CML, comprehensive molecular annotation-based analyses of this disease population have been scarce, and it is currently difficult to identify the optimal treatment for atypical CML. To explore atypical CML genomic landscape and treatment options, we performed a systematic retrospective of the clinical data and outcomes of 31 atypical CML patients. We observed that the molecular landscape of atypical CML was highly heterogeneous, with multiple molecular events driving its pathogenesis. Patients with atypical CML had a low response to current therapies, with an overall response rate (ORR) of 33.3% to hypomethylating agent (HMA)-based therapy. The current treatment strategies, including hematopoietic stem cell transplantation (HSCT), did not improve overall survival (OS) in atypical CML patients, with a median survival of 20 months. Thus, the benefits from HSCT and candidates for HSCT remain to be further evaluated. Acute myeloid leukemia (AML)-like chemotherapy followed by bridging allogeneic HSCT may be an ideal regimen for suitable individuals. The large-scale and prospective clinical studies will help to address the dilemma.
Collapse
Affiliation(s)
- Yingxin Sun
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Qinrong Wang
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xingxia Zhang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, Huai'an, China
| | - Zhiyu Zhang
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Qian Wang
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Jiannong Cen
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Mingqing Zhu
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Jinlan Pan
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Dandan Liu
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Depei Wu
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Yifeng Cai
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.
| | - Suning Chen
- Department of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China. .,Department of Thrombosis and Hemostasis, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.
| |
Collapse
|
27
|
Patnaik MM, Tefferi A. Atypical chronic myeloid leukemia and myelodysplastic/myeloproliferative neoplasm, not otherwise specified: 2023 update on diagnosis, risk stratification, and management. Am J Hematol 2023; 98:681-689. [PMID: 36601682 DOI: 10.1002/ajh.26828] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023]
Abstract
DISEASE OVERVIEW Atypical chronic myeloid leukemia (aCML) and myelodysplastic/myeloproliferative (MDS/MPN) neoplasms, not otherwise specified (NOS), are MDS/MPN overlap neoplasms characterized by leukocytosis, in the absence of monocytosis and eosinophilia, with <20% blasts in the blood and bone marrow. DIAGNOSIS aCML, previously known as aCML, BCR::ABL1 negative, was renamed as aCML by the ICC classification, and as MDS/MPN with neutrophilia by the 5th edition of the WHO classification. This entity is characterized by dysplastic neutrophilia with immature myeloid cells comprising ≥10% of the white blood cell count, with prominent dysgranulopoiesis. MDS/MPN-NOS consists of MDS/MPN overlap neoplasms not meeting criteria for defined categories such as chronic myelomonocytic leukemia (CMML), MDS/MPN-ring sideroblasts-thrombocytosis (MDS/MPN-RS-T), and aCML. MUTATIONS AND KARYOTYPE Cytogenetic abnormalities are seen in 40-50% of patients in both categories. In aCML, somatic mutations commonly encountered include ASXL1, SETBP1, ETNK1, and EZH2 whereas MDS/MPN-NOS can be further stratified by mutational profiles into CMML-like, MDS/MPN-RS-T-like, aCML-like, TP35-mutated, and "others", respectively. RISK STRATIFICATION The Mayo Clinic aCML model stratifies patients based on age >67 years, hemoglobin <10 g/dl, and the presence of TET2 mutations into low-risk (0-1 points) and high-risk (>2 points) groups, with median survivals of 18 and 7 months, respectively. MDS/MPN-NOS patients have traditionally been risk stratified using MDS risk models such as IPSS and IPSS-R. TREATMENT Leukocytosis and anemia are managed like lower risk MPN and MDS. DNMT inhibitors have been used in both entities with suboptimal response rates. Allogeneic stem cell transplant remains the only curative strategy but is associated with high morbidity and mortality.
Collapse
MESH Headings
- Humans
- Aged
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/diagnosis
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/therapy
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/therapy
- Leukocytosis
- Myelodysplastic-Myeloproliferative Diseases/diagnosis
- Myelodysplastic-Myeloproliferative Diseases/genetics
- Myelodysplastic-Myeloproliferative Diseases/therapy
- Leukemia, Myelomonocytic, Chronic/diagnosis
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/therapy
- Thrombocytosis/genetics
- Mutation
- Risk Assessment
Collapse
Affiliation(s)
- Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ayalew Tefferi
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
28
|
Jain T, Tsai HL, Elmariah H, Vachhani P, Karantanos T, Wall S, Gondek L, Bashey A, Keyzner A, Tamari R, Grunwald M, Abedin S, Nadiminti K, Iqbal M, Gerds A, Viswabandya A, McCurdy S, Malki MA, Varadhan R, Ali H, Gupta V, Jones RJ, Otoukesh S. Haploidentical Donor Blood or Marrow Transplantation for Myelodysplastic/Myeloproliferative Overlap Neoplasms: Results from a North American Collaboration. RESEARCH SQUARE 2023:rs.3.rs-2691216. [PMID: 36993719 PMCID: PMC10055643 DOI: 10.21203/rs.3.rs-2691216/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Haploidentical donors offer a potentially readily available donor, especially for non-White patients, for blood or marrow transplantation (BMT). In this collaboration across North America, we retrospectively analyzed outcomes of first BMT using haploidentical donor and posttransplantation cyclophosphamide (PTCy) in MDS/MPN-overlap neoplasms (MDS/MPN), an otherwise incurable hematological neoplasm. We included 120 patients, 38% of non-White/Caucasian ethnicity, across 15 centers with median age at BMT 62.5 years. The median follow-up is 2.4 years. Graft failure was reported in 6% patients. At 3-years, nonrelapse mortality (NRM) was 25%, relapse 27%, grade 3-4 acute graft versus host disease (GVHD) 12%, chronic GVHD requiring systemic immunosuppression 14%, progression-free survival (PFS) 48% and overall survival (OS) 56%. On multivariable analysis, statistically significant associations included older age at BMT (per decade increment) with NRM (sdHR 3.28, 95%CI 1.30-8.25), PFS (HR 1.98, 95% 1.13-3.45) and OS (HR 2.01, 95% CI 1.11-3.63), presence of mutation in EZH2/RUNX1/SETBP1 with relapse (sdHR 2.61, 95%CI 1.06-6.44), and splenomegaly at BMT/prior splenectomy with OS (HR 2.20, 95%CI 1.04-4.65). Haploidentical donors are a viable option for BMT in MDS/MPN, especially for those disproportionately represented in the unrelated donor registry. Disease-related factors including splenomegaly and high-risk mutations dominate outcomes following BMT.
Collapse
Affiliation(s)
- Tania Jain
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University
| | | | | | - Pankit Vachhani
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham
| | | | | | | | | | | | - Roni Tamari
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Itzykson R, Santini V, Thepot S, Ades L, Chaffaut C, Giagounidis A, Morabito M, Droin N, Lübbert M, Sapena R, Nimubona S, Goasguen J, Wattel E, Zini G, Torregrosa Diaz JM, Germing U, Pelizzari AM, Park S, Jaekel N, Metzgeroth G, Onida F, Navarro R, Patriarca A, Stamatoullas A, Götze K, Puttrich M, Mossuto S, Solary E, Gloaguen S, Chevret S, Chermat F, Platzbecker U, Fenaux P. Decitabine Versus Hydroxyurea for Advanced Proliferative Chronic Myelomonocytic Leukemia: Results of a Randomized Phase III Trial Within the EMSCO Network. J Clin Oncol 2022; 41:1888-1897. [PMID: 36455187 DOI: 10.1200/jco.22.00437] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
PURPOSE Hydroxyurea (HY) is a reference treatment of advanced myeloproliferative neoplasms. We conducted a randomized phase III trial comparing decitabine (DAC) and HY in advanced myeloproliferative chronic myelomonocytic leukemias (CMML). PATIENTS AND METHODS Newly diagnosed myeloproliferative CMML patients with advanced disease were randomly assigned 1:1 to intravenous DAC (20 mg/m2/d days 1-5) or HY (1-4 g/d) in 28-day cycles. The primary end point was event-free survival (EFS), events being death and acute myelomonocytic leukemia (AML) transformation or progression. RESULTS One-hundred seventy patients received DAC (n = 84) or HY (n = 86). Median age was 72 and 74 years, and median WBC count 32.5 × 109/L and 31.2 × 109/L in the DAC and HY arms, respectively. Thirty-three percent of DAC and 31% of HY patients had CMML-2. Patients received a median of five DAC and six HY cycles. With a median follow-up of 17.5 months, median EFS was 12.1 months in the DAC arm and 10.3 months in the HY arm (hazard ratio [HR], 0.83; 95% CI, 0.59 to 1.16; P = .27). There was no significant interaction between treatment effect and blast or platelet count, anemia, CMML Prognostic Scoring System, Groupe Francophone des Myelodysplasies, or CMML Prognostic Scoring System–mol risk. Fifty-three (63%) DAC patients achieved a response compared with 30 (35%) HY patients ( P = .0004). Median duration of response was similar in both arms (DAC, 16.3 months; HY, 17.4 months; P = .90). Median overall survival was 18.4 months in the DAC arm and 21.9 months in the HY arm ( P = .67). Compared with HY, DAC significantly reduced the risk of CMML progression or transformation to acute myelomonocytic leukemia (cause-specific HR, 0.62; 95% CI, 0.41 to 0.94; P = .005) at the expense of death without progression or transformation (cause-specific HR, 1.55; 95% CI, 0.82 to 2.9; P = .04). CONCLUSION Compared with HY, frontline treatment with DAC did not improve EFS in patients with advanced myeloproliferative CMML (ClinicalTrials.gov identifier: NCT02214407 ).
Collapse
Affiliation(s)
- Raphael Itzykson
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université de Paris, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, Paris, France
- Groupe Francophone des Myélodysplasies, Paris, France
| | - Valeria Santini
- MDS Unit, DMSC; AOU Careggi, University of Florence, Florence, Italy
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM-ets), Bologna, Italy
| | - Sylvain Thepot
- Groupe Francophone des Myélodysplasies, Paris, France
- Hematology Department CHU Angers, Université Angers, Angers, France
| | - Lionel Ades
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
- Groupe Francophone des Myélodysplasies, Paris, France
- Service Hématologie Seniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Cendrine Chaffaut
- SBIM, APHP, Hôpital Saint-Louis, INSERM, UMR-1153, ECSTRA Team, Paris, France
| | - Aristoteles Giagounidis
- Marien Hospital, Klinik für Hämatologie, Onkologie und klinische Immunologie, D-Düsseldorf, Germany
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
| | - Margot Morabito
- Université Paris Saclay, INSERM U1287, Gustave Roussy Cancer Center, Villejuif, France
| | - Nathalie Droin
- Université Paris Saclay, INSERM U1287, Gustave Roussy Cancer Center, Villejuif, France
| | - Michael Lübbert
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine—University Medical Center Freiburg, Freiburg, Germany
| | - Rosa Sapena
- Groupe Francophone des Myélodysplasies, Paris, France
| | - Stanislas Nimubona
- Groupe Francophone des Myélodysplasies, Paris, France
- Service Hématologie Clinique adulte, CHU de Rennes, Rennes, France
| | | | - Eric Wattel
- Groupe Francophone des Myélodysplasies, Paris, France
- Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Gina Zini
- Hematology, Università Cattolica del S. Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Jose Miguel Torregrosa Diaz
- Groupe Francophone des Myélodysplasies, Paris, France
- Service d’Hématologie Oncologique et Thérapie Cellulaire, CIC INSERM 1402, University Hospital of Poitiers, Poitiers, France
| | - Ulrich Germing
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- Heinrich-Heine University Düsseldorf, Universitätsklinik Düsseldorf, Klinik für Hämatologie, Onkologie und Klinische Immunologie, Düsseldorf, Germany
| | - Anna Maria Pelizzari
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM-ets), Bologna, Italy
- Hematology Unit, ASST Spedali Civili, Brescia, Italy
| | - Sophie Park
- Groupe Francophone des Myélodysplasies, Paris, France
- Université Grenoble Alpes, Hematology Department, CHU Grenoble Alpes, Grenoble, France
| | - Nadja Jaekel
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- University Hospital Halle, Halle, Germany
| | - Georgia Metzgeroth
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Francesco Onida
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM-ets), Bologna, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico - University of Milan, Hematology-BMT Unit, Milan, Italy
| | - Robert Navarro
- Groupe Francophone des Myélodysplasies, Paris, France
- Service d’Hématologie, CHU Montpellier, Montpellier, France
| | - Andrea Patriarca
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM-ets), Bologna, Italy
- Hematology Unit, AOU «Maggiore della Carità» and University of Eastern Piedmont, I-28100, Novara, Italy
| | - Aspasia Stamatoullas
- Groupe Francophone des Myélodysplasies, Paris, France
- Centre Henri Becquerel, Rouen, France
| | - Katharina Götze
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- Technical University of Munich, Department of Medicine III, Munich, Germany
| | - Martin Puttrich
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- GWT-TUD GmbH, Dresden, Germany
| | - Sandra Mossuto
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM-ets), Bologna, Italy
| | - Eric Solary
- Groupe Francophone des Myélodysplasies, Paris, France
- Université Paris Saclay, INSERM U1287, Gustave Roussy Cancer Center, Villejuif, France
- Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Silke Gloaguen
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- Clinic and Polyclinic for Hematology, Cellular Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Sylvie Chevret
- SBIM, APHP, Hôpital Saint-Louis, INSERM, UMR-1153, ECSTRA Team, Paris, France
| | | | - Uwe Platzbecker
- Deutsche MDS-Studiengruppe, D-04103 Leipzig, Germany
- Clinic and Polyclinic for Hematology, Cellular Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Pierre Fenaux
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
- Groupe Francophone des Myélodysplasies, Paris, France
- Service Hématologie Seniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
30
|
Duncavage EJ, Bagg A, Hasserjian RP, DiNardo CD, Godley LA, Iacobucci I, Jaiswal S, Malcovati L, Vannucchi AM, Patel KP, Arber DA, Arcila ME, Bejar R, Berliner N, Borowitz MJ, Branford S, Brown AL, Cargo CA, Döhner H, Falini B, Garcia-Manero G, Haferlach T, Hellström-Lindberg E, Kim AS, Klco JM, Komrokji R, Lee-Cheun Loh M, Loghavi S, Mullighan CG, Ogawa S, Orazi A, Papaemmanuil E, Reiter A, Ross DM, Savona M, Shimamura A, Skoda RC, Solé F, Stone RM, Tefferi A, Walter MJ, Wu D, Ebert BL, Cazzola M. Genomic profiling for clinical decision making in myeloid neoplasms and acute leukemia. Blood 2022; 140:2228-2247. [PMID: 36130297 PMCID: PMC10488320 DOI: 10.1182/blood.2022015853] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/27/2022] [Indexed: 11/20/2022] Open
Abstract
Myeloid neoplasms and acute leukemias derive from the clonal expansion of hematopoietic cells driven by somatic gene mutations. Although assessment of morphology plays a crucial role in the diagnostic evaluation of patients with these malignancies, genomic characterization has become increasingly important for accurate diagnosis, risk assessment, and therapeutic decision making. Conventional cytogenetics, a comprehensive and unbiased method for assessing chromosomal abnormalities, has been the mainstay of genomic testing over the past several decades and remains relevant today. However, more recent advances in sequencing technology have increased our ability to detect somatic mutations through the use of targeted gene panels, whole-exome sequencing, whole-genome sequencing, and whole-transcriptome sequencing or RNA sequencing. In patients with myeloid neoplasms, whole-genome sequencing represents a potential replacement for both conventional cytogenetic and sequencing approaches, providing rapid and accurate comprehensive genomic profiling. DNA sequencing methods are used not only for detecting somatically acquired gene mutations but also for identifying germline gene mutations associated with inherited predisposition to hematologic neoplasms. The 2022 International Consensus Classification of myeloid neoplasms and acute leukemias makes extensive use of genomic data. The aim of this report is to help physicians and laboratorians implement genomic testing for diagnosis, risk stratification, and clinical decision making and illustrates the potential of genomic profiling for enabling personalized medicine in patients with hematologic neoplasms.
Collapse
Affiliation(s)
- Eric J. Duncavage
- Department of Pathology and Immunology, Washington University, St. Louis, MO
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Courtney D. DiNardo
- Division of Cancer Medicine, Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Lucy A. Godley
- Section of Hematology and Oncology, Departments of Medicine and Human Genetics, The University of Chicago, Chicago, IL
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia & Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Alessandro M. Vannucchi
- Department of Hematology, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence and Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Keyur P. Patel
- Division of Pathology/Lab Medicine, Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Maria E. Arcila
- Department of Pathology, Memorial Sloan Lettering Cancer Center, New York, NY
| | - Rafael Bejar
- Division of Hematology and Oncology, University of California San Diego, La Jolla, CA
| | - Nancy Berliner
- Division of Hematology, Brigham and Women’s Hospital, Harvard University, Boston, MA
| | - Michael J. Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Susan Branford
- Department of Genetics and Molecular Pathology, Center for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Anna L. Brown
- Department of Pathology, South Australia Heath Alliance, Adelaide, Australia
| | - Catherine A. Cargo
- Haematological Malignancy Diagnostic Service, St James’s University Hospital, Leeds, United Kingdom
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Brunangelo Falini
- Department of Hematology, CREO, University of Perugia, Perugia, Italy
| | | | | | - Eva Hellström-Lindberg
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annette S. Kim
- Department of Pathology, Brigham and Women’s Hospital, Harvard University, Boston, MA
| | - Jeffery M. Klco
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Rami Komrokji
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Mignon Lee-Cheun Loh
- Department of Pediatrics, Ben Towne Center for Childhood Cancer Research, Seattle Children’s Hospital, University of Washington, Seattle, WA
| | - Sanam Loghavi
- Division of Pathology/Lab Medicine, Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Seishi Ogawa
- University of Kyoto School of Medicine, Kyoto, Japan
| | - Attilio Orazi
- Department of Pathology, Texas Tech University Health Sciences Center, El Paso, TX
| | | | - Andreas Reiter
- University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - David M. Ross
- Haematology Directorate, SA Pathology, Adelaide, Australia
| | - Michael Savona
- Department of Medicine, Vanderbilt University, Nashville, TN
| | - Akiko Shimamura
- Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Radek C. Skoda
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Francesc Solé
- MDS Group, Institut de Recerca contra la Leucèmia Josep Carreras, Barcelona, Spain
| | - Richard M. Stone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | | | | - David Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mario Cazzola
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| |
Collapse
|
31
|
Moyo TK, Mendler JH, Itzykson R, Kishtagari A, Solary E, Seegmiller AC, Gerds AT, Ayers GD, Dezern AE, Nazha A, Valent P, van de Loosdrecht AA, Onida F, Pleyer L, Cirici BX, Tibes R, Geissler K, Komrokji RS, Zhang J, Germing U, Steensma DP, Wiseman DH, Pfeilstöecker M, Elena C, Cross NCP, Kiladjian JJ, Luebbert M, Mesa RA, Montalban-Bravo G, Sanz GF, Platzbecker U, Patnaik MM, Padron E, Santini V, Fenaux P, Savona MR. The ABNL-MARRO 001 study: a phase 1-2 study of randomly allocated active myeloid target compound combinations in MDS/MPN overlap syndromes. BMC Cancer 2022; 22:1013. [PMID: 36153475 PMCID: PMC9509596 DOI: 10.1186/s12885-022-10073-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) comprise several rare hematologic malignancies with shared concomitant dysplastic and proliferative clinicopathologic features of bone marrow failure and propensity of acute leukemic transformation, and have significant impact on patient quality of life. The only approved disease-modifying therapies for any of the MDS/MPN are DNA methyltransferase inhibitors (DNMTi) for patients with dysplastic CMML, and still, outcomes are generally poor, making this an important area of unmet clinical need. Due to both the rarity and the heterogeneous nature of MDS/MPN, they have been challenging to study in dedicated prospective studies. Thus, refining first-line treatment strategies has been difficult, and optimal salvage treatments following DNMTi failure have also not been rigorously studied. ABNL-MARRO (A Basket study of Novel therapy for untreated MDS/MPN and Relapsed/Refractory Overlap Syndromes) is an international cooperation that leverages the expertise of the MDS/MPN International Working Group (IWG) and provides the framework for collaborative studies to advance treatment of MDS/MPN and to explore clinical and pathologic markers of disease severity, prognosis, and treatment response. METHODS ABNL MARRO 001 (AM-001) is an open label, randomly allocated phase 1/2 study that will test novel treatment combinations in MDS/MPNs, beginning with the novel targeted agent itacitinib, a selective JAK1 inhibitor, combined with ASTX727, a fixed dose oral combination of the DNMTi decitabine and the cytidine deaminase inhibitor cedazuridine to improve decitabine bioavailability. DISCUSSION Beyond the primary objectives of the study to evaluate the safety and efficacy of novel treatment combinations in MDS/MPN, the study will (i) Establish the ABNL MARRO infrastructure for future prospective studies, (ii) Forge innovative scientific research that will improve our understanding of pathogenetic mechanisms of disease, and (iii) Inform the clinical application of diagnostic criteria, risk stratification and prognostication tools, as well as response assessments in this heterogeneous patient population. TRIAL REGISTRATION This trial was registered with ClinicalTrials.gov on August 19, 2019 (Registration No. NCT04061421).
Collapse
Affiliation(s)
- Tamara K Moyo
- Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN, 777 PRB, USA
- Levine Cancer Institute, Charlotte, NC, USA
| | - Jason H Mendler
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Ashwin Kishtagari
- Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN, 777 PRB, USA
| | - Eric Solary
- Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Adam C Seegmiller
- Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN, 777 PRB, USA
| | | | - Gregory D Ayers
- Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN, 777 PRB, USA
| | | | | | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Francesco Onida
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Lisa Pleyer
- Third Medical Department With Hematology, Medical Oncology, Rheumatology and Infectiology, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
| | - Blanca Xicoy Cirici
- Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Bellaterr, Spain
| | | | | | | | - Jing Zhang
- University of Wisconsin-Madison, Madison, WI, USA
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, University of Duesseldorf, Duesseldorf, Germany
| | | | | | - Michael Pfeilstöecker
- Hanusch Hospital and Ludwig Boltzmann Institute for Hematology and Oncology, Vienna, Austria
| | | | | | - Jean-Jacques Kiladjian
- Université de Paris, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | | | - Ruben A Mesa
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | | | | | | | | | - Eric Padron
- H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | | | - Michael R Savona
- Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN, 777 PRB, USA.
| |
Collapse
|
32
|
Hergott CB, Kim AS. Molecular Diagnostic Testing for Hematopoietic Neoplasms: Linking Pathogenic Drivers to Personalized Diagnosis. Clin Lab Med 2022; 42:325-347. [PMID: 36150815 DOI: 10.1016/j.cll.2022.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Molecular diagnostics inhabit an increasingly central role in characterizing hematopoietic malignancies. This brief review summarizes the genomic targets important for many major categories of hematopoietic neoplasia by focusing on disease pathogenesis. In myeloid disease, recurrent mutations in key functional classes drive clonal hematopoiesis, on which additional variants can specify clinical presentation and accelerate progression. Lymphoblastic leukemias are frequently initiated by oncogenic fusions that block lymphoid maturation while, in concert with additional mutations, driving proliferation. The links between genetic aberrations and lymphoma patient outcomes have been clarified substantially through the clustering of genomic profiles. Finally, the addition of next-generation sequencing strategies to cytogenetics is refining risk stratification for plasma cell myeloma. In all categories, molecular diagnostics shed light on the unique mechanistic underpinnings of each individual malignancy, thereby empowering more rational, personalized care for these patients.
Collapse
Affiliation(s)
- Christopher B Hergott
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Castaño-Díez S, López-Guerra M, Bosch-Castañeda C, Bataller A, Charry P, Esteban D, Guijarro F, Jiménez-Vicente C, Castillo-Girón C, Cortes A, Martínez-Roca A, Triguero A, Álamo JR, Beà S, Costa D, Colomer D, Rozman M, Esteve J, Díaz-Beyá M. Real-World Data on Chronic Myelomonocytic Leukemia: Clinical and Molecular Characteristics, Treatment, Emerging Drugs, and Patient Outcomes. Cancers (Basel) 2022; 14:cancers14174107. [PMID: 36077644 PMCID: PMC9455040 DOI: 10.3390/cancers14174107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Despite emerging molecular information on chronic myelomonocytic leukemia (CMML), patient outcome remains unsatisfactory and little is known about the transformation to acute myeloid leukemia (AML). In a single-center cohort of 219 CMML patients, we explored the potential correlation between clinical features, gene mutations, and treatment regimens with overall survival (OS) and clonal evolution into AML. The most commonly detected mutations were TET2, SRSF2, ASXL1, and RUNX1. Median OS was 34 months and varied according to age, cytogenetic risk, FAB, CPSS and CPSS-Mol categories, and number of gene mutations. Hypomethylating agents were administered to 37 patients, 18 of whom responded. Allogeneic stem cell transplantation (alloSCT) was performed in 22 patients. Two-year OS after alloSCT was 60.6%. Six patients received targeted therapy with IDH or FLT3 inhibitors, three of whom attained a long-lasting response. AML transformation occurred in 53 patients and the analysis of paired samples showed changes in gene mutation status. Our real-world data emphasize that the outcome of CMML patients is still unsatisfactory and alloSCT remains the only potentially curative treatment. However, targeted therapies show promise in patients with specific gene mutations. Complete molecular characterization can help to improve risk stratification, understand transformation, and personalize therapy.
Collapse
Affiliation(s)
- Sandra Castaño-Díez
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Medical School, University of Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Mónica López-Guerra
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | | | - Alex Bataller
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Medical School, University of Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
| | - Paola Charry
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Daniel Esteban
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Francesca Guijarro
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Medical School, University of Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Jiménez-Vicente
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Carlos Castillo-Girón
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Albert Cortes
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Hematology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Alexandra Martínez-Roca
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Ana Triguero
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - José Ramón Álamo
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Silvia Beà
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Dolors Costa
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Dolors Colomer
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - María Rozman
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Esteve
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- Medical School, University of Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
| | - Marina Díaz-Beyá
- Hematology and Hematopathology Departments, Hospital Clínic Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
- Correspondence: ; Tel.: +34-9-227-54-28
| |
Collapse
|
34
|
Bi X, Gergis U, Wagner JL, Carabasi M, Filicko-O’Hara J, O’Hara W, Klumpp T, Porcu P, Flomenberg N, Grosso D. Outcomes of two-step haploidentical allogeneic stem cell transplantation in elderly patients with hematologic malignancies. Bone Marrow Transplant 2022; 57:1671-1680. [PMID: 35986105 PMCID: PMC9388981 DOI: 10.1038/s41409-022-01780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) remains the best curative option for the majority of patients with hematologic malignancies (HM); however, many elderly patients are excluded from transplant and outcome data in this population is still limited. The novel two-step graft engineering approach has been the main platform for allo-SCT at Thomas Jefferson University since 2006. Following administration of the preparative regimen, we infuse donor lymphocytes, followed by cyclophosphamide to induce bidirectional tolerance, then infusion of CD34-selected cells. A total of 76 patients ≥ 65 years old with HM underwent haploidentical (haplo) allo-SCT on the two-step transplant platform between 2007 and 2021. The median time to neutrophil engraftment was 11 days and platelet engraftment was 18 days. With a median follow up of 44 months, the 3-year overall survival (OS) and progression-free survival (PFS) were 36.3% and 35.6%, respectively. The cumulative incidences of non-relapse mortality (NRM) and relapse at 3 years were 43.5% and 21.0% at 3 years, respectively. The cumulative incidence of grade III-IV acute graft-versus-host-disease (GVHD) was 11.1% at 6 months, and chronic GVHD requiring treatment was 15.1% at 2 years. The two-step haplo allo-SCT is a novel alternative platform for high-risk older HM patients, achieving fast engraftment, low relapse rates and promising survival.
Collapse
|
35
|
Karantanos T, Tsai HL, Gondek LP, DeZern AE, Ghiaur G, Dalton WB, Gojo I, Prince GT, Webster J, Ambinder A, Smith BD, Levis MJ, Varadhan R, Jones RJ, Jain T. Genomic landscape of myelodysplastic/myeloproliferative neoplasm can predict response to hypomethylating agent therapy. Leuk Lymphoma 2022; 63:1942-1948. [PMID: 35379077 PMCID: PMC9847567 DOI: 10.1080/10428194.2022.2057488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
There are currently no known predictors of myelodysplastic syndrome (MDS)/myeloproliferative overlap neoplasm (MPN) patients' response to hypomethylating agents (HMA). Forty-three patients with MDS/MPN who were treated with HMA during chronic phase and had next-generation sequencing using the established 63-genes panel were identified. Complete and partial remission and marrow response were assessed based on the MDS/MPN International Working Group response criteria. On univariate analysis, younger age, higher number of mutations, and mutations in SETBP1, RUNX1, or EZH2 were associated with no response. Multivariable analysis for modeling response were conducted via least absolute shrinkage and selection operator logistic regression approach, and showed that mutations in SETBP1, RUNX1, or EZH2 predict lack of HMA response. While limited by sample size, our findings suggest that genomic landscape can potentially identify MDS/MPN patients with lower likelihood of response to HMA.
Collapse
Affiliation(s)
- Theodoras Karantanos
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Hua-Ling Tsai
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Lukasz P. Gondek
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Amy E. DeZern
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Gabriel Ghiaur
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - W. Brian Dalton
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Ivana Gojo
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielis T. Prince
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan Webster
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Alexander Ambinder
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - B. Douglas Smith
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Mark J Levis
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Richard J. Jones
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimrnel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
36
|
Hagino T, Sato T, Saga R, Hidai H, Murai Y, Akiyama H, Motomura S. Myeloid leukemoid reaction after initial azacitidine therapy for chronic myelomonocytic leukemia. Int J Hematol 2022; 116:961-965. [DOI: 10.1007/s12185-022-03422-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022]
|
37
|
Nathan DI, Feld J, El Jamal SM, Mascarenhas J, Tremblay D. Myelodysplastic syndrome/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis: Ringing in a new future. Leuk Res 2022; 115:106820. [DOI: 10.1016/j.leukres.2022.106820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/19/2023]
|
38
|
Enjeti AK, Agarwal R, Blombery P, Chee L, Chua CC, Grigg A, Hamad N, Iland H, Lane S, Perkins A, Singhal D, Tate C, Tiong IS, Ross DM. Panel-based gene testing in myelodysplastic/myeloproliferative neoplasm- overlap syndromes: Australasian Leukaemia and Lymphoma Group (ALLG) consensus statement. Pathology 2022; 54:389-398. [DOI: 10.1016/j.pathol.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
|
39
|
Walsh C, Hunter A, Lasho T, Finke C, Ketterling R, Komrokji R, Tefferi A, Mangaonkar A, Howard M, Gangat N, Al-Kali A, Ali NA, Padron E, Patnaik MM. Differential prognostic impact of IDH1 and IDH2 mutations in chronic myelomonocytic leukemia. Leukemia 2022; 36:1693-1696. [DOI: 10.1038/s41375-022-01551-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/09/2022]
|
40
|
Patnaik MM, Tefferi A. Chronic myelomonocytic leukemia: 2022 update on diagnosis, risk stratification, and management. Am J Hematol 2022; 97:352-372. [PMID: 34985762 DOI: 10.1002/ajh.26455] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022]
Abstract
DISEASE OVERVIEW Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms, with an inherent risk for leukemic transformation (~15% over 3-5 years). DIAGNOSIS Diagnosis is based on the presence of sustained (>3 months) peripheral blood monocytosis (≥1 × 109 /L; monocytes ≥10%), usually with accompanying bone marrow dysplasia. Clonal cytogenetic abnormalities occur in ~30% of patients, while >90% have somatic gene mutations. Mutations involving TET2 (~60%), SRSF2 (~50%), ASXL1 (~40%), and the oncogenic RAS pathway (~30%) are frequent, while the presence of ASXL1 and DNMT3A mutations and the absence of TET2 mutations negatively impact overall survival. RISK-STRATIFICATION Molecularly integrated prognostic models include the Groupe Français des Myélodysplasies, Mayo Molecular Model (MMM), and the CMML specific prognostic model. Risk factors incorporated into the MMM include presence of truncating ASXL1 mutations, absolute monocyte count >10 × 109 /L, hemoglobin <10 g/dL, platelet count <100 × 109 /L, and the presence of circulating immature myeloid cells. The MMM stratifies CMML patients into four groups: high (≥3 risk factors), intermediate-2 (2 risk factors), intermediate-1 (1 risk factor), and low (no risk factors), with median survivals of 16, 31, 59, and 97 months, respectively. RISK-ADAPTED THERAPY Hypomethylating agents such as 5-azacitidine and decitabine are commonly used, with overall response rates of ~40%-50% and complete remission rates of ~7%-17%; with no impact on mutational allele burdens. Allogeneic stem cell transplant is the only potentially curative option but is associated with significant morbidity and mortality.
Collapse
Affiliation(s)
- Mrinal M. Patnaik
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota USA
| | - Ayalew Tefferi
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota USA
| |
Collapse
|
41
|
Triguero A, Xicoy B, Zamora L, Jiménez MJ, García O, Calabuig M, Díaz-Beyá M, Arzuaga J, Ramos F, Medina A, Bernal T, Talarn C, Coll R, Collado R, Chen TH, Borrás J, Brunet S, Marchante I, Marco V, López F, Calbacho M, Simiele A, Cortés M, Cedena MT, Pedreño M, Aguilar C, Pedró C, Fernández M, Stoica C, Ribera JM, Sanz G. RESPONSE TO AZACITIDINE IN PATIENTS WITH CHRONIC MYELOMONOCYTIC LEUKEMIA ACCORDING TO OVERLAP MYELODYSPLASTIC/MYELOPROLIFERATIVE NEOPLASMS CRITERIA. Leuk Res 2022; 116:106836. [DOI: 10.1016/j.leukres.2022.106836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
42
|
Emergence of clone with PHF6 nonsense mutation in chronic myelomonocytic leukemia at relapse after allogeneic HCT. Int J Hematol 2022; 115:748-752. [PMID: 34988909 DOI: 10.1007/s12185-021-03284-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 10/19/2022]
Abstract
Disease relapse is a major cause of treatment failure after allogeneic hematopoietic cell transplantation (HCT) and the mechanisms of relapse remain unclear. We encountered a 58-year-old man with chronic myelomonocytic leukemia (CMML) that relapsed after haploidentical HCT from his daughter. Peripheral blood samples collected at HCT and at relapse were analyzed, and CD14+/CD16- monocytes that typically accumulate in CMML were isolated by flow cytometry. Whole-exome sequencing of the monocytes revealed 8 common mutations in CMML at HCT. In addition, a PHF6 nonsense mutation not detected at HCT was detected at relapse. RNA sequencing could not detect changes in expression of HLA or immune-checkpoint molecules, which are important mechanisms of immune evasion. However, gene set enrichment analysis (GSEA) revealed that a TNF-α signaling pathway was downregulated at relapse. Ubiquitination of histone H2B at lysine residue 120 (H2BK120ub) at relapse was significantly decreased at the protein level, indicating that PHF6 loss might downregulate a TNF-α signaling pathway by reduction of H2BK120ub. This case illustrates that PHF6 loss contributes to a competitive advantage for the clone under stress conditions and leads to relapse after HCT.
Collapse
|
43
|
Mazza GL, Mead-Harvey C, Mascarenhas J, Yacoub A, Kosiorek HE, Hoffman R, Dueck AC, Mesa RA. Symptom burden and quality of life in patients with high-risk essential thrombocythaemia and polycythaemia vera receiving hydroxyurea or pegylated interferon alfa-2a: a post-hoc analysis of the MPN-RC 111 and 112 trials. Lancet Haematol 2022; 9:e38-e48. [PMID: 34971581 PMCID: PMC9098160 DOI: 10.1016/s2352-3026(21)00343-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Patients with essential thrombocythaemia or polycythaemia vera have several symptoms that can worsen their quality of life. We aimed to assess how symptom burden changes over time with cytoreductive therapy. METHODS We performed a post-hoc analysis of data from MPN-RC 111-a single-arm, open-label, phase 2, multicentre trial at 17 hospitals and cancer centres in Italy and the USA, evaluating the clinical-haematological response to pegylated interferon alfa-2a in patients who were resistant or intolerant to hydroxyurea (NCT01259817)-and MPN-RC 112-a randomised, open-label, phase 3, multicentre trial at 25 hospitals and cancer centres in France, Germany, Israel, Italy, the UK, and the USA, comparing the clinical-haematological response to pegylated interferon alfa-2a versus hydroxyurea in therapy-naive patients with either high-risk essential thrombocythaemia or polycythaemia vera (NCT01258856). Patients completed the Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF) and the European Organisation for the Research and Treatment of Cancer Core Quality of Life Questionnaire through 12 months after initiation of treatment as secondary endpoints. In this post-hoc analysis, we examined the association of symptom burden with the clinical-haematological response at 12 months and the effect of baseline symptom burden (ie, high burden [total symptom score ≥20] vs low burden [total symptom score <20]) on subsequent changes in symptoms, estimated via mixed models. A clinically significant improvement in symptom burden was defined as 50% or greater improvement in the MPN-SAF total symptom score from baseline to 12 months in patients with a total symptom score greater than zero at baseline. FINDINGS 135 patients were enrolled in MPN-RC 111 between Feb 15, 2012, and Dec 23, 2015, and 168 were enrolled in MPN-RC 112 between Sept 24, 2011, and June 30, 2016. For this analysis, we included data from 114 patients from MPN-RC 111 (64 [56%] with essential thrombocythaemia and 50 [44%] with polycythaemia vera; 56 [49%] were female, and 100 [91%] of 110 were white) and 166 patients from MPN-RC 112 (79 [48%] with essential thrombocythaemia and 87 [52%] with polycythaemia vera; 68 [41%] were female, and 145 [93%] of 156 were white). At 12 months, a clinically significant improvement in symptom burden was reported by 12 (32%) of 38 complete responders and seven (20%) of 35 partial responders treated with pegylated interferon alfa-2a in MPN-RC 111; five (19%) of 27 complete responders and six (18%) of 34 partial responders treated with pegylated interferon alfa-2a in MPN-112; and eight (27%) of 30 complete responders and six (22%) of 27 partial responders treated with hydroxyurea in MPN-112. More complete and partial responders reported a clinically significant improvement than did non-responders (44 [22%] of 191 complete and partial responders vs four [5%] of 76 non-responders; Fisher's exact p=0·0003). Symptom burden improved between 3 and 12 months in patients with high baseline symptom burden, both those treated with pegylated interferon alfa-2a (mean total symptom score change -10·2, 95% CI -13·2 to -7·2) and those treated with hydroxyurea (-6·8, -11·2 to -2·4). However, symptom burden worsened between 3 and 12 months in patients with low baseline symptom burden (patients treated with pegylated interferon alfa-2a: mean total symptom score change 3·2, 95% CI 0·9 to 5·4; patients treated with hydroxyurea: 3·4, 0·6 to 6·2). INTERPRETATION Results can inform treatment decisions, including treatment timing and goals in managing essential thrombocythaemia and polycythaemia vera, because measuring symptom burden from the patient perspective is crucial to understanding treatment efficacy and tolerability. FUNDING US National Cancer Institute of the National Institutes of Health, and Roche Genentech.
Collapse
Affiliation(s)
- Gina L. Mazza
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | | | - John Mascarenhas
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Abdulraheem Yacoub
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS
| | - Heidi E. Kosiorek
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | - Ronald Hoffman
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Amylou C. Dueck
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | - Ruben A. Mesa
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX
| |
Collapse
|
44
|
Garcia JS, Kim HT, Murdock HM, Cutler CS, Brock J, Gooptu M, Ho VT, Koreth J, Nikiforow S, Romee R, Shapiro R, Loschi F, Ryan J, Fell G, Karp HQ, Lucas F, Kim AS, Potter D, Mashaka T, Stone RM, DeAngelo DJ, Letai A, Lindsley RC, Soiffer RJ, Antin JH. Adding venetoclax to fludarabine/busulfan RIC transplant for high-risk MDS and AML is feasible, safe, and active. Blood Adv 2021; 5:5536-5545. [PMID: 34614506 PMCID: PMC8714724 DOI: 10.1182/bloodadvances.2021005566] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/20/2021] [Indexed: 01/03/2023] Open
Abstract
Adding the selective BCL-2 inhibitor venetoclax to reduced-intensity conditioning chemotherapy (fludarabine and busulfan [FluBu2]) may enhance antileukemic cytotoxicity and thereby reduce the risk of posttransplant relapse. This phase 1 study investigated the recommended phase 2 dose (RP2D) of venetoclax, a BCL-2 selective inhibitor, when added to FluBu2 in adult patients with high-risk acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and MDS/myeloproliferative neoplasms (MPN) undergoing transplant. Patients received dose-escalated venetoclax (200-400 mg daily starting day -8 for 6-7 doses) in combination with fludarabine 30 mg/m2 per day for 4 doses and busulfan 0.8 mg/kg twice daily for 8 doses on day -5 to day -2 (FluBu2). Transplant related-toxicity was evaluated from the first venetoclax dose on day -8 to day 28. Twenty-two patients were treated. At study entry, 5 patients with MDS and MDS/MPN had 5% to 10% marrow blasts, and 18 (82%) of 22 had a persistent detectable mutation. Grade 3 adverse events included mucositis, diarrhea, and liver transaminitis (n = 3 each). Neutrophil/platelet recovery and acute/chronic graft-versus-host-disease rates were similar to those of standard FluBu2. No dose-limiting toxicities were observed. The RP2D of venetoclax was 400 mg daily for 7 doses. With a median follow-up of 14.7 months (range, 8.6-24.8 months), median overall survival was not reached, and progression-free survival was 12.2 months (95% confidence interval, 6.0-not estimable). In patients with high-risk AML, MDS, and MDS/MPN, adding venetoclax to FluBu2 was feasible and safe. To further address relapse risk, assessment of maintenance therapy after venetoclax plus FluBu2 transplant is ongoing. This study was registered at clinicaltrials.gov as #NCT03613532.
Collapse
Affiliation(s)
| | - Haesook T. Kim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA; and
| | | | | | | | | | | | | | | | | | | | | | | | - Geoffrey Fell
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA; and
| | | | - Fabienne Lucas
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Annette S. Kim
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Saliba AN, Litzow MR, Gangat N, Al‐Kali A, Foran JM, Hogan WJ, Palmer JM, Mangaonkar AA, Tefferi A, Patnaik MM. Outcomes of venetoclax-based therapy in chronic phase and blast transformed chronic myelomonocytic leukemia. Am J Hematol 2021; 96:E433-E436. [PMID: 34428328 DOI: 10.1002/ajh.26334] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Affiliation(s)
| | - Mark R. Litzow
- Division of Hematology Mayo Clinic Rochester Minnesota USA
| | - Naseema Gangat
- Division of Hematology Mayo Clinic Rochester Minnesota USA
| | - Aref Al‐Kali
- Division of Hematology Mayo Clinic Rochester Minnesota USA
| | - James M. Foran
- Division of Hematology and Oncology Mayo Clinic Jacksonville Florida USA
| | | | - Jeanne M. Palmer
- Division of Hematology and Oncology Mayo Clinic Phoenix Arizona USA
| | | | - Ayalew Tefferi
- Division of Hematology Mayo Clinic Rochester Minnesota USA
| | | |
Collapse
|
46
|
Kazmi F, Shrestha N, Booth S, Dodwell D, Aroldi F, Foord T, Nicholson BD, Heesen P, Lord S, Yeoh KW, Blagden S. Next-generation sequencing for guiding matched targeted therapies in people with relapsed or metastatic cancer. Hippokratia 2021. [DOI: 10.1002/14651858.cd014872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Farasat Kazmi
- Department of Oncology; University of Oxford; Oxford UK
| | - Nipun Shrestha
- Department of Primary Care and Mental Health; University of Liverpool; Liverpool UK
| | - Stephen Booth
- Department of Oncology; University of Oxford; Oxford UK
| | - David Dodwell
- Nuffield Department of Population Health; University of Oxford; Oxford UK
| | | | | | - Brian D Nicholson
- Nuffield Department of Primary Care Health Sciences; University of Oxford; Oxford UK
| | | | - Simon Lord
- Department of Oncology; University of Oxford; Oxford UK
| | - Kheng-Wei Yeoh
- Radiation Oncology; National Cancer Centre; Singapore Singapore
| | - Sarah Blagden
- Department of Oncology; University of Oxford; Oxford UK
| |
Collapse
|
47
|
Renneville A, Patnaik MM, Chan O, Padron E, Solary E. Increasing recognition and emerging therapies argue for dedicated clinical trials in chronic myelomonocytic leukemia. Leukemia 2021; 35:2739-2751. [PMID: 34175902 DOI: 10.1038/s41375-021-01330-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023]
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN). Median overall survival of this aggressive myeloid malignancy is only 2-3 years, with a 15-30% risk of acute leukemic transformation. The paucity of clinical trials specifically designed for CMML has made therapeutic management of CMML patients challenging. As a result, treatment paradigms for CMML patients are largely borrowed from MDS and MPN. The standard of care still relies on hydroxyurea, hypomethylating agents (HMA), and allogeneic stem cell transplantation, this latter option remaining the only potentially curative therapy. To date, approved drugs for CMML treatment are HMA, including azacitidine, decitabine, and more recently the oral combination of decitabine and cedazuridine. However, HMA treatment does not meaningfully alter the natural course of this disease. New treatment approaches for improving CMML-associated cytopenias or targeting the CMML malignant clone are emerging. More than 25 therapeutic agents are currently being evaluated in phase 1 or phase 2 clinical trials for CMML and other myeloid malignancies, often in combination with a HMA backbone. Several novel agents, such as sotatercept, ruxolitinib, lenzilumab, and tagraxofusp have shown promising clinical efficacy in CMML. Current evidence supports the idea that effective treatment in CMML will likely require combination therapy targeting multiple pathways, which emphasizes the need for additional new therapeutic options. This review focuses on recent therapeutic advances and innovative treatment strategies in CMML, including global and molecularly targeted approaches. We also discuss what may help to make progress in the design of rationally derived and disease-modifying therapies for CMML.
Collapse
Affiliation(s)
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Onyee Chan
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Eric Solary
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France. .,Faculty of Medicine, Université Paris-Sud, Le Kremlin-Bicêtre, France. .,Department of Hematology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
48
|
Hammond D, Montalban-Bravo G. Management and Outcomes of Blast Transformed Chronic Myelomonocytic Leukemia. Curr Hematol Malig Rep 2021; 16:405-417. [PMID: 34499330 DOI: 10.1007/s11899-021-00643-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Despite recent advances in the treatment of de novo acute myeloid leukemia (AML), AML arising from antecedent chronic myelomonocytic leukemia (CMML) continues to have dismal outcomes. While the unique biological drivers of CMML and subsequent leukemic transformation (LT) have been revealed with advances in molecular characterization, this has not yet translated to the bedside. Here, we review these biologic drivers, outcomes with current therapies, and rationale avenues of future investigation specifically in blast phase CMML (CMML-BP). RECENT FINDINGS CMML-BP outcomes are studied as an aggregate with more common categories of AML with myelodysplasia-related changes (AML-MRCs) or the even broader category of secondary AML (sAML), which illustrates the crux of the problem. While a modest survival advantage with allogeneic hematopoietic stem cell transplant exists, the difficulty is bridging patients to transplant and managing patients that require an allograft-sparing approach. Limited data suggest that short-lived remissions can be obtained employing CPX-351 or venetoclax-based lower intensity combination therapy. Promising future strategies include repurposing cladribine, exploiting the supportive role of dendritic cell subsets with anti-CD123 therapies, MCL-1 inhibition, dual MEK/PLK1 inhibition, FLT3 inhibition in RAS-mutated and CBL-mutated subsets, and immune therapies targeting novel immune checkpoint molecules such as the leukocyte immunoglobulin-like receptor B4 (LILRB4), an immune-modulatory transmembrane protein restrictively expressed on monocytic cells. The successful management of an entity as unique as CMML-BP will require a cooperative, concerted effort to design and conduct clinical trials dedicated to this rare form of sAML.
Collapse
Affiliation(s)
- Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
49
|
Rozema J, van Roon EN, Kibbelaar RE, Veeger NJGM, Slim CL, de Wit H, Hoogendoorn M. Patterns of transfusion burden in an unselected population of patients with myelodysplastic syndromes: A population-based study. Transfusion 2021; 61:2877-2884. [PMID: 34480360 PMCID: PMC9293228 DOI: 10.1111/trf.16631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/27/2022]
Abstract
Background Ineffective hematopoiesis in patients with myelodysplastic syndromes (MDS) often results in transfusion dependence. The burden of frequent transfusions in the real‐world MDS population is largely unknown. Study design and methods An observational, retrospective, population‐based study, using the HemoBase registry, was performed including all patients diagnosed with MDS between 2005 and 2017 in Friesland, a province in the Netherlands with approximately 650,000 inhabitants. Detailed clinical information was collected from the electronic health records. Transfusion burden was classified according to the International Working Group 2018 criteria: not transfusion dependent, low (LTB), or high transfusion burden (HTB). Univariate and multivariable regression analyses were performed. Results Of 292 patients, 136 (46.6%) had a HTB of ≥8 units/16 weeks and 17 (5.8%) had a LTB of 3–7 units/16 weeks. This was present in all types of MDS patients, but patients aged 75–84 years (odds ratio [OR] 4.02, 95% confidence interval [CI]: 1.84–8.82), high‐risk MDS patients (OR 2.88, 95% CI: 1.08–7.68) and MDS‐EB‐2 patients (OR 7.07, 95% CI: 2.17–22.90) were particularly at risk for a HTB. Discussion This study provides a reliable estimate of the transfusion burden in real‐world MDS patients, with almost half of the patients having a HTB. A HTB was observed in all MDS subtypes and both low‐ and high‐risk MDS. Therefore, we conclude that the entire MDS population might benefit from novel agents that reduce the transfusion need and that might have beneficial effects on patient outcomes and healthcare utilization outcomes.
Collapse
Affiliation(s)
- Johanne Rozema
- Unit of Pharmacotherapy, Epidemiology and Economics, Department of Pharmacy, University of Groningen, Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Eric N van Roon
- Unit of Pharmacotherapy, Epidemiology and Economics, Department of Pharmacy, University of Groningen, Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Robby E Kibbelaar
- Department of Pathology, Pathology Friesland, Leeuwarden, The Netherlands
| | - Nic J G M Veeger
- Science Bureau Department, Science Bureau, Medical Center Leeuwarden, Leeuwarden, The Netherlands.,Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Christiaan L Slim
- Location Medical Center Leeuwarden, Certe Medical Diagnostics & Advice, Leeuwarden, The Netherlands
| | - Harry de Wit
- Location Medical Center Leeuwarden, Certe Medical Diagnostics & Advice, Leeuwarden, The Netherlands
| | - Mels Hoogendoorn
- Department of Internal Medicine, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | | |
Collapse
|
50
|
Sadigh S, Kim AS. Molecular Pathology of Myeloid Neoplasms: Molecular Pattern Recognition. Surg Pathol Clin 2021; 14:517-528. [PMID: 34373100 DOI: 10.1016/j.path.2021.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Despite the apparent complexity of the molecular genetic underpinnings of myeloid neoplasms, most myeloid mutational profiles can be understood within a simple framework. Somatic mutations accumulate in hematopoietic stem cells with aging and toxic insults, termed clonal hematopoiesis. These "old stem cells" mutations, predominantly in the epigenetic and RNA spliceosome pathways, act as "founding" driver mutations leading to a clonal myeloid neoplasm when sufficient in number and clone size. Subsequent mutations can create the genetic flavor of the myeloid neoplasm ("backseat" drivers) due to their enrichment in certain entities or act as progression events ("aggressive" drivers) during clonal evolution.
Collapse
Affiliation(s)
- Sam Sadigh
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|