403 results on '"LUCA MALCOVATI"'
Search Results
52. Dynamics of Mortality and Transformation Risk within Different Risk Groups of Patients with Myelodysplastic Syndromes Stratified According to the IPSS-R - Comparison of Treated and Untreated Patients and Consequences for the Description of Risk Categories
- Author
-
Michael Pfeilstocker, Heinz Tuechler, Lionel Ades, Jaroslav Cermak, Fatiha Chermat, Matteo G. Della Porta, Pierre Fenaux, Guillermo Garcia-Manero, Ulrich Germing, Detlef Haase, Andrea Kuendgen, Michael Luebbert, Silvia Maria Meira Magalhaes, Luca Malcovati, Yasushi Miyazaki, Guillermo Sanz, Valeria Santini, Mikkael A. Sekeres, Matthew J Walter, Peter Valent, and Peter L Greenberg
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
53. Consensus proposal for revised International Working Group response criteria for higher risk myelodysplastic syndromes
- Author
-
Amer M. Zeidan, Uwe Platzbecker, Jan Philipp Bewersdorf, Maximilian Stahl, Lionel Adès, Uma Borate, David T Bowen, Rena J. Buckstein, Andrew M. Brunner, Hetty E Carraway, Naval G. Daver, Maria Díez-Campelo, Theo M de Witte, Amy E. DeZern, Fabio Efficace, Guillermo Garcia-Manero, Jacqueline S. Garcia, Ulrich Germing, Aristoteles Giagounidis, Elizabeth A Griffiths, Robert P Hasserjian, Eva Hellström-Lindberg, Marcelo C Iastrebner, Rami S. Komrokji, Austin G Kulasekararaj, Luca Malcovati, Yasushi Miyazaki, Olatoyosi Odenike, Valeria Santini, Guillermo F. Sanz, Phillip Scheinberg, Reinhard Stauder, Arjan A Van de Loosdrecht, Andrew H Wei, Mikkael A. Sekeres, and Pierre Fenaux
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Myelodysplastic syndromes/neoplasms (MDS) are associated with variable clinical presentations and outcomes. The initial response criteria developed by the International Working Group (IWG) in 2000 have been used in clinical practice, clinical trials, regulatory reviews, and drug labels. While the IWG criteria were revised in 2006 and 2018 (the latter focusing on lower-risk disease), limitations persist in their application to higher-risk MDS and in their ability to fully capture clinical benefits of novel investigational drugs or to serve as valid surrogates for longer-term clinical endpoints (e.g., overall survival). Further, issues related to ambiguity and practicality of some criteria lead to variability in interpretation and inter-observer inconsistency in reporting results from the same sets of data. Thus, we convened an international panel of 36 MDS experts and used an established modified Delphi process to develop consensus recommendations for updated response criteria that would be more reflective of patient-centered and clinically relevant outcomes in higher-risk MDS. Among others, the IWG 2023 criteria include changes in the hemoglobin threshold for complete remission (CR), the introduction of CR with limited count recovery (CRL) and CR with partial hematologic recovery (CRh) as provisional response criteria, elimination of marrow CR, and specific recommendations for standardization of time-to-event endpoints and the derivation and reporting of responses. The updated criteria should lead to better correlation between patient-centered outcomes and clinical trial results in an era of multiple emerging new agents with novel mechanisms of action.
- Published
- 2023
- Full Text
- View/download PDF
54. Cause of death and excess mortality in patients with lower-risk myelodysplastic syndromes (MDS): A report from the European MDS registry
- Author
-
Krzysztof, Mądry, Karol, Lis, Pierre, Fenaux, David, Bowen, Argiris, Symeonidis, Moshe, Mittelman, Reinhard, Stauder, Jaroslav, Čermák, Guillermo, Sanz, Eva, Hellström-Lindberg, Saskia, Langemeijer, Luca, Malcovati, Ulrich, Germing, Mette Skov, Holm, Agnes, Guerci-Bresler, Dominic, Culligan, Laurence, Sanhes, Ioannis, Kotsianidis, Corine, van Marrewijk, Simon, Crouch, Theo, de Witte, and Alex, Smith
- Subjects
Cancer development and immune defence Radboud Institute for Health Sciences [Radboudumc 2] ,All institutes and research themes of the Radboud University Medical Center ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Hematology - Abstract
Contains fulltext : 290857.pdf (Publisher’s version ) (Closed access) Information on causes of death (CoDs) and the impact of myelodysplastic syndromes (MDS) on survival in patients with lower-risk MDS (LR-MDS) is limited. A better understanding of the relationship between disease characteristics, clinical interventions and CoDs may improve outcomes of patients with LR-MDS. We prospectively collected data on patients with LR-MDS in the European MDS registry from 2008 to 2019. Clinical, laboratory and CoDs data were obtained. To examine MDS-specific survival, relative survival (RS) was estimated using national life tables. Of 2396 evaluated subjects, 900 died (median overall survival [OS]: 4.7 years; median follow-up: 3.5 years). The most common CoDs were acute myeloid leukaemia/MDS (20.1%), infection (17.8%) and cardiovascular disease (CVD; 9.8%). Patients with isolated del(5q) and with red cell transfusion needed during the disease course, had a higher risk of fatal CVD. The 5-year OS was 47.3% and the 5-year RS was 59.6%, indicating that most patients died due to their underlying MDS. Older patients (aged >80 years) and the lowest-risk patients were more likely to die from competing causes. This study shows that MDS and its related complications play crucial role in the outcome of patients with LR-MDS. 01 februari 2023
- Published
- 2023
- Full Text
- View/download PDF
55. Prognostic impact of organomegaly in mastocytosis : an analysis of the European Competence Network on Mastocytosis
- Author
-
Johannes Lübke, Juliana Schwaab, Deborah Christen, Hanneke Oude Elberink, Bart Span, Marek Niedoszytko, Aleksandra Gorska, Magdalena Lange, Karoline V. Gleixner, Emir Hadzijusufovic, Oleksii Solomianyi, Irena Angelova-Fischer, Roberta Zanotti, Massimiliano Bonifacio, Patrizia Bonadonna, Khalid Shoumariyeh, Nikolas von Bubnoff, Sabine Müller, Cecelia Perkins, Chiara Elena, Luca Malcovati, Hans Hagglund, Mattias Mattsson, Roberta Parente, Judit Varkonyi, Anna Belloni Fortina, Francesca Caroppo, Alexander Zink, Knut Brockow, Christine Breynaert, Dominique Bullens, Akif Selim Yavuz, Michael Doubek, Vito Sabato, Tanja Schug, Dietger Niederwieser, Karin Hartmann, Massimo Triggiani, Jason Gotlib, Olivier Hermine, Michel Arock, Hanneke C. Kluin-Nelemans, Jens Panse, Wolfgang R. Sperr, Peter Valent, Andreas Reiter, Mohamad Jawhar, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Hepatomegaly ,Lymphadenopathy ,Mastocytosis ,Organomegaly ,Splenomegaly ,Systemic mastocytosis ,Immunology and Allergy ,Human medicine - Abstract
BACKGROUND: Organomegaly, including splenomegaly, hepatomegaly, and/or lymphadenopathy, are important diagnostic and prognostic features in patients with cutaneous mastocytosis (CM) or systemic mastocytosis (SM). OBJECTIVES: To investigate the prevalence and prognostic impact of 1 or more organomegalies on clinical course and survival in patients with CM/SM. METHODS: Therefore, 3155 patients with CM (n = 1002 [32%]) or SM (n = 2153 [68%]) enrolled within the registry of the European Competence Network on Mastocytosis were analyzed. RESULTS: Overall survival (OS) was adversely affected by the number of organomegalies (OS: #0 vs #1 hazard ratio [HR], 4.9; 95% CI, 3.4-7.1, P < .001; #1 vs #2 HR, 2.1, 95% CI, 1.4-3.1, P < .001; #2 vs #3 HR, 1.7, 95% CI, 1.2-2.5, P = .004). Lymphadenopathy was frequently detected in patients with smoldering SM (SSM, 18 of 60 [30%]) or advanced SM (AdvSM, 137 of 344 [40%]). Its presence confered an inferior outcome in patients with AdvSM compared with patients with AdvSM without lymphadenopathy (median OS, 3.8 vs 2.6 years; HR, 1.6; 95% CI, 1.2-2.2; P = .003). OS was not different between patients having organomegaly with either ISM or SSM (median, 25.5 years vs not reached; P = .435). At time of disease progression, a new occurrence of any organomegaly was observed in 17 of 40 (43%) patients with ISM, 4 of 10 (40%) patients with SSM, and 33 of 86 (38%) patients with AdvSM, respectively. CONCLUSIONS: Organomegalies including lymphadenopathy are often found in SSM and AdvSM. ISM with organomegaly has a similar course and prognosis compared with SSM. The number of organomegalies is adversely associated with OS. A new occurrence of organomegaly in all variants of SM may indicate disease progression. (c) 2022 American Academy of Allergy, Asthma & Immunology
- Published
- 2023
56. Germ line DDX41 mutations define a unique subtype of myeloid neoplasms
- Author
-
Hideki Makishima, Ryunosuke Saiki, Yasuhito Nannya, Sophia Korotev, Carmelo Gurnari, June Takeda, Yukihide Momozawa, Steve Best, Pramila Krishnamurthy, Tetsuichi Yoshizato, Yoshiko Atsuta, Yusuke Shiozawa, Yuka Iijima-Yamashita, Kenichi Yoshida, Yuichi Shiraishi, Yasunobu Nagata, Nobuyuki Kakiuchi, Makoto Onizuka, Kenichi Chiba, Hiroko Tanaka, Ayana Kon, Yotaro Ochi, Masahiro M. Nakagawa, Rurika Okuda, Takuto Mori, Akinori Yoda, Hidehiro Itonaga, Yasushi Miyazaki, Masashi Sanada, Takayuki Ishikawa, Shigeru Chiba, Hisashi Tsurumi, Senji Kasahara, Carsten Müller-Tidow, Akifumi Takaori-Kondo, Kazuma Ohyashiki, Toru Kiguchi, Fumihiko Matsuda, Joop H. Jansen, Chantana Polprasert, Piers Blombery, Yoichiro Kamatani, Satoru Miyano, Luca Malcovati, Torsten Haferlach, Michiaki Kubo, Mario Cazzola, Austin G. Kulasekararaj, Lucy A. Godley, Jaroslaw P. Maciejewski, and Seishi Ogawa
- Subjects
All institutes and research themes of the Radboud University Medical Center ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Cell Biology ,Hematology ,Settore MED/15 ,Biochemistry - Abstract
Germ line DDX41 variants have been implicated in late-onset myeloid neoplasms (MNs). Despite an increasing number of publications, many important features of DDX41-mutated MNs remain to be elucidated. Here we performed a comprehensive characterization of DDX41-mutated MNs, enrolling a total of 346 patients with DDX41 pathogenic/likely-pathogenic (P/LP) germ line variants and/or somatic mutations from 9082 MN patients, together with 525 first-degree relatives of DDX41-mutated and wild-type (WT) patients. P/LP DDX41 germ line variants explained ∼80% of known germ line predisposition to MNs in adults. These risk variants were 10-fold more enriched in Japanese MN cases (n = 4461) compared with the general population of Japan (n = 20 238). This enrichment of DDX41 risk alleles was much more prominent in male than female (20.7 vs 5.0). P/LP DDX41 variants conferred a large risk of developing MNs, which was negligible until 40 years of age but rapidly increased to 49% by 90 years of age. Patients with myelodysplastic syndromes (MDS) along with a DDX41-mutation rapidly progressed to acute myeloid leukemia (AML), which was however, confined to those having truncating variants. Comutation patterns at diagnosis and at progression to AML were substantially different between DDX41-mutated and WT cases, in which none of the comutations affected clinical outcomes. Even TP53 mutations made no exceptions and their dismal effect, including multihit allelic status, on survival was almost completely mitigated by the presence of DDX41 mutations. Finally, outcomes were not affected by the conventional risk stratifications including the revised/molecular International Prognostic Scoring System. Our findings establish that MDS with DDX41-mutation defines a unique subtype of MNs that is distinct from other MNs.
- Published
- 2023
57. A continuous-time Markov model approach for modeling myelodysplastic syndromes progression from cross-sectional data.
- Author
-
Giovanna Nicora, F. Moretti, Elisabetta Sauta, Matteo Giovanni Della Porta, Luca Malcovati, Mario Cazzola, Silvana Quaglini, and Riccardo Bellazzi
- Published
- 2020
- Full Text
- View/download PDF
58. Incidence and prognosis of clonal hematopoiesis in patients with chronic idiopathic neutropenia
- Author
-
Gabriele Todisco, Irene Fragiadaki, Stavros Papadakis, Helen A. Papadaki, Chiara Elena, Anastasios Batas, Anna Gallì, Elisabetta Molteni, Elisa Bono, Grigorios Tsaknakis, Luca Malcovati, Peggy Kanellou, Irene Mavroudi, Charalampos Pontikoglou, Stella Maxouri, Emmanouela Linardaki, Ettore Rizzo, and Nektarios Tavernarakis
- Subjects
Adult ,Male ,medicine.medical_specialty ,Neutropenia ,Myeloid ,IDH1 ,Immunology ,Biochemistry ,Gastroenterology ,Malignant transformation ,Young Adult ,Gene Frequency ,Internal medicine ,medicine ,Humans ,Clinical significance ,Aged ,Aged, 80 and over ,Cytopenia ,business.industry ,Incidence ,Incidence (epidemiology) ,Cancer ,Cell Biology ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,medicine.anatomical_structure ,Mutation ,Female ,Clonal Hematopoiesis ,business ,Follow-Up Studies - Abstract
The incidence and prognosis of clonal hematopoiesis in patients with isolated neutropenia among patients with idiopathic cytopenia of undetermined significance (ICUS), known as ICUS-N or chronic idiopathic neutropenia (CIN) patients, is poorly defined. The current study sought to investigate the frequency and clinical significance of mutations of genes implicated in myeloid malignancies using next-generation sequencing in patients with CIN (n = 185) with a long follow-up. We found that 21 (11.35%) of 185 patients carried a total of 25 somatic mutations in 6 genes with a median variant allele frequency of 12.75%. The most frequently mutated genes were DNMT3A and TET2 involving >80% of patients, followed by IDH1/2, SRSF2, and ZRSR2. The frequency of transformation to a myeloid malignancy was low in the total group of patients (5 of 185 patients [2.70%]). However, from the transformed patients, 4 belonged to the clonal group (4 of 21 [19.05%]) and 1 to the nonclonal group (1 of 164 [0.61%]), indicating that the presence of mutation(s) confers a relative risk for transformation of 31.24 (P = .0017). The variant allele frequency of the mutant clones in the transformed patients was >10% in all cases, and the genes most frequently associated with malignant transformation were SRSF2 and IDH1. No significant differences were identified between the clonal and nonclonal groups in the severity of neutropenia. Patients with clonal disease were older compared with nonclonal patients. These data contribute to the better understanding of the heterogeneous entities underlying ICUS and highlight the importance of mutation analysis for the diagnosis and prognosis of patients with unexplained neutropenias.
- Published
- 2021
- Full Text
- View/download PDF
59. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data
- Author
-
Daniel A. Arber, Attilio Orazi, Robert P. Hasserjian, Michael J. Borowitz, Katherine R. Calvo, Hans-Michael Kvasnicka, Sa A. Wang, Adam Bagg, Tiziano Barbui, Susan Branford, Carlos E. Bueso-Ramos, Jorge E. Cortes, Paola Dal Cin, Courtney D. DiNardo, Hervé Dombret, Eric J. Duncavage, Benjamin L. Ebert, Elihu H. Estey, Fabio Facchetti, Kathryn Foucar, Naseema Gangat, Umberto Gianelli, Lucy A. Godley, Nicola Gökbuget, Jason Gotlib, Eva Hellström-Lindberg, Gabriela S. Hobbs, Ronald Hoffman, Elias J. Jabbour, Jean-Jacques Kiladjian, Richard A. Larson, Michelle M. Le Beau, Mignon L.-C. Loh, Bob Löwenberg, Elizabeth Macintyre, Luca Malcovati, Charles G. Mullighan, Charlotte Niemeyer, Olatoyosi M. Odenike, Seishi Ogawa, Alberto Orfao, Elli Papaemmanuil, Francesco Passamonti, Kimmo Porkka, Ching-Hon Pui, Jerald P. Radich, Andreas Reiter, Maria Rozman, Martina Rudelius, Michael R. Savona, Charles A. Schiffer, Annette Schmitt-Graeff, Akiko Shimamura, Jorge Sierra, Wendy A. Stock, Richard M. Stone, Martin S. Tallman, Jürgen Thiele, Hwei-Fang Tien, Alexandar Tzankov, Alessandro M. Vannucchi, Paresh Vyas, Andrew H. Wei, Olga K. Weinberg, Agnieszka Wierzbowska, Mario Cazzola, Hartmut Döhner, Ayalew Tefferi, Arber, Daniel A, Orazi, Attilio, Hasserjian, Robert P, Borowitz, Michael J, Branford, Susan, Tefferi, Ayalew, and Hematology
- Subjects
Consensus ,Leukemia ,Myeloproliferative Disorders ,Immunology ,Genomics ,Cell Biology ,Hematology ,Settore MED/08 - Anatomia Patologica ,World Health Organization ,Biochemistry ,Hematologic Neoplasms ,Acute Disease ,Humans ,myeloid neoplasia ,lymphoid neoplasia - Abstract
The classification of myeloid neoplasms and acute leukemias was last updated in 2016 within a collaboration between the World Health Organization (WHO), the Society for Hematopathology, and the European Association for Haematopathology. This collaboration was primarily based on input from a clinical advisory committees (CACs) composed of pathologists, hematologists, oncologists, geneticists, and bioinformaticians from around the world. The recent advances in our understanding of the biology of hematologic malignancies, the experience with the use of the 2016 WHO classification in clinical practice, and the results of clinical trials have indicated the need for further revising and updating the classification. As a continuation of this CAC-based process, the authors, a group with expertise in the clinical, pathologic, and genetic aspects of these disorders, developed the International Consensus Classification (ICC) of myeloid neoplasms and acute leukemias. Using a multiparameter approach, the main objective of the consensus process was the definition of real disease entities, including the introduction of new entities and refined criteria for existing diagnostic categories, based on accumulated data. The ICC is aimed at facilitating diagnosis and prognostication of these neoplasms, improving treatment of affected patients, and allowing the design of innovative clinical trials.
- Published
- 2022
60. Genomic profiling for clinical decision making in myeloid neoplasms and acute leukemia
- Author
-
Eric J. Duncavage, Adam Bagg, Robert P. Hasserjian, Courtney D. DiNardo, Lucy A. Godley, Ilaria Iacobucci, Siddhartha Jaiswal, Luca Malcovati, Alessandro M. Vannucchi, Keyur P. Patel, Daniel A. Arber, Maria E. Arcila, Rafael Bejar, Nancy Berliner, Michael J. Borowitz, Susan Branford, Anna L. Brown, Catherine A. Cargo, Hartmut Döhner, Brunangelo Falini, Guillermo Garcia-Manero, Torsten Haferlach, Eva Hellström-Lindberg, Annette S. Kim, Jeffery M. Klco, Rami Komrokji, Mignon Lee-Cheun Loh, Sanam Loghavi, Charles G. Mullighan, Seishi Ogawa, Attilio Orazi, Elli Papaemmanuil, Andreas Reiter, David M. Ross, Michael Savona, Akiko Shimamura, Radek C. Skoda, Francesc Solé, Richard M. Stone, Ayalew Tefferi, Matthew J. Walter, David Wu, Benjamin L. Ebert, and Mario Cazzola
- Subjects
Leukemia, Myeloid, Acute ,Myeloproliferative Disorders ,Neoplasms ,Hematologic Neoplasms ,Immunology ,Mutation ,Clinical Decision-Making ,Humans ,Cell Biology ,Hematology ,Genomics ,Biochemistry - 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.
- Published
- 2022
61. Monocytosis and its association with clonal hematopoiesis in community-dwelling individuals
- Author
-
Isabelle A. van Zeventer, Aniek O. de Graaf, Theresia N. Koorenhof-Scheele, Bert A. van der Reijden, Melanie M. van der Klauw, Avinash G. Dinmohamed, Arjan Diepstra, Jan Jacob Schuringa, Luca Malcovati, Gerwin Huls, Joop H. Jansen, Life Course Epidemiology (LCE), Stem Cell Aging Leukemia and Lymphoma (SALL), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Hematology
- Subjects
Adult ,All institutes and research themes of the Radboud University Medical Center ,Myeloproliferative Disorders ,SDG 3 - Good Health and Well-being ,Leukocytosis ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Mutation ,Humans ,Leukemia, Myelomonocytic, Chronic ,Hematology ,Independent Living ,Clonal Hematopoiesis - Abstract
Monocytosis may occur in numerous inflammatory conditions but is also the defining feature of chronic myelomonocytic leukemia (CMML). Clonal somatic mutations detectable in CMML may occur with aging in otherwise healthy individuals, so-called “clonal hematopoiesis” (CH). We investigated whether the combination of CH and monocytosis would represent an early developmental stage of CMML. We studied community-dwelling individuals with monocytosis (≥1 × 109/L and ≥10% of leukocytes) in the population-based Lifelines cohort (n = 144 676 adults). The prevalence and spectrum of CH were evaluated for individuals ≥60 years with monocytosis (n = 167 [0.8%]), and control subjects 1:3 matched for age and sex (n = 501). Diagnoses of hematological malignancies were retrieved by linkage to the Netherlands Cancer Registry (NCR). Monocyte counts and the prevalence of monocytosis increased with advancing age. Older individuals with monocytosis more frequently carried CH (50.9% vs 35.5%; P < .001). Monocytosis is associated with enrichment of multiple gene mutations (P = .006) and spliceosome mutations (P = .007) but not isolated mutated DNMT3A, TET2, or ASXL1. Persistent monocytosis over 4 years was observed in 30/102 evaluable individuals and associated with a higher prevalence of CH (63%). Myeloid malignancies, including 1 case of CMML, developed in 4 individuals with monocytosis who all carried CH. In conclusion, monocytosis and CH both occur at an older age and do not necessarily reflect clonal monocytic proliferation. In a fraction of older subjects with monocytosis, CH might constitute early clonal dominance in developing malignant myelomonocytic disease. Mutational spectra deviating from age-related CH require attention.
- Published
- 2022
62. Molecular International Prognostic Scoring System for Myelodysplastic Syndromes
- Author
-
Elsa Bernard, Heinz Tuechler, Peter L. Greenberg, Robert P. Hasserjian, Juan E. Arango Ossa, Yasuhito Nannya, Sean M. Devlin, Maria Creignou, Philippe Pinel, Lily Monnier, Gunes Gundem, Juan S. Medina-Martinez, Dylan Domenico, Martin Jädersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Lurdes Zamora, Ronald F. Pinheiro, Andrea Pellagatti, Harold K. Elias, Detlef Haase, Christina Ganster, Lionel Ades, Magnus Tobiasson, Laura Palomo, Matteo Giovanni Della Porta, Akifumi Takaori-Kondo, Takayuki Ishikawa, Shigeru Chiba, Senji Kasahara, Yasushi Miyazaki, Agnes Viale, Kety Huberman, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio P. S. Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Kazuma Ohyashiki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Luca Malcovati, Mario Cazzola, Seishi Ogawa, Eva Hellström-Lindberg, and Elli Papaemmanuil
- Published
- 2022
- Full Text
- View/download PDF
63. Co-mutation pattern, clonal hierarchy, and clone size concur to determine disease phenotype of SRSF2P95-mutated neoplasms
- Author
-
Maria Creignou, Anna Gallì, Paola Guglielmelli, Mario Cazzola, Johanna Ungerstedt, Seishi Ogawa, Silvia Catricalà, Ettore Rizzo, Marios Dimitriou, Eva Hellström-Lindberg, Elisa Bono, Alessandro M. Vannucchi, Martina Sarchi, Elisa Rumi, Luca Malcovati, Vittorio Rosti, Yasuhito Nannya, Marco Roncador, Daniela Pietra, Chiara Elena, Elisabetta Molteni, and Gabriele Todisco
- Subjects
0301 basic medicine ,Genetics ,Cancer Research ,education.field_of_study ,Myeloid ,Genetic heterogeneity ,Myelodysplastic syndromes ,Population ,Myeloid leukemia ,Hematology ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Monocytosis ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,education ,Myelofibrosis ,Dominance (genetics) - Abstract
Somatic mutations in splicing factor genes frequently occur in myeloid neoplasms. While SF3B1 mutations are associated with myelodysplastic syndromes (MDS) with ring sideroblasts, SRSF2P95 mutations are found in different disease categories, including MDS, myeloproliferative neoplasms (MPN), myelodysplastic/myeloproliferative neoplasms (MDS/MPN), and acute myeloid leukemia (AML). To identify molecular determinants of this phenotypic heterogeneity, we explored molecular and clinical features of a prospective cohort of 279 SRSF2P95-mutated cases selected from a population of 2663 patients with myeloid neoplasms. Median number of somatic mutations per subject was 3. Multivariate regression analysis showed associations between co-mutated genes and clinical phenotype, including JAK2 or MPL with myelofibrosis (OR = 26.9); TET2 with monocytosis (OR = 5.2); RAS-pathway genes with leukocytosis (OR = 5.1); and STAG2, RUNX1, or IDH1/2 with blast phenotype (MDS or AML) (OR = 3.4, 1.9, and 2.1, respectively). Within patients with SRSF2-JAK2 co-mutation, JAK2 dominance was invariably associated with clinical feature of MPN, whereas SRSF2 mutation was dominant in MDS/MPN. Within patients with SRSF2-TET2 co-mutation, clinical expressivity of monocytosis was positively associated with co-mutated clone size. This study provides evidence that co-mutation pattern, clone size, and hierarchy concur to determine clinical phenotype, tracing relevant genotype-phenotype associations across disease entities and giving insight on unaccountable clinical heterogeneity within current WHO classification categories.
- Published
- 2020
- Full Text
- View/download PDF
64. Mutational spectrum and dynamics of clonal hematopoiesis in anemia of older individuals
- Author
-
Isabelle A van Zeventer, Gerwin Huls, Luca Malcovati, Hanneke J C M Wouters, Marianne A. Jonker, Aniek O. de Graaf, Melanie M. van der Klauw, Joop H. Jansen, Theo de Witte, Bert A. van der Reijden, Life Course Epidemiology (LCE), Stem Cell Aging Leukemia and Lymphoma (SALL), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
0301 basic medicine ,Male ,SF3B1 MUTATION ,Aging ,Anemia ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Population ,Physiology ,Kaplan-Meier Estimate ,medicine.disease_cause ,Biochemistry ,Somatic evolution in cancer ,03 medical and health sciences ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,Medicine ,Humans ,Prospective Studies ,Prospective cohort study ,education ,Aged ,RISK ,Mutation ,education.field_of_study ,business.industry ,ORIGIN ,Clonal hematopoiesis ,Age Factors ,Cell Biology ,Hematology ,SOMATIC MUTATIONS ,Middle Aged ,medicine.disease ,EVOLUTION ,3. Good health ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,Hematopoiesis ,PREVALENCE ,Haematopoiesis ,030104 developmental biology ,Cohort ,DNMT3A ,Female ,business ,030215 immunology - Abstract
Anemia is a major and currently poorly understood clinical manifestation of hematopoietic aging. Upon aging, hematopoietic clones harboring acquired leukemia-associated mutations expand and become detectable, now referred to as clonal hematopoiesis (CH). To investigate the relationship between CH and anemia of the elderly, we explored the landscape and dynamics of CH in older individuals with anemia. From the prospective, population-based Lifelines cohort (n = 167 729), we selected all individuals at least 60 years old who have anemia according to World Health Organization criteria (n = 676) and 1:1 matched control participants. Peripheral blood of 1298 individuals was analyzed for acquired mutations at a variant allele frequency (VAF) of 1% or higher in 27 driver genes. To track clonal evolution over time, we included all available follow-up samples (n = 943). CH was more frequently detected in individuals with anemia (46.6%) compared with control individuals (39.1%; P = .007). Although no differences were observed regarding commonly detected DTA mutations (DNMT3A, TET2, ASXL1) in individuals with anemia compared with control individuals, other mutations were enriched in the anemia cohort, including TP53 and SF3B1. Unlike individuals with nutrient deficiency (P = .84), individuals with anemia of chronic inflammation and unexplained anemia revealed a higher prevalence of CH (P = .035 and P = .017, respectively) compared with their matched control individuals. Follow-up analyses revealed that clones may expand and decline, generally showing only a subtle increase in VAF (mean, 0.56%) over the course of 44 months, irrespective of the presence of anemia. Specific mutations were associated with different growth rates and propensities to acquire an additional hit. In contrast to smaller clones (
- Published
- 2020
- Full Text
- View/download PDF
65. Abstract 6168: Implementation and adoption of a web tool to support precision diagnostic and treatment decisions for patient with myelodysplastic syndromes
- Author
-
Elsa Bernard, Juan E. Arango Ossa, Heinz Tuechler, Peter L. Greenberg, Robert P. Hasserjian, Yasuhito Nannya, Sean M. Devlin, Maria Creignou, Philippe Pinel, Lily Monier, Juan S. Medina-Martinez, Dylan Domenico, Martin Jädersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Lurdes Zamora, Ronald F. Pinheiro, Andrea Pellagatti, Detlef Haase, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio P. Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Kazuma Ohyashiki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Luca Malcovati, Mario Cazzola, Seishi Ogawa, Eva Hellström-Lindberg, and Elli Papaemmanuil
- Subjects
Cancer Research ,Oncology - Abstract
Despite a detailed understanding of the genes mutated in myelodysplastic syndromes (MDS), diagnostic and treatment decisions for patients with MDS rely primarily on clinical and cytogenetic variables as considered by the Revised International Prognostic Scoring System (IPSS-R). Here we describe the recently developed Molecular IPSS (IPSS-M), a clinico-genomic risk stratification system that considers clinical, cytogenetic and genetic parameters; the implementation of a web portal to facilitate its adoption, a strategy to handle missing variables, and the worldwide utilization of the web calculator as a clinical support tool. The IPSS-M was trained on 2,957 clinically annotated diagnostic MDS samples profiled for mutations in 156 driver genes. To maximize the clinical applicability of the IPSS-M and account for missing genetic data (i.e genes missing from a sequencing panel), we implemented a strategy to calculate a risk score under three scenarios: best, worst and average. Last, we developed an online calculator as a standalone single-page web application using VueJs, and D3Js for the interactive visualizations, deployed through a CI/CD pipeline on AWS, where collection of anonymous usage analytics allows to track adoption and usability of the new proposed model. The model incorporates clinical, morphological, genetic variables informed by cytogenetics and constructed from the presence of oncogenic mutations in 31 genes. It delivers a unique risk score for each individual patient, as well as an assignment to one of six IPSS-M risk strata. Compared to the IPSS-R the IPSS-M re-stratified 46% of MDS patients. The model was validated in an external dataset of 754 MDS patients. We released an open-access IPSS-M web calculator available at https://mds-risk-model.com. By specifying the patient clinical and molecular profiles, the tool returns the patient-specific IPSS-M risk score and category, and the probability estimates over time for three clinical endpoints, i.e. leukemia free survival (LFS), overall survival, and incidence of leukemic transformation. Since its launch in June 2022, the calculator has been used by >6000 users in >75 countries, reaching a daily average of 100 users per day. Risks have been calculated for >45,000 patient profiles. 99.28% of the sessions initiated reach an IPSS-M score, suggesting that the calculator is intuitive and easy to use. We trained and validated the IPSS-M on 3,711 patients, a patient tailored risk stratification tool for patients with MDS that considers clinical, morphological and genetic variables inclusive of cytogenetics and mutations in one of 31 genes. The development of a web based tool was instrumental to the global dissemination of the model, enabling non-expert users to leverage the power of molecular biomarkers in risk stratification for patients with MDS. Citation Format: Elsa Bernard, Juan E. Arango Ossa, Heinz Tuechler, Peter L. Greenberg, Robert P. Hasserjian, Yasuhito Nannya, Sean M. Devlin, Maria Creignou, Philippe Pinel, Lily Monier, Juan S. Medina-Martinez, Dylan Domenico, Martin Jädersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Lurdes Zamora, Ronald F. Pinheiro, Andrea Pellagatti, Detlef Haase, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio P. Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Kazuma Ohyashiki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Luca Malcovati, Mario Cazzola, Seishi Ogawa, Eva Hellström-Lindberg, Elli Papaemmanuil. Implementation and adoption of a web tool to support precision diagnostic and treatment decisions for patient with myelodysplastic syndromes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6168.
- Published
- 2023
- Full Text
- View/download PDF
66. Silk Bone Marrow-like Niches for Red Cell Production Ex Vivo
- Author
-
Christian A Di Buduo, Virginia Camilotto, Francesca Careddu, Marco Lunghi, Rosangela Invernizzi, Claudia Del Fante, Luca Malcovati, and Alessandra Balduini
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
67. Clonal Trajectories and Therapeutic Targeting of High-Risk SF3B1-Mutant Myelodysplastic Syndromes
- Author
-
Martina Sarchi, Courtnee A Clough, Anna Gallì, Gabriele Todisco, J. Philip Creamer, Luca Malcovati, and Sergei Doulatov
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
68. Management of MDS with Isolated Del(5q) Patients in the European MDS (EUMDS) Registry: A Report on 197 Cases
- Author
-
Inga Mandac Smoljanovic, Adele Taylor, Pierre Fenaux, Agnès Guerci-Bresler, Raphael Itzykson, Argiris Symeonidis, Ioannis Kotsianidis, Moshe Mittelman, Guillermo Sanz, Dominic J. Culligan, Jaroslav Cermak, Reinhard Stauder, Eva Hellström-Lindberg, Luca Malcovati, Saskia M.C. Langemeijer, Ulrich Germing, Krzysztof Madry, Howard S. Oster, Alexandra Smith, Corine van Marrewijk, Theo M de Witte, and David Bowen
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
69. Somatic mutational landscape of hereditary hematopoietic malignancies associated with germline variants in RUNX1, GATA2 and DDX41
- Author
-
Anna L. Brown, Claire Homan, Michael W. Drazer, Kai Yu, David Lawrence, Jinghua Feng, Luis Arriola-Martinez, Matthew Pozsgai, Kelsey McNeely, Thuong Ha, Parvathy Venugopal, Peer Arts, Sarah King-Smith, Jesse JC Cheah, Mark Armstrong, Csaba Bödör, Paul Wang, Alan B. Cantor, Mario Cazzola, Erin Degelman, Courtney D. DiNardo, Nicolas Duployez, Remi Favier, Stefan Fröhling, Ana Rio-Machin, Jeffery M. Klco, Alwin Krämer, Mineo Kurokawa, Joanne Lee, Luca Malcovati, Neil V Morgan, Georges Natsoulis, Carolyn Owen, Keyur P. Patel, Claude Preudhomme, Hana Raslova, Hugh Young Rienhoff, Tim Ripperger, Rachael Schulte, Kiran Tawana, Elvira Deolinda Rodrigues Pereira Velloso, Benedict Yan, Raman Sood, Amy Hsu, Steven M. Holland, Kerry Phillips, Nicola Poplawski, Milena Babic, Erika M Kwon Kim, Andrew H. Wei, Cecily Forsyth, Helen Mar Fan, Ian D Lewis, Julian Cooney, Rachel Susman, Lucy C Fox, Piers Blombery, Deepak Singhal, Devendra Hiwase, Andreas W Schreiber, Christopher N Hahn, Hamish S Scott, Paul P. Liu, Lucy A. Godley, Brown, Anna L, Homan, Claire, Drazer, Michael W, Yu, Kai, Ha, Thuong, Arts, Peer, King Smith, Sarah, Cheah, Jesse JC, Armstrong, Mark, Wang, Paul, Babic, Milena, Hahn, Christopher N, Scott, Hamish S, Godley, Lucy, and 64th Annual Meeting and Exposition of the American-Society-of-Hematology (ASH) New Orleans, US 10-13 December 2022
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Germline variants in RUNX1, GATA2 and DDX41 may confer a predisposition to hereditary haematopoietic malignancies (HHMs) such as MDS and AML yet have distinct age ranges of malignancy diagnosis and a highly variable overall risk for leukemogenesis. The increased awareness and identification of carriers of these germline variants, particularly before development of malignancy, has changed the way in which individuals and families need to be managed in the clinic. Individuals need lifelong monitoring and may also need modification to treatments when malignancy does develop, compared to sporadic counterparts. Gaps in understanding pre-malignant states in HHM syndromes have hampered efforts to design effective clinical surveillance regimes, provide personalized pre-emptive treatments, and appropriate counselling to patients.
- Published
- 2022
70. Refined diagnostic criteria for bone marrow mastocytosis : a proposal of the European competence network on mastocytosis
- Author
-
Anna Belloni Fortina, Khalid Shoumariyeh, Aleksandra Górska, Hans Hägglund, Oleksii Solomianyi, Francesca Caroppo, Andreas Reiter, Michel Arock, Bjorn van Anrooij, Peter Valent, Akif Selim Yavuz, Cecelia Perkins, Jason Gotlib, Cornelius Miething, Alexander Zink, Massimiliano Bonifacio, William Shomali, Christine Breynaert, Julien Rossignol, Mohamad Jawhar, Chiara Elena, Michael Doubek, Marek Niedoszytko, Sabine Müller, Jens Panse, Wolfgang R. Sperr, Luca Malcovati, Emir Hadzijusufovic, Vladan Vucinic, Vito Sabato, Judit Várkonyi, Patrizia Bonadonna, Massimo Triggiani, Anja Illerhaus, Luigi Scaffidi, Roberta Parente, Roberta Zanotti, Friederike Wortmann, Hanneke Oude Elberink, Hanneke C. Kluin-Nelemans, Magdalena Lange, Mattias Mattsson, Karin Hartmann, Olivier Hermine, Irena Angelova-Fischer, Tanja Schug, Knut Brockow, Giuseppe Lucchini, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Tryptase ,Skin Diseases ,World health ,Mastocytosis, Systemic ,Bone Marrow ,Internal medicine ,Overall survival ,medicine ,Humans ,Mast Cells ,Mastocytosis ,Systemic mastocytosis ,Aged ,Aged, 80 and over ,biology ,business.industry ,Network on ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Europe ,Survival Rate ,medicine.anatomical_structure ,biology.protein ,Female ,Tryptases ,Bone marrow ,Human medicine ,business ,Skin lesion ,Follow-Up Studies - Abstract
In the current classification of the World Health Organization (WHO), bone marrow mastocytosis (BMM) is a provisional variant of indolent systemic mastocytosis (ISM) defined by bone marrow involvement and absence of skin lesions. However, no additional diagnostic criteria for BMM have been proposed. Within the registry dataset of the European Competence Network on Mastocytosis, we compared characteristics and outcomes of 390 patients with BMM and 1175 patients with typical ISM. BMM patients were significantly older, predominantly male, had lower tryptase and lower burden of neoplastic mast cells, and displayed a higher frequency of allergic reactions, mainly triggered by Hymenoptera, than patients with typical ISM. The estimated 10-year progression-free survival of BMM and typical ISM was 95.9% and 92.6%, respectively. In BMM patients defined by WHO-based criteria, the presence of one B-Finding and tryptase level ≥125 ng/mL were identified as risk factors for progression in multivariate analyses. BMM patients without any of these risk factors were found to have better progression-free survival (p < 0.05) and better overall survival (p < 0.05) than other ISM patients. These data support the proposal to define BMM as a separate SM variant characterized by SM criteria, absence of skin lesions, absence of B-Findings, and tryptase levels
- Published
- 2022
71. Autoantibodies against type I IFNs in patients with Ph-negative myeloproliferative neoplasms
- Author
-
Oscar Borsani, Paul Bastard, Jérémie Rosain, Adrian Gervais, Emanuela Sant’Antonio, Daniele Vanni, Ilaria Carola Casetti, Daniela Pietra, Chiara Trotti, Silvia Catricalà, Virginia Valeria Ferretti, Luca Malcovati, Luca Arcaini, Jean-Laurent Casanova, Alessandro Borghesi, Elisa Rumi, University of Pavia, Human genetics of infectious diseases : Mendelian predisposition (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPC), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Département de Pédiatrie et maladies infectieuses [CHU Necker], CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Azienda Usl Toscana centro [Firenze], Fondazione IRCCS Policlinico San Matteo [Pavia], Università di Pavia, Howard Hughes Medical Institute (HHMI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Studies performed at the Department of Hematology, Fondazione IRCCS Policlinico San Matteo and Department of Molecular Medicine, University of Pavia were supported by grants from the Italian Ministry of Health for young researchers (GR-2016-02361272) (E.R.) and by Associazione Italiana per la Ricerca sul Cancro (AIRC: IG 2021 ID 25703) through the project 'Actionable targets in clonal progression and systemic spreading of myeloid neoplasms,' (IG 2021 ID 25703) (E.R.) MYNERVA project (Project Code: 21267) (E.R. and L.M.). This work was supported by The European Union’s Horizon 2020 research and innovation program (ATAC, 101003650), the Center for Innovative Medicine at Karolinska Institutet, the Swedish Research Council, the Knut and Alice Wallenberg Foundation (KAW). The Laboratory of Human Genetics of Infectious Diseases is supported by the Howard Hughes Medical Institute, the Rockefeller University, the St. Giles Foundation, the National Institute of Allergy and Infectious Diseases (NIAID) (grants R01AI088364 and R01AI163029), the National Center for Advancing Translational Sciences (NCATS), National Institutes of Health Clinical and Translational Science Award (CTSA) program (UL1TR001866), a Fast Grant from Emergent Ventures, Mercatus Center at George Mason University, the Fisher Center for Alzheimer’s Research Foundation, the Meyer Foundation, the JPB Foundation, the French National Research Agency (ANR) under the 'Investments for the Future' program (ANR-10-IAHU-01), ANR grants (ANR-14-CE14-0008-01, ANR-18-CE15-0020-02, ANR-20-CE93-003, ANR-20-CO11-000,1 and ANR-21-COVR-0039), the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID), the French Foundation for Medical Research (FRM) (EQU201903007798), the FRM and ANR GENCOVID project (ANR-20-COVI-0003), ANRS Nord-Sud (ANRS-COV05), the European Union’s Horizon 2020 Research and Innovation Program (grant agreement no. 824110) (EASI-Genomics), the Square Foundation, Grandir-Fonds de solidarité pour l’enfance, the SCOR Corporate Foundation for Science, Fondation du Souffle, Institut National de la Santé et de la Recherche Médicale (INSERM), REACTing-INSERM, and the University of Paris. P.B. received support from the French Foundation for Medical Research (FRM, EA20170638020) and was supported by the PhD program of the Imagine Institute (with the support of the Fondation Bettencourt-Schueller)., ANR-14-CE14-0008,IEIHSEER,L'encéphalite Herpétique de l'enfant résulte de déficits héréditaires d'immunité contre l'HSV-1: une exception ou une règle?(2014), ANR-18-CE15-0020,SEAe-HostFactors,Facteurs de susceptibilité de l'hôte à l'encéphalite pédiatrique en Asie du Sud Est(2018), ANR-20-CE93-0003,GENVIR,Analyse multi-omique de l'immunité anti-virale: de l'identification des circuits biologiques pertinents à la découverte de défauts monogéniques héréditaires de l'immunité chez les patients avec infections virales sévères(2020), ANR-20-CO11-0001,AABIFNCOV,Bases génétiques et immunologiques des auto-anticorps contre les interférons de type I prédisposant aux formes sévères de COVID-19.(2020), ANR-21-COVR-0039,GenMIS-C,Recherche des Déficits immunitaires innées monogéniques prédisposant au syndrome inflammatoire multisystémique chez l'enfant.(2021), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-20-COVI-0003,GENCOVID,Identification des défauts monogéniques de l'immunité responsables des formes sévères de COVID-19 chez les patients précédemment en bonne santé(2020), European Project: 824110,H2020-INFRAIA-2018-1,EASI-Genomics(2019), Università degli Studi di Pavia = University of Pavia (UNIPV), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)
- Subjects
MESH: Interferon Type I ,MESH: Humans ,Myeloproliferative Disorders ,[SDV]Life Sciences [q-bio] ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Neoplasms ,Interferon Type I ,MESH: Autoantibodies ,Humans ,MESH: Neoplasms ,ComputingMilieux_MISCELLANEOUS ,risk ,MESH: Myeloproliferative Disorders ,Autoantibodies - Abstract
International audience
- Published
- 2021
- Full Text
- View/download PDF
72. A Personalized Clinical-Decision Tool to Improve the Diagnostic Accuracy of Myelodysplastic Syndromes
- Author
-
Nathan Radakovich, Manja Meggendorfer, Luca Malcovati, Mikkael A. Sekeres, Jacob Shreve, Cameron Beau Hilton, Yazan Rouphail, Wencke Walter, Stephan Hutter, Sudipto Mukherjee, Cassandra M. Kerr, Babal K. Jha, Anna Gallì, Sarah Pozzi, Aaron T. Gerds, Cassandra M Kerr, Claudia Haferlach, Jaroslaw P. Maciejewski, Torsten Haferlach, and Aziz Nazha
- Subjects
Cytopenia ,medicine.medical_specialty ,business.industry ,Essential thrombocythemia ,Myelodysplastic syndromes ,Immunology ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,hemic and lymphatic diseases ,Internal medicine ,Cohort ,medicine ,Data monitoring committee ,business ,Myelofibrosis ,Myeloproliferative neoplasm - Abstract
Background While histo- and cytomorphological examinations are central to the diagnosis of myelodysplastic syndromes (MDS), significant inter-observer variability exists. The diagnosis can be challenging in pancytopenic patients (pts) without evidence of dysplasia and is contingent on observer expertise. We developed and externally validated a geno-clinical model that uses mutational data and peripheral blood counts/clinical variables to distinguish MDS from other myeloid malignancies. Methods Clinical and genomic data, including commercially available next-generation sequencing panels, were obtained for patients (pts) treated at the Cleveland Clinic (CC; 652 pts), Munich Leukemia Laboratory (MLL; 1509 pts), and the University of Pavia in Italy (UP, 536 pts). All patients had carried a diagnosis of MDS, chronic myelomonocytic leukemia (CMML), MDS/myeloproliferative neoplasm overlap (MDS/MPN), myeloproliferative neoplasm (MPN; either polycythemia vera, essential thrombocythemia, or myelofibrosis), clonal cytopenia of undetermined significance (CCUS), or idiopathic cytopenia of undetermined significance (ICUS). All diagnoses were established with bone marrow aspiration and according to World Health Organization 2017 criteria. The training cohort included data from CC and UP and randomly divided into learner (80%) and test (20%) cohorts. The final model was independently validated in the MLL cohort. A machine learning algorithm was used to build the model; multiple extraction algorithms were used to extract genomic/clinical variables on both the cohort and individual levels. Performance was evaluated according to the area under the curve of the receiver operating characteristic (ROC-AUC) and accuracy matrices. Results Among the 2697 pts included from all sites, the median age was 70 years [36 - 86]. Median hemoglobin (Hb) was 10.4g/dl [6.9 - 15.7], median platelet count (PLT) was 132 k/dL [14 - 722], median WBC count was 5.3 k/dL [1.4 - 49.9], median ANC was 2.8 k/dL [0.3 - 27.7], median monocyte count was 0.3 k/dL [0 - 9.9], and median lymphocyte count (ALC) was 1.1 k/dL [0.1 - 5.4], and median peripheral blast percentage 0% [0 - 8]. The most commonly mutated genes in all patients were (list top 5 genes) and among pts with MDS were SF3B1 (27%), TET2 (25%), ASXL1 (19%), SRSF2 (16%), and DNMT3A (11%); among patients with MDS-MPN/CMML, the most commonly mutated genes were MDS-MPN/CMML (TET2 46%, ASXL1 34%, SRSF2 29%, RUNX1 13%, CBL 12%) ; among patients with MPNs, the most commonly mutated genes were (JAK2 64%, ASXL1 27%, TET2 14%, DNMT3A 8%, U2AF1 7%); among patients with CCUS the most commonly mutated genes were (TET2 41%, DNMT3A 27%, ASXL1 19%, SRSF2 17%, ZRSR2 10%). The most important features for model predictions (ranked from the most to the least important) included: number of mutations detected/sample, peripheral blast percentage, AMC, JAK2 status, Hb, basophil count, age, eosinophil count, ALC, WBC, EZH2 mutation status, ANC, mutation status of KRAS and SF3B1, platelets, and gender. The final model achieved an average AUROC of 0.95 (95% CI 0.93-0.96) when applied to the test cohort and 0.93 (95% CI 0.91 - 0.94) when it was applied to the MLL cohort. The model also provides individual-level explanations for predictions, providing top differential diagnoses and individual-level explanations of how features influence a putative diagnosis (Figure 1b). Conclusions We developed and externally validated a highly accurate and interpretable model that can distinguish MDS from other myeloid malignancies using clinical and mutational data from a large international cohort. The model can provide personalized interpretations of its outcome and can aid physicians and hematopathologists in recognizing MDS with high accuracy when encountering pts with pancytopenia and with a suspected diagnosis of MDS. Disclosures Sekeres: Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda/Millenium: Consultancy, Membership on an entity's Board of Directors or advisory committees; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees. Mukherjee:Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Partnership for Health Analytic Research, LLC (PHAR, LLC): Honoraria; Bristol Myers Squib: Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Aplastic Anemia and MDS International Foundation: Honoraria; Celgene/Acceleron: Membership on an entity's Board of Directors or advisory committees; EUSA Pharma: Consultancy. Gerds:Sierra Oncology: Research Funding; Imago Biosciences: Research Funding; Apexx Oncology: Consultancy; Celgene: Consultancy, Research Funding; Incyte Corporation: Consultancy, Research Funding; Roche/Genentech: Research Funding; CTI Biopharma: Consultancy, Research Funding; AstraZeneca/MedImmune: Consultancy; Gilead Sciences: Research Funding; Pfizer: Research Funding. Maciejewski:Alexion, BMS: Speakers Bureau; Novartis, Roche: Consultancy, Honoraria. Nazha:Jazz: Research Funding; Incyte: Speakers Bureau; Novartis: Speakers Bureau; MEI: Other: Data monitoring Committee.
- Published
- 2020
- Full Text
- View/download PDF
73. Divergent mutational processes distinguish hypoxic and normoxic tumours
- Author
-
Justin Whalley, Robert Bristow, Choon Kiat Ong, Samuel Aparicio, Paul Boutros, Raquel Rabionet, Elizabeth Christie, Markus Ringnér, Rosa Karlić, Shuto Hayashi, Sergei Yakneen, Vinayak Bhandari, Nigel Jamieson, Benedikt Brors, and LUCA MALCOVATI
- Subjects
0301 basic medicine ,Unknown aetiology ,Medizin ,Genes, myc ,General Physics and Astronomy ,Genome ,0302 clinical medicine ,Neoplasms ,Cancer genomics ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,Aetiology ,lcsh:Science ,Cancer ,0303 health sciences ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,Multidisciplinary ,Manchester Cancer Research Centre ,Single Nucleotide ,myc ,Cell Hypoxia ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,3. Good health ,030220 oncology & carcinogenesis ,medicine.symptom ,Human ,Cancer microenvironment ,Tumour heterogeneity ,Science ,Genomic Structural Variation ,Biology ,Polymorphism, Single Nucleotide ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Genetics ,PTEN ,Humans ,Polymorphism ,Gene ,030304 developmental biology ,Whole genome sequencing ,Whole Genome Sequencing ,Genome, Human ,ResearchInstitutes_Networks_Beacons/mcrc ,Human Genome ,PTEN Phosphohydrolase ,PCAWG Consortium ,General Chemistry ,Hypoxia (medical) ,medicine.disease ,030104 developmental biology ,Good Health and Well Being ,Genes ,Mutation ,Cancer research ,biology.protein ,Tumor Hypoxia ,lcsh:Q ,Molecular oxygen ,Tumor Suppressor Protein p53 - Abstract
Many primary tumours have low levels of molecular oxygen (hypoxia), and hypoxic tumours respond poorly to therapy. Pan-cancer molecular hallmarks of tumour hypoxia remain poorly understood, with limited comprehension of its associations with specific mutational processes, non-coding driver genes and evolutionary features. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2658 cancers across 38 tumour types, we quantify hypoxia in 1188 tumours spanning 27 cancer types. Elevated hypoxia associates with increased mutational load across cancer types, irrespective of underlying mutational class. The proportion of mutations attributed to several mutational signatures of unknown aetiology directly associates with the level of hypoxia, suggesting underlying mutational processes for these signatures. At the gene level, driver mutations in TP53, MYC and PTEN are enriched in hypoxic tumours, and mutations in PTEN interact with hypoxia to direct tumour evolutionary trajectories. Overall, hypoxia plays a critical role in shaping the genomic and evolutionary landscapes of cancer., Many tumours exhibit hypoxia (low oxygen) and hypoxic tumours often respond poorly to therapy. Here, the authors quantify hypoxia in 1188 tumours from 27 cancer types, showing elevated hypoxia links to increased mutational load, directing evolutionary trajectories.
- Published
- 2020
- Full Text
- View/download PDF
74. Loss of lenalidomide-induced megakaryocytic differentiation leads to therapy resistance in del(5q) myelodysplastic syndrome
- Author
-
Austin G. Kulasekararaj, Martin Jädersten, Patricia Umlandt, Jihong Jiang, Aly Karsan, Sergio Martinez-Høyer, Megan Fuller, Uwe Platzbecker, Nadia Gharaee, Luca Malcovati, T. Roderick Docking, Eva Hellström-Lindberg, Yu Deng, Angela Mo, Alan F. List, Jenny Li, Jeremy Parker, and Hematology
- Subjects
0303 health sciences ,Mutation ,Megakaryocyte differentiation ,Myelodysplastic syndromes ,GATA2 ,Context (language use) ,Cell Biology ,Biology ,medicine.disease ,medicine.disease_cause ,3. Good health ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,030304 developmental biology ,Lenalidomide ,medicine.drug - Abstract
Interstitial deletion of the long arm of chromosome 5 (del(5q)) is the most common structural genomic variant in myelodysplastic syndromes (MDS)1. Lenalidomide (LEN) is the treatment of choice for patients with del(5q) MDS, but half of the responding patients become resistant2 within 2 years. TP53 mutations are detected in ~20% of LEN-resistant patients3. Here we show that patients who become resistant to LEN harbour recurrent variants of TP53 or RUNX1. LEN upregulated RUNX1 protein and function in a CRBN- and TP53-dependent manner in del(5q) cells, and mutation or downregulation of RUNX1 rendered cells resistant to LEN. LEN induced megakaryocytic differentiation of del(5q) cells followed by cell death that was dependent on calpain activation and CSNK1A1 degradation4,5. We also identified GATA2 as a LEN-responsive gene that is required for LEN-induced megakaryocyte differentiation. Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Overexpression of GATA2 restored LEN sensitivity in the context of RUNX1 or TP53 mutations by enhancing LEN-induced megakaryocytic differentiation. Screening for mutations that block LEN-induced megakaryocytic differentiation should identify patients who are resistant to LEN.
- Published
- 2020
- Full Text
- View/download PDF
75. Guideline-based indicators for adult patients with myelodysplastic syndromes
- Author
-
Juerg Bernhard, Antonio Almeida, Monika Heger, Nicolas Bonadies, Jaroslav Cermak, Christine Morgenthaler, Avinash G Dinmohamed, Luca Malcovati, Theo de Witte, Georg Stussi, Reinhard Stauder, Sämi Schär, Michael Pfeilstöcker, Kristina Stojkov, Ulrich Germing, David P. Steensma, Charlotte Maddox, Moshe Mittelman, David L. B. Schwappach, Manuel Haschke, Sabrina Eggmann, Argiris Symeonidis, Julia Bohlius, Jakob Passweg, Pierre Fenaux, Corien Eeltink, Arjan A. van de Loosdrecht, Eva Hellström-Lindberg, Tobias Silzle, David G. Bowen, Valeria Santini, Uwe Platzbecker, Hematology, CCA - Cancer Treatment and quality of life, and Public Health
- Subjects
Adult ,medicine.medical_specialty ,Evidence-based practice ,Standardization ,Best practice ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,MEDLINE ,Standardized test ,610 Medicine & health ,All institutes and research themes of the Radboud University Medical Center ,Multidisciplinary approach ,360 Social problems & social services ,Health care ,medicine ,Humans ,Aged ,myelodysplastic syndromes, treatment guidelines ,business.industry ,Health Services and Outcomes ,Hematology ,Guideline ,Family medicine ,Myelodysplastic Syndromes ,business - Abstract
Myelodysplastic syndromes (MDSs) represent a heterogeneous group of hematological stem cell disorders with an increasing burden on health care systems. Evidence-based MDS guidelines and recommendations (G/Rs) are published but do not necessarily translate into better quality of care if adherence is not maintained in daily clinical practice. Guideline-based indicators (GBIs) are measurable elements for the standardized assessment of quality of care and, thus far, have not been developed for adult MDS patients. To this end, we screened relevant G/Rs published between 1999 and 2018 and aggregated all available information as candidate GBIs into a formalized handbook as the basis for the subsequent consensus rating procedure. An international multidisciplinary expert panel group (EPG) of acknowledged MDS experts (n = 17), health professionals (n = 7), and patient advocates (n = 5) was appointed. The EPG feedback rates for the first and second round were 82% (23 of 28) and 96% (26 of 27), respectively. A final set of 29 GBIs for the 3 domains of diagnosis (n = 14), therapy (n = 8), and provider/infrastructural characteristics (n = 7) achieved the predefined agreement score for selection (>70%). We identified shortcomings in standardization of patient-reported outcomes, toxicity, and geriatric assessments that need to be optimized in the future. Our GBIs represent the first comprehensive consensus on measurable elements addressing best practice performance, outcomes, and structural resources. They can be used as a standardized instrument with the goal of assessing, comparing, and fostering good quality of care within clinical development cycles in the daily care of adult MDS patients.
- Published
- 2020
- Full Text
- View/download PDF
76. Clinical, histopathological and molecular characterization of hypoplastic myelodysplastic syndrome
- Author
-
Luca Malcovati, Erica Travaglino, Anna Gallì, Kavita Raj, Virginia Valeria Ferretti, Elisa Bono, Jie Jiang, Chiara Elena, Emanuela Boveri, Antonio Bianchessi, Gabriele Todisco, Robin M. Ireland, Austin G. Kulasekararaj, Alesia Abigael Khan, Judith C. W. Marsh, Ghulam J. Mufti, Donal P. McLornan, Mario Cazzola, and Shreyans Gandhi
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Adolescent ,Myeloid Neoplasm ,Cohort Studies ,Diagnosis, Differential ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Bone Marrow ,hemic and lymphatic diseases ,medicine ,Humans ,Aplastic anemia ,Aged ,Aged, 80 and over ,Chromosome Aberrations ,business.industry ,Myelodysplastic syndromes ,Anemia, Aplastic ,Hematology ,Middle Aged ,medicine.disease ,030104 developmental biology ,Hypocellularity ,medicine.anatomical_structure ,Oncology ,Karyotyping ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Cohort ,Female ,Bone marrow ,Differential diagnosis ,business - Abstract
Diagnostic criteria for hypoplastic myelodysplasic syndrome (h-MDS) have not been clearly established, making the differential diagnosis from other bone marrow failure syndromes (BMF) challenging. In this study, we aimed to delineate clinical, histopathological, and molecular features of h-MDS, based on a large and well-annotated cohort of patients with bone marrow (BM) hypocellularity. The study included 534 consecutive adult patients with hypocellular BM (278 h-MDS and 136 aplastic anemia), and 727 with normo- or hypercellular MDS (n-MDS). Comparison of clinical features of patients with h-MDS as defined by BM cellularity ≤25% (n = 204) or reduced age-adjusted cellularity (n = 74) did not reveal significant differences. We developed a diagnostic score to discriminate h-MDS from non-malignant BMF based on histological and cytological variables with the highest specificity for MDS (h-score). The information from chromosomal abnormalities and somatic mutation patterns was then integrated into a cyto-histological/genetic score (hg-score). This score was able to segregate two groups of h-MDS with a significantly different risk of blast progression (P
- Published
- 2019
- Full Text
- View/download PDF
77. Incidence, Clinical Associations, and Co-Mutation Patterns of UBA1 Mutations in MDS
- Author
-
Maria Sirenko, Elsa Bernard, David B. Beck, Maria Creignou, Dylan Domenico, Andrea Farina, Juan E Arango, Olivier Kosmider, Robert P. Hasserjian, Martin Jadersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Matilde Y Follo, Felicitas R. Thol, Lurdes Zamora, Ronald Feitosa Pinheiro, Andrea Pellagatti, Harold K. Elias, Detlef Haase, Christina Ganster, Lionel Ades, Magnus Tobiasson, Laura Palomo, Matteo G. Della Porta, Kety Huberman, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio Pires de Souza Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Seishi Ogawa, Michaela Fontenay, Joop H. Jansen, Jose Cervera, Benjamin L. Ebert, Rafael Bejar, Peter L Greenberg, Norbert Gattermann, Luca Malcovati, Mario Cazzola, Eva Hellström-Lindberg, and Elli Papaemmanuil
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
78. Investigating the Molecular Mechanism of H3B-8800: A Splicing Modulator Inducing Preferential Lethality in Spliceosome-Mutant Cancers
- Author
-
Jure Borišek, Angelo Spinello, Luca Malcovati, Alessandra Magistrato, Spinello A., Borisek J., Malcovati L., and Magistrato A.
- Subjects
Spliceosome ,QH301-705.5 ,Protein Conformation ,Pyridines ,RNA Splicing ,Mutant ,Druggability ,H3B-8800 ,Molecular Dynamics Simulation ,medicine.disease_cause ,Catalysis ,Piperazines ,Article ,Inorganic Chemistry ,Neoplasms ,spliceosome-mutant cancer ,medicine ,Humans ,Physical and Theoretical Chemistry ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,Mutation ,splicing modulators ,Chemistry ,Organic Chemistry ,Wild type ,Intron ,leukemia ,General Medicine ,Phosphoproteins ,H3B‐8800 ,molecular dynamics ,Computer Science Applications ,Cell biology ,Phenotype ,Cancer cell ,RNA splicing ,RNA Splicing Factors ,Spliceosome‐mutant cancer - Abstract
The SF3B1 protein, part of the SF3b complex, recognizes the intron branch point sequence of precursor messenger RNA (pre-mRNA), thus contributing to splicing fidelity. SF3B1 is frequently mutated in cancer and is the target of distinct families of splicing modulators (SMs). Among these, H3B-8800 is of particular interest, as it induces preferential lethality in cancer cells bearing the frequent and highly pathogenic K700E SF3B1 mutation. Despite the potential of H3B-8800 to treat myeloid leukemia and other cancer types hallmarked by SF3B1 mutations, the molecular mechanism underlying its preferential lethality towards spliceosome-mutant cancer cells remains elusive. Here, microsecond-long all-atom simulations addressed the binding/dissociation mechanism of H3B-8800 to wild type and K700E SF3B1-containing SF3b (K700ESB3b) complexes at the atomic level, unlocking that the K700E mutation little affects the thermodynamics and kinetic traits of H3B-8800 binding. This supports the hypothesis that the selectivity of H3B-8800 towards mutant cancer cells is unrelated to its preferential targeting of K700ESB3b. Nevertheless, this set of simulations discloses that the K700E mutation and H3B-8800 binding affect the overall SF3b internal motion, which in turn may influence the way SF3b interacts with other spliceosome components. Finally, we unveil the existence of a putative druggable SF3b pocket in the vicinity of K700E that could be harnessed in future rational drug-discovery efforts to specifically target mutant SF3b.
- Published
- 2021
79. Clonal hematopoiesis and myeloid malignancies: clonal dynamics and clinical implications
- Author
-
Gabriele Todisco, Chiara Elena, Anna Gallì, Elisa Bono, and Luca Malcovati
- Subjects
Oncology ,Cytopenia ,medicine.medical_specialty ,Myeloid ,Myeloproliferative Disorders ,business.industry ,Anemia ,Preleukemia ,Context (language use) ,Hematology ,medicine.disease ,Malignancy ,Myeloid Neoplasm ,medicine.anatomical_structure ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,Hematologic Neoplasms ,medicine ,Humans ,Aplastic anemia ,Clonal Hematopoiesis ,business - Abstract
Purpose of review Clinical and experimental studies have uncovered relevant clinical implications of clonal hematopoiesis. However, the true magnitude of this process, clonal dynamics over time and mechanisms of progression into overt malignancy remain to be largely elucidated. In this article, the consequences of clonal hematopoiesis, its significance in the context of cytopenia, and its implications in the clinical management of patients with myeloid malignancies are reviewed and discussed. Recent findings Clonal hematopoiesis has been associated with higher risk of hematologic cancers, as well as of death from cardiovascular causes. Clonal hematopoiesis has been proven clinically relevant in the context of disorders characterized by peripheral blood cytopenia, including aplastic anemia, cytopenia of undetermined significance, as well as unexplained anemia of the elderly. Summary The available evidence has been proving the utility of somatic mutational analysis in patients with unexplained cytopenia, as well as in those receiving a diagnosis of myeloid neoplasm, enabling more accurate diagnosis, risk assessment, effective therapeutic strategies and residual disease monitoring. The access to a minimally invasive assessment is paving the way for screening programs of clonal hematopoiesis in individuals with absent or mild hematologic phenotype, as well as for therapeutic targeting of preleukemia cells.
- Published
- 2021
80. Deep deconvolution of the hematopoietic stem cell regulatory microenvironment reveals a high degree of specialization and conservation between mouse and human
- Author
-
Diego Alignani, Gabriele Todisco, Marta Miñana Barrios, Jesper Tegnér, Amaia Vilas, Ignacio González, Nuria Planell, Miren Lasaga, Juan P. Romero, Ana C. Vinado, Xabier Martinez-de-Morentin, Felipe Prosper, Isabel A. Calvo, Delia Quilez Agreda, Jin Ye, Borja Saez, David Gomez-Cabrero, Luca Malcovati, Bruno Paiva, Itziar Cenzano, and Patxi San Martin-Uriz
- Subjects
Haematopoiesis ,medicine.anatomical_structure ,Mesenchymal stem cell ,Specialization (functional) ,Cell ,medicine ,RNA ,Hematopoietic stem cell ,Computational biology ,Biology ,Intermediate cell - Abstract
Understanding the regulation of normal and malignant human hematopoiesis requires comprehensive cell atlas of the hematopoietic stem cell (HSC) regulatory microenvironment. Here, we develop a tailored bioinformatic pipeline to integrate public and proprietary single-cell RNA sequencing (scRNA-seq) datasets. As a result, we robustly identify for the first time 14 intermediate cell states and 11 stages of differentiation in the endothelial and mesenchymal BM compartments, respectively. Our data provide the most comprehensive description to date of the murine HSC-regulatory microenvironment and suggests a higher level of specialization of the cellular circuits than previously anticipated. Furthermore, this deep characterization allows to infer conserved features in human, suggesting that the layers of microenvironmental regulation of hematopoiesis may also be shared between species. Our resource and methodology are a steppingstone towards a comprehensive cell atlas of the BM microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
81. ZBTB33 is mutated in clonal hematopoiesis and myelodysplastic syndromes and impacts RNA splicing
- Author
-
Ruth J. F. Loos, Monkol Lek, Elli Papaemmanuil, Amaro Taylor-Weiner, Benjamin L. Ebert, Gad Getz, Björn Nilsson, Michael H. Cho, Elsa Bernard, Abhishek Niroula, Luca Malcovati, Monica Schenone, Pinkal Desai, Mendy Miller, Edyta Malolepsza, Daniel G. MacArthur, Joshua S. Weinstock, Waihay J. Wong, Anna Gallì, Kasper Lage, Marie McConkey, Maria Creignou, Andrew Dunford, Gabriele Todisco, Eva Hellström-Lindberg, Bruce M. Psaty, Shankara Anand, Chip Stewart, Akansha Tarun, Andrew D. Johnson, Benjamin Tanenbaum, Ellen M. Beauchamp, Cecilia A. Castellano, Caroline Stanclift, Emma R. Hoppe, Alexander G. Bick, Matthew Leventhal, Timothy Wood, Siddhartha Jaiswal, Alex Barbera, Robert K. Bradley, Donna Neuberg, and Steven A. Carr
- Subjects
Genetics ,Myelodysplastic syndromes ,Intron ,General Medicine ,Biology ,medicine.disease ,Article ,Haematopoiesis ,DNA methylation ,RNA splicing ,medicine ,Progenitor cell ,Stem cell ,Gene - Abstract
Clonal hematopoiesis results from somatic mutations in cancer driver genes in hematopoietic stem cells. We sought to identify novel drivers of clonal expansion using an unbiased analysis of sequencing data from 84,683 persons and identified common mutations in the 5-methylcytosine reader, ZBTB33, as well as in YLPM1, SRCAP, and ZNF318. We also identified these mutations at low frequency in myelodysplastic syndrome patients. Zbtb33 edited mouse hematopoietic stem and progenitor cells exhibited a competitive advantage in vivo and increased genome-wide intron retention. ZBTB33 mutations potentially link DNA methylation and RNA splicing, the two most commonly mutated pathways in clonal hematopoiesis and MDS.
- Published
- 2021
82. The journey of a thousand miles begins with 1 step
- Author
-
Luca Malcovati
- Subjects
Cloning ,Genetics ,Transplantation ,business.industry ,Immunology ,Myeloproliferative disease ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Congenital tufting enteropathy ,Telomere ,Chromatin ,Chronic traumatic encephalopathy ,Mutation (genetic algorithm) ,Medicine ,business ,Gene - Abstract
Germline pathogenic TERT variants are associated with short telomeres and an increased risk of developing myelodysplastic syndrome (MDS) among patients with a telomere biology disorder. We identified TERT rare variants in 41 of 1514 MDS patients (2.7%) without a clinical diagnosis of a telomere biology disorder who underwent allogeneic transplantation. Patients with a TERT rare variant had shorter telomere length (P < .001) and younger age at MDS diagnosis (52 vs 59 years, P = .03) than patients without a TERT rare variant. In multivariable models, TERT rare variants were associated with inferior overall survival (P = .034) driven by an increased incidence of nonrelapse mortality (NRM; P = .015). Death from a noninfectious pulmonary cause was more frequent among patients with a TERT rare variant. Most variants were missense substitutions and classified as variants of unknown significance. Therefore, we cloned all rare missense variants and quantified their impact on telomere elongation in a cell-based assay. We found that 90% of TERT rare variants had severe or intermediate impairment in their capacity to elongate telomeres. Using a homology model of human TERT bound to the shelterin protein TPP1, we inferred that TERT rare variants disrupt domain-specific functions, including catalysis, protein–RNA interactions, and recruitment to telomeres. Our results indicate that the contribution of TERT rare variants to MDS pathogenesis and NRM risk is underrecognized. Routine screening for TERT rare variants in MDS patients regardless of age or clinical suspicion may identify clinically inapparent telomere biology disorders and improve transplant outcomes through risk-adapted approaches.
- Published
- 2021
83. Relationship between clone metrics and clinical outcome in clonal cytopenia
- Author
-
Chiara Elena, Cinzia Sala, Martina Sarchi, Anna Gallì, Clara Camaschella, Mario Cazzola, Daniela Toniolo, Nicolas Fiorelli, Ettore Rizzo, Emanuela Boveri, Virginia Valeria Ferretti, Paolo Gasparini, Luca Malcovati, Sara Pozzi, Silvia Zibellini, Eulalia Catamo, Elisa Bono, Gabriele Todisco, Jacqueline Ferrari, Elisabetta Molteni, Galli, A., Todisco, G., Catamo, E., Sala, C., Elena, C., Pozzi, S., Bono, E., Ferretti, V. V., Rizzo, E., Molteni, E., Zibellini, S., Sarchi, M., Boveri, E., Ferrari, J., Fiorelli, N., Camaschella, C., Gasparini, P., Toniolo, D., Cazzola, M., and Malcovati, L.
- Subjects
Adult ,Male ,Myeloid ,Immunology ,Clone (cell biology) ,clonal cytopenia ,Biology ,Biochemistry ,Myeloid Neoplasm ,DNA Methyltransferase 3A ,Cohort Studies ,Young Adult ,Gene Frequency ,medicine ,Humans ,Expressivity (genetics) ,Aged ,Aged, 80 and over ,Cytopenia ,clone metrics ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Phenotype ,medicine.anatomical_structure ,Dysplasia ,Mutation (genetic algorithm) ,Mutation ,clone metric ,Female ,Clonal Hematopoiesis - Abstract
Clonal cytopenia of undetermined significance (CCUS) is associated with an increased risk of developing a myeloid neoplasm with myelodysplasia (MN). To identify the features of the mutant clone(s) that is associated with clinical phenotype and progression, we studied the following cohorts of individuals: 311 patients with idiopathic cytopenia of undetermined significance (ICUS), 532 community-dwelling individuals without hematologic phenotype (n = 355) or with unexplained anemia (n = 177), and 592 patients with overt MN. Ninety-two of 311 (30%) patients with ICUS carried a somatic genetic lesion that signaled CCUS. Clonal hematopoiesis (CH) was detected in 19.7% and 27.7% of nonanemic and anemic community-dwelling individuals, respectively. Different mutation patterns and variant allele frequencies (VAFs) (clone metrics parameters) were observed in the conditions studied. Recurrent mutation patterns exhibited different VAFs associated with marrow dysplasia (0.17-0.48), indicating variable clinical expressivity of mutant clones. Unsupervised clustering analysis based on mutation profiles identified 2 major clusters, characterized by isolated DNMT3A mutations (CH-like cluster) or combinatorial mutation patterns (MN-like cluster), and showing different overall survival (HR, 1.8). In patients with CCUS, the 2 clusters had different risk of progression to MN (HR, 2.7). Within the MN-like cluster, distinct subsets with different risk of progression to MN were identified based on clone metrics. These findings unveil marked variability in the clinical expressivity of myeloid driver genes and underline the limitations of morphologic dysplasia for clinical staging of mutant hematopoietic clones. Clone metrics appears to be critical for informing clinical decision-making in patients with clonal cytopenia.
- Published
- 2021
84. Multicenter Next-Generation Sequencing Studies between Theory and Practice
- Author
-
Pierre Fenaux, Joop H. Jansen, Eva Hellström-Lindberg, Olivier Kosmider, Emmanuelle Clappier, Luca Malcovati, María Díez-Campelo, Bert A. van der Reijden, Magnus Tobiasson, Sarah Sandmann, María Abáigar, Anna Gallì, Aniek O. de Graaf, Martin Dugas, and Michaela Fontenay
- Subjects
0301 basic medicine ,Relation (database) ,medicine.diagnostic_test ,Computer science ,business.industry ,Sequencing data ,Harmonization ,Context (language use) ,Computational biology ,DNA sequencing ,3. Good health ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Region of interest ,030220 oncology & carcinogenesis ,medicine ,Molecular Medicine ,Personalized medicine ,business ,Genetic testing - Abstract
In the age of personalized medicine, genetic testing by means of targeted sequencing has taken a key role. However, when comparing different sets of targeted sequencing data, these are often characterized by a considerable lack of harmonization. Laboratories follow their own best practices, analyzing their own target regions. The question on how to best integrate data from different sites remains unanswered. Studying the example of myelodysplastic syndrome (MDS), we analyzed 11 targeted sequencing sets, collected from six different centers (n = 831). An intersecting target region of 43,076 bp (30 genes) was identified; whereas, the original target regions covered up to 499,097 bp (117 genes). Considering a region of interest in the context of MDS, a target region of 55,969 bp (31 genes) was identified. For each gene, coverage and sequencing data quality was evaluated, calculating a sequencing score. Analyses revealed huge differences between different data sets as well as between different genes. Analysis of the relation between sequencing score and mutation frequency in MDS revealed that most genes with high frequency in MDS could be sequenced without expecting low coverage or quality. Still, no gene appeared consistently unproblematic for all data sets. To allow for comparable results in a multicenter setting analyzing MDS, we propose to use a predefined target region of interest and to perform centralized data analysis using harmonized criteria.
- Published
- 2021
- Full Text
- View/download PDF
85. Cytogenetic and molecular aberrations and worse outcome for male patients in systemic mastocytosis
- Author
-
Vito Sabato, Jens Panse, Michael Doubek, Akif Selim Yavuz, Marek Niedoszytko, Knut Brockow, Peter Valent, Michel Arock, Olivier Hermine, Sabine Müller, Roberta Zanotti, Juliana Schwaab, Luca Malcovati, Julien Rossignol, Madlen Jentzsch, Massimo Triggiani, Andreas Reiter, Nikolas von Bubnoff, Chiara Elena, Hanneke C. Kluin-Nelemans, Roberta Parente, Hans Hägglund, Judit Várkonyi, Patrizia Bonadonna, Karin Hartmann, Anna Belloni Fortina, Bjorn van Anrooij, Magdalena Lange, Anja Illerhaus, Khalid Shoumariyeh, Aleksandra Górska, Luigi Scaffidi, Michael Kundi, Hanneke Oude Elberink, Alexander Zink, Irena Angelova-Fischer, Vanessa E Kennedy, Mattias Mattsson, Tanja Schug, David Fuchs, Wolfgang R. Sperr, Anne Simonowski, Jason Gotlib, Mohamad Jawhar, Francesca Caroppo, Cecelia Perkins, Christine Breynaert, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Male ,Survival ,Gastrointestinal Diseases ,Medicine (miscellaneous) ,Leukemia, Mast-Cell ,Research & Experimental Medicine ,Gastroenterology ,cytogenetics ,0302 clinical medicine ,Neoplasm ,Systemic mastocytosis ,Child ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Aged, 80 and over ,Serine-Arginine Splicing Factors ,CLONAL HEMATOPOIESIS ,Hematology ,Middle Aged ,Prognosis ,Progression-Free Survival ,ddc ,PREVALENCE ,3. Good health ,Survival Rate ,Leukemia, Myeloid, Acute ,Proto-Oncogene Proteins c-kit ,Leukemia ,Medicine, Research & Experimental ,Child, Preschool ,Hematologic Neoplasms ,030220 oncology & carcinogenesis ,Core Binding Factor Alpha 2 Subunit ,MAST-CELLS ,Female ,medicine.symptom ,Life Sciences & Biomedicine ,Mastocytosis ,molecular mutations ,Research Paper ,Hepatomegaly ,Male predominance ,Adult ,medicine.medical_specialty ,Cytogenetics ,Molecular mutations ,Sex difference ,Adolescent ,sex difference ,Skin Diseases ,survival ,CLASSIFICATION ,Organomegaly ,Young Adult ,03 medical and health sciences ,Sex Factors ,Mastocytosis, Systemic ,Internal medicine ,medicine ,Humans ,Hematologi ,Aged ,Chromosome Aberrations ,Science & Technology ,MUTATIONS ,business.industry ,Disease progression ,Infant, Newborn ,Infant ,ADULTS ,medicine.disease ,Repressor Proteins ,Male patient ,Myelodysplastic Syndromes ,Splenomegaly ,Human medicine ,business ,LEUKEMIA ,030215 immunology - Abstract
Theranostics 11(1), 292-303 (2021). doi:10.7150/thno.51872, Published by Ivyspring, Wyoming, NSW
- Published
- 2021
- Full Text
- View/download PDF
86. Peripheral blood cytopenias in the aging general population and risk of incident hematological disease and mortality
- Author
-
Aniek O. de Graaf, Isabelle A van Zeventer, Gerwin Huls, Edo Vellenga, Jan Jacob Schuringa, Melanie M. van der Klauw, Joop H. Jansen, Bert A. van der Reijden, Arjan Diepstra, Luca Malcovati, Life Course Epidemiology (LCE), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Stem Cell Aging Leukemia and Lymphoma (SALL)
- Subjects
medicine.medical_specialty ,Aging ,Neutropenia ,Anemia ,Clinical Trials and Observations ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Population ,Context (language use) ,UNDETERMINED SIGNIFICANCE ICUS ,HEMOGLOBIN CONCENTRATION ,All institutes and research themes of the Radboud University Medical Center ,AGE ,Internal medicine ,hemic and lymphatic diseases ,Medicine ,Humans ,Cumulative incidence ,education ,CLINICAL-SIGNIFICANCE ,Aged ,MILD ANEMIA ,Cytopenia ,education.field_of_study ,business.industry ,Hazard ratio ,CLONAL HEMATOPOIESIS ,Hematology ,PLATELET COUNT ,medicine.disease ,Hematologic Diseases ,Thrombocytopenia ,PREVALENCE ,Cohort ,IDIOPATHIC CYTOPENIAS ,SEX ,business - Abstract
Peripheral blood cytopenias may precede the development of hematological malignancies and frequently pose clinical challenges in the older population. The natural course of (mild) cytopenias during aging and their association with hematological disorders in community-dwelling individuals are not well studied. Within the population-based Lifelines cohort (n = 167729), we studied changes in peripheral blood counts, occurrence of cytopenias, and associated hematological outcomes in the context of aging. Development of hematological malignancies and (cause-specific) mortality were evaluated by linkage to nationwide registries. Anemia and thrombocytopenia emerged with older age, in line with a general age-related decline in these blood counts. For neutropenia, no increase in prevalence with older age was observed. Using standard reference limits to define cytopenias, anemia (hazard ratio [HR], 1.84; 95% confidence interval [CI], 1.59-2.12), thrombocytopenia (HR, 1.58; 95% CI, 1.32-1.89), and, especially the concomitant presence of anemia and thrombocytopenia (HR, 4.75; 95% CI, 2.98-7.55) were associated with inferior overall survival. Only a minor proportion of deaths was explained by diagnosed hematological malignancies, with the majority attributable to other causes. Neutropenia, either isolated (HR, 0.88; 95% CI, 0.73-1.06) or combined with another cytopenia, did not affect overall survival. For individuals aged ≥60 years, 5-year cumulative incidence of hematological malignancies was 0.60% (95% CI, 0.50-0.70), with higher incidences among those with anemia (P < .001) or thrombocytopenia (P < .001) but not neutropenia (P = .201). Highest cumulative incidences of diagnoses and mortality from hematological malignancies were observed in individuals with >1 cytopenia. We conclude that anemia and thrombocytopenia, but not neutropenia, are associated with inferior overall survival of community-dwelling individuals. Hematological malignancies develop in a small fraction of these cases.
- Published
- 2021
87. A predictive algorithm using clinical and laboratory parameters may assist in ruling out and in diagnosing MDS
- Author
-
Ioannis Kotsianidis, Theo de Witte, Bander Abu Shrkihe, Mette Holm, Pierre Fenaux, Corine van Marrewijk, Albert Kolomansky, Guillermo Sanz, Eva Hellström-Lindberg, Alexandra Smith, Jaroslav Cermak, Dominic Culligan, Argiris Symeonidis, Juliet Mills, David T. Bowen, Saskia Langemeijer, Ulrich Germing, Moshe Mittelman, Simon Crouch, Shoham Baruch, Ge Yu, Howard S. Oster, Laurence Sanhes, Reinhard Stauder, Agnès Guerci-Bresler, Luca Malcovati, Shachar Naor, Krzysztof Madry, and Jonathan Ben-Ezra
- Subjects
Oncology ,medicine.medical_specialty ,Anemia ,SCORING SYSTEM ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,METABOLISM ,VALIDATION ,Cancer development and immune defence Radboud Institute for Health Sciences [Radboudumc 2] ,chemistry.chemical_compound ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,CELLULARITY ,Bone Marrow Diseases ,Mean corpuscular volume ,Myelodysplastic Syndromes/diagnosis ,Creatinine ,Myeloid Neoplasia ,Receiver operating characteristic ,medicine.diagnostic_test ,business.industry ,BONE-MARROW EXAMINATION ,UNEXPLAINED CYTOPENIAS ,EPICARDIAL POTENTIALS ,Bone Marrow Examination ,PRIMARY MYELODYSPLASTIC SYNDROMES ,CLONAL HEMATOPOIESIS ,LOCALIZATION ,Hematology ,medicine.disease ,Predictive value ,Peripheral blood ,Confidence interval ,medicine.anatomical_structure ,chemistry ,Myelodysplastic Syndromes ,Bone marrow ,Laboratories ,business ,Algorithms - Abstract
Contains fulltext : 237720.pdf (Publisher’s version ) (Open Access) We present a noninvasive Web-based app to help exclude or diagnose myelodysplastic syndrome (MDS), a bone marrow (BM) disorder with cytopenias and leukemic risk, diagnosed by BM examination. A sample of 502 MDS patients from the European MDS (EUMDS) registry (n > 2600) was combined with 502 controls (all BM proven). Gradient-boosted models (GBMs) were used to predict/exclude MDS using demographic, clinical, and laboratory variables. Area under the receiver operating characteristic curve (AUC), sensitivity, and specificity were used to evaluate the models, and performance was validated using 100 times fivefold cross-validation. Model stability was assessed by repeating its fit using different randomly chosen groups of 502 EUMDS cases. AUC was 0.96 (95% confidence interval, 0.95-0.97). MDS is predicted/excluded accurately in 86% of patients with unexplained anemia. A GBM score (range, 0-1) of less than 0.68 (GBM < 0.68) resulted in a negative predictive value of 0.94, that is, MDS was excluded. GBM ≥ 0.82 provided a positive predictive value of 0.88, that is, MDS. The diagnosis of the remaining patients (0.68 ≤ GBM < 0.82) is indeterminate. The discriminating variables: age, sex, hemoglobin, white blood cells, platelets, mean corpuscular volume, neutrophils, monocytes, glucose, and creatinine. A Web-based app was developed; physicians could use it to exclude or predict MDS noninvasively in most patients without a BM examination. Future work will add peripheral blood cytogenetics/genetics, EUMDS-based prospective validation, and prognostication.
- Published
- 2021
- Full Text
- View/download PDF
88. Atomic-level mechanism of Pre-mRNA splicing in health and disease
- Author
-
Lorenzo Casalino, Jure Borišek, Suzanne G. Mays, Andrea Saltalamacchia, Alessandra Magistrato, and Luca Malcovati
- Subjects
Therapeutic gene modulation ,Spliceosome ,RNA Splicing ,Antineoplastic Agents ,Computational biology ,Saccharomyces cerevisiae ,Biology ,Molecular Dynamics Simulation ,Proteòmica ,010402 general chemistry ,01 natural sciences ,Transcriptome ,Neoplasms ,Gene expression ,RNA Precursors ,Humans ,Empalmament (Genètica) ,Salut ,RNA, Messenger ,010405 organic chemistry ,Ribozyme ,General Medicine ,General Chemistry ,Group II intron ,Expressió gènica ,0104 chemical sciences ,RNA splicing ,Proteome ,biology.protein ,Spliceosomes ,Malalties ,Quantum Theory ,Proteïnes - Abstract
Intron removal from premature-mRNA (pre-mRNA splicing) is an essential part of gene expression and regulation that is required for the production of mature, protein-coding mRNA. The spliceosome (SPL), a majestic machine composed of five small nuclear RNAs and hundreds of proteins, behaves as an eminent transcriptome tailor, efficiently performing splicing as a protein-directed metallo-ribozyme. To select and excise long and diverse intronic sequences with single-nucleotide precision, the SPL undergoes a continuous compositional and conformational remodeling, forming eight distinct complexes throughout each splicing cycle. Splicing fidelity is of paramount importance to preserve the integrity of the proteome. Mutations in splicing factors can severely compromise the accuracy of this machinery, leading to aberrant splicing and altered gene expression. Decades of biochemical and genetic studies have provided insights into the SPL's composition and function, but its complexity and plasticity have prevented an in-depth mechanistic understanding. Single-particle cryogenic electron microscopy techniques have ushered in a new era for comprehending eukaryotic gene regulation, providing several near-atomic resolution structures of the SPL from yeast and humans. Nevertheless, these structures represent isolated snapshots of the splicing process and are insufficient to exhaustively assess the function of each SPL component and to unravel particular facets of the splicing mechanism in a dynamic environment.In this Account, building upon our contributions in this field, we discuss the role of biomolecular simulations in uncovering the mechanistic intricacies of eukaryotic splicing in health and disease. Specifically, we showcase previous applications to illustrate the role of atomic-level simulations in elucidating the function of specific proteins involved in the architectural reorganization of the SPL along the splicing cycle. Moreover, molecular dynamics applications have uniquely contributed to decrypting the channels of communication required for critical functional transitions of the SPL assemblies. They have also shed light on the role of carcinogenic mutations in the faithful selection of key intronic regions and the molecular mechanism of splicing modulators. Additionally, we emphasize the role of quantum-classical molecular dynamics in unraveling the chemical details of pre-mRNA cleavage in the SPL and in its evolutionary ancestors, group II intron ribozymes. We discuss methodological pitfalls of multiscale calculations currently used to dissect the splicing mechanism, presenting future challenges in this field. The results highlight how atomic-level simulations can enrich the interpretation of experimental results. We envision that the synergy between computational and experimental approaches will aid in developing innovative therapeutic strategies and revolutionary gene modulation tools to fight the over 200 human diseases associated with splicing misregulation, including cancer and neurodegeneration. J.B. thanks the Slovenian Research Agency (P1-0017 and Z1-1855), and A.M. is thankful for the financial support of the Italian Association for Cancer Research (IG 24514) and of the project “Against bRain cancEr: finding personalized therapies with in Silico and in vitro strategies” (ARES) CUP: D93D19000020007 POR FESR 2014 2020-1.3.b- Friuli Venezia Giulia. L.M. is thankful for the financial support of the Associazione Italiana Ricerca sul Cancro (AIRC) (International Accelerator Program #22796, 5x1000 project #21267, IG 2017 #20125). S.G.M. is supported by the European Union’s H2020 research and innovation programme under Marie Skłodowska-Curie Grant Agreement No. 754422
- Published
- 2021
89. Deep Deconvolution of the Hematopoietic Stem Cell Regulatory Microenvironment Reveals a High Degree of Specialization and Conservation Between Mouse and Human
- Author
-
Jin Ye, Isabel A. Calvo, Itziar Cenzano, Amaia Vilas-Zornoza, Xavier Martinez-de-Morentin, Miren Lasaga, Diego Alignani, Bruno Pavia, Ana C. Vinado, Patxi San Martin-Uriz, Juan Pablo Romero, Delia Quilez-Agreda, Marta Minana-Barrios, Ignacio Sancho-Gonzalez, Gabriele Todisco, Luca Malcovati, Nuria Planell, Borja Saez, Jesper Tegnér, Felipe Prosper, and David Gomez-Cabrero
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2021
- Full Text
- View/download PDF
90. Pseudouridine-modified tRNA fragments repress aberrant protein synthesis and predict leukaemic progression in myelodysplastic syndrome
- Author
-
Nicola Guzzi, Sowndarya Muthukumar, Maciej Cieśla, Gabriele Todisco, Phuong Cao Thi Ngoc, Magdalena Madej, Roberto Munita, Serena Fazio, Simon Ekström, Teresa Mortera-Blanco, Monika Jansson, Yasuhito Nannya, Mario Cazzola, Seishi Ogawa, Luca Malcovati, Eva Hellström-Lindberg, Marios Dimitriou, and Cristian Bellodi
- Subjects
Leukemia, Myeloid, Acute ,RNA, Transfer ,Peptide Initiation Factors ,Myelodysplastic Syndromes ,Humans ,RNA-Binding Proteins ,Cell Biology ,Hematopoietic Stem Cells ,Pseudouridine - Abstract
Transfer RNA-derived fragments (tRFs) are emerging small noncoding RNAs that, although commonly altered in cancer, have poorly defined roles in tumorigenesis1. Here we show that pseudouridylation (Ψ) of a stem cell-enriched tRF subtype2, mini tRFs containing a 5′ terminal oligoguanine (mTOG), selectively inhibits aberrant protein synthesis programmes, thereby promoting engraftment and differentiation of haematopoietic stem and progenitor cells (HSPCs) in patients with myelodysplastic syndrome (MDS). Building on evidence that mTOG-Ψ targets polyadenylate-binding protein cytoplasmic 1 (PABPC1), we employed isotope exchange proteomics to reveal critical interactions between mTOG and functional RNA-recognition motif (RRM) domains of PABPC1. Mechanistically, this hinders the recruitment of translational co-activator PABPC1-interacting protein 1 (PAIP1)3 and strongly represses the translation of transcripts sharing pyrimidine-enriched sequences (PES) at the 5′ untranslated region (UTR), including 5′ terminal oligopyrimidine tracts (TOP) that encode protein machinery components and are frequently altered in cancer4. Significantly, mTOG dysregulation leads to aberrantly increased translation of 5′ PES messenger RNA (mRNA) in malignant MDS-HSPCs and is clinically associated with leukaemic transformation and reduced patient survival. These findings define a critical role for tRFs and Ψ in difficult-to-treat subsets of MDS characterized by high risk of progression to acute myeloid leukaemia (AML).
- Published
- 2020
91. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes
- Author
-
Elsa Bernard, Yasuhito Nannya, Robert P. Hasserjian, Sean M. Devlin, Heinz Tuechler, Juan S. Medina-Martinez, Tetsuichi Yoshizato, Yusuke Shiozawa, Ryunosuke Saiki, Luca Malcovati, Max F. Levine, Juan E. Arango, Yangyu Zhou, Francesc Solé, Catherine A. Cargo, Detlef Haase, Maria Creignou, Ulrich Germing, Yanming Zhang, Gunes Gundem, Araxe Sarian, Arjan A. van de Loosdrecht, Martin Jädersten, Magnus Tobiasson, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Ronald F. Pinheiro, Valeria Santini, Ioannis Kotsianidis, Jacqueline Boultwood, Fabio P. S. Santos, Julie Schanz, Senji Kasahara, Takayuki Ishikawa, Hisashi Tsurumi, Akifumi Takaori-Kondo, Toru Kiguchi, Chantana Polprasert, John M. Bennett, Virginia M. Klimek, Michael R. Savona, Monika Belickova, Christina Ganster, Laura Palomo, Guillermo Sanz, Lionel Ades, Matteo Giovanni Della Porta, Harold K. Elias, Alexandra G. Smith, Yesenia Werner, Minal Patel, Agnès Viale, Katelynd Vanness, Donna S. Neuberg, Kristen E. Stevenson, Kamal Menghrajani, Kelly L. Bolton, Pierre Fenaux, Andrea Pellagatti, Uwe Platzbecker, Michael Heuser, Peter Valent, Shigeru Chiba, Yasushi Miyazaki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Yoshiko Atsuta, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Peter L. Greenberg, Mario Cazzola, Eva Hellström-Lindberg, Seishi Ogawa, Elli Papaemmanuil, Bernard E., Nannya Y., Hasserjian R.P., Devlin S.M., Tuechler H., Medina-Martinez J.S., Yoshizato T., Shiozawa Y., Saiki R., Malcovati L., Levine M.F., Arango J.E., Zhou Y., Sole F., Cargo C.A., Haase D., Creignou M., Germing U., Zhang Y., Gundem G., Sarian A., van de Loosdrecht A.A., Jadersten M., Tobiasson M., Kosmider O., Follo M.Y., Thol F., Pinheiro R.F., Santini V., Kotsianidis I., Boultwood J., Santos F.P.S., Schanz J., Kasahara S., Ishikawa T., Tsurumi H., Takaori-Kondo A., Kiguchi T., Polprasert C., Bennett J.M., Klimek V.M., Savona M.R., Belickova M., Ganster C., Palomo L., Sanz G., Ades L., Della Porta M.G., Smith A.G., Werner Y., Patel M., Viale A., Vanness K., Neuberg D.S., Stevenson K.E., Menghrajani K., Bolton K.L., Fenaux P., Pellagatti A., Platzbecker U., Heuser M., Valent P., Chiba S., Miyazaki Y., Finelli C., Voso M.T., Shih L.-Y., Fontenay M., Jansen J.H., Cervera J., Atsuta Y., Gattermann N., Ebert B.L., Bejar R., Greenberg P.L., Cazzola M., Hellstrom-Lindberg E., Ogawa S., Papaemmanuil E., Hematology, and CCA - Cancer biology and immunology
- Subjects
Male ,0301 basic medicine ,Oncology ,endocrine system diseases ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,DNA Mutational Analysis ,Loss of Heterozygosity ,Medical and Health Sciences ,Cohort Studies ,Loss of heterozygosity ,0302 clinical medicine ,Gene Frequency ,2.1 Biological and endogenous factors ,Medicine ,Aetiology ,Cancer ,DNA Copy Number Variation ,Allele ,0303 health sciences ,Myeloid leukemia ,Hematology ,General Medicine ,Prognosis ,3. Good health ,Phenotype ,Treatment Outcome ,International Prognostic Scoring System ,030220 oncology & carcinogenesis ,Cohort ,Female ,Survival Analysi ,Human ,medicine.medical_specialty ,DNA Copy Number Variations ,Prognosi ,Immunology ,Myelodysplastic Syndrome ,Locus (genetics) ,Article ,Genomic Instability ,General Biochemistry, Genetics and Molecular Biology ,DNA Mutational Analysi ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Complex Karyotype ,Genetics ,Humans ,Clinical significance ,Allele frequency ,neoplasms ,Gene ,Alleles ,Survival analysis ,030304 developmental biology ,business.industry ,Myelodysplastic syndromes ,Stem Cell Research ,Settore MED/15 ,medicine.disease ,Survival Analysis ,030104 developmental biology ,Myelodysplastic Syndromes ,Mutation ,Cohort Studie ,Tumor Suppressor Protein p53 ,TP53 mutations, myelodyspastic syndromes, prognosis, lenalidomide ,business - Abstract
Tumor protein p53 (TP53) is the most frequently mutated gene in cancer1,2. In patients with myelodysplastic syndromes (MDS), TP53 mutations are associated with high-risk disease3,4, rapid transformation to acute myeloid leukemia (AML)5, resistance to conventional therapies6–8 and dismal outcomes9. Consistent with the tumor-suppressive role of TP53, patients harbor both mono- and biallelic mutations10. However, the biological and clinical implications of TP53 allelic state have not been fully investigated in MDS or any other cancer type. We analyzed 3,324 patients with MDS for TP53 mutations and allelic imbalances and delineated two subsets of patients with distinct phenotypes and outcomes. One-third of TP53-mutated patients had monoallelic mutations whereas two-thirds had multiple hits (multi-hit) consistent with biallelic targeting. Established associations with complex karyotype, few co-occurring mutations, high-risk presentation and poor outcomes were specific to multi-hit patients only. TP53 multi-hit state predicted risk of death and leukemic transformation independently of the Revised International Prognostic Scoring System (IPSS-R)11. Surprisingly, monoallelic patients did not differ from TP53 wild-type patients in outcomes and response to therapy. This study shows that consideration of TP53 allelic state is critical for diagnostic and prognostic precision in MDS as well as in future correlative studies of treatment response. Clinical sequencing across a large prospective cohort of patients with myelodysplasic syndrome uncovers distinct associations between the mono- and biallelic states of TP53 and clinical presentation
- Published
- 2020
- Full Text
- View/download PDF
92. Multicenter Next-Generation Sequencing Studies between Theory and Practice: Harmonization of Data Analysis Using Real-World Myelodysplastic Syndrome Data
- Author
-
Sarah, Sandmann, Aniek O, de Graaf, Magnus, Tobiasson, Olivier, Kosmider, María, Abáigar, Emmanuelle, Clappier, Anna, Gallì, Bert A, van der Reijden, Luca, Malcovati, Pierre, Fenaux, María, Díez-Campelo, Michaela, Fontenay, Eva, Hellström-Lindberg, Joop H, Jansen, and Martin, Dugas
- Subjects
Gene Frequency ,Data Interpretation, Statistical ,Myelodysplastic Syndromes ,Mutation ,High-Throughput Nucleotide Sequencing ,Humans ,Reproducibility of Results ,Genetic Testing ,Sensitivity and Specificity ,Algorithms ,Alleles ,Biomarkers - Abstract
In the age of personalized medicine, genetic testing by means of targeted sequencing has taken a key role. However, when comparing different sets of targeted sequencing data, these are often characterized by a considerable lack of harmonization. Laboratories follow their own best practices, analyzing their own target regions. The question on how to best integrate data from different sites remains unanswered. Studying the example of myelodysplastic syndrome (MDS), we analyzed 11 targeted sequencing sets, collected from six different centers (n = 831). An intersecting target region of 43,076 bp (30 genes) was identified; whereas, the original target regions covered up to 499,097 bp (117 genes). Considering a region of interest in the context of MDS, a target region of 55,969 bp (31 genes) was identified. For each gene, coverage and sequencing data quality was evaluated, calculating a sequencing score. Analyses revealed huge differences between different data sets as well as between different genes. Analysis of the relation between sequencing score and mutation frequency in MDS revealed that most genes with high frequency in MDS could be sequenced without expecting low coverage or quality. Still, no gene appeared consistently unproblematic for all data sets. To allow for comparable results in a multicenter setting analyzing MDS, we propose to use a predefined target region of interest and to perform centralized data analysis using harmonized criteria.
- Published
- 2020
93. Combined loss of function of two different loci of miR-15/16 drives the pathogenesis of acute myeloid leukemia
- Author
-
Luca Malcovati, Michael A. Caligiuri, Mark D. Minden, Anna Gallì, Narmin Ibrahimova, Rosario Distefano, Alexey Palamarchuk, Francesca Lovat, Pierluigi Gasparini, Silvia Catricalà, Tatsuya Nakamura, Paolo Fadda, Giovanni Nigita, Carlo M. Croce, and Luisa Tomasello
- Subjects
Male ,Disease ,Biology ,Pathogenesis ,Cohort Studies ,hemic and lymphatic diseases ,microRNA ,medicine ,Gene silencing ,Humans ,neoplasms ,Loss function ,Aged ,Aged, 80 and over ,Multidisciplinary ,Gene Expression Regulation, Leukemic ,Myelodysplastic syndromes ,Myeloid leukemia ,Methylation ,Middle Aged ,Biological Sciences ,medicine.disease ,Leukemia, Myeloid, Acute ,MicroRNAs ,Cancer research ,Disease Progression ,Female - Abstract
Double knockout of the two miR-15/16 loci in mouse resulted in the development of acute myeloid leukemia (AML). This result suggested that, at least, a fraction of human AMLs could be due to a similar mechanism. We analyzed the role of the two miR-15/16 clusters in 93 myelodysplastic syndrome (MDS) patients divided in three subgroups: patients with MDS, patients with MDS before transforming into AML (MDS-T), and patients with AML evolving from MDS (MDS-AML). Then, we tested 139 AML cases and 14 different AML cell lines by assessing microRNA (miRNA) expression, target protein expression, genetic loss, and silencing. MDS-T and MDS-AML patients show a reduction of the expression of miR-15a/-15b/-16 compared to MDS patients. Each miRNA can significantly predict MDS and MDS-T groups. Then, 79% of primary AMLs show a reduced expression of miR-15a and/or miR-15b. The expression of miR-15a/-15b/-16 significantly stratified AML patients in two prognostic classes. Furthermore, 40% of AML cell lines showed a combined loss of the expression of miR-15a/-15b and overexpression of their direct/indirect targets. As potential mechanisms involved in the silencing of the two miR-15/16 loci, we identified a genetic loss of miR-15a and miR-15b and silencing of these two loci by methylation. We identified a potential driver oncogenic role in the loss of expression of both miR-15/16 clusters in the progression of MDS into AML and in AML pathogenesis. The stratification of AML patients, based on miR-15/16 expression, can lead to targeted and combination therapies for the treatment of this incurable disease.
- Published
- 2020
94. SF3B1-mutant MDS as a distinct disease subtype: a proposal from the International Working Group for the Prognosis of MDS
- Author
-
Jacqueline Boultwood, Manja Meggendorfer, Seishi Ogawa, Donna Neuberg, Luca Malcovati, David T. Bowen, Felicitas Thol, Michael Heuser, Elli Papaemmanuil, David P. Steensma, Michaela Fontenay, Kristen E. Stevenson, Mario Cazzola, David A. Sallman, Heinz Tüchler, Mikkael A. Sekeres, Michael R. Savona, Rami S. Komrokji, Detlef Haase, Sudhir Tauro, Andrea Pellagatti, Torsten Haferlach, Peter J. Campbell, Pierre Fenaux, Rafael Bejar, Benjamin L. Ebert, Paresh Vyas, Matthew J. Walter, Joop H. Jansen, Aly Karsan, Timothy A. Graubert, Peter L. Greenberg, Jaroslaw P. Maciejewski, Arjan A. van de Loosdrecht, Eva Hellström-Lindberg, Guillermo Garcia-Manero, Hematology, and CCA - Cancer biology and immunology
- Subjects
Ineffective erythropoiesis ,Oncology ,medicine.medical_specialty ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Plenary Paper ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,Internal medicine ,Molecular genetics ,hemic and lymphatic diseases ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,Cytopenia ,business.industry ,Leukemia, Myelomonocytic, Chronic ,Cell Biology ,Hematology ,medicine.disease ,Phosphoproteins ,Prognosis ,3. Good health ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Dysplasia ,030220 oncology & carcinogenesis ,Concomitant ,Mutation (genetic algorithm) ,Erythropoiesis ,Bone marrow ,RNA Splicing Factors ,business - Abstract
The 2016 revision of the World Health Organization classification of tumors of hematopoietic and lymphoid tissues is characterized by a closer integration of morphology and molecular genetics. Notwithstanding, the myelodysplastic syndrome (MDS) with isolated del(5q) remains so far the only MDS subtype defined by a genetic abnormality. Approximately half of MDS patients carry somatic mutations in spliceosome genes, with SF3B1 being the most commonly mutated one. SF3B1 mutation identifies a condition characterized by ring sideroblasts (RS), ineffective erythropoiesis, and indolent clinical course. A large body of evidence supports recognition of SF3B1-mutant MDS as a distinct nosologic entity. To further validate this notion, we interrogated the data set of the International Working Group for the Prognosis of MDS (IWG-PM). Based on the findings of our analyses, we propose the following diagnostic criteria for SF3B1-mutant MDS: (1) cytopenia as defined by standard hematologic values, (2) somatic SF3B1 mutation, (3) morphologic dysplasia (with or without RS), and (4) bone marrow blasts
- Published
- 2020
- Full Text
- View/download PDF
95. The Data Registry of the European Competence Network on Mastocytosis (ECNM): Set Up, Projects, and Perspectives
- Author
-
Magdalena Lange, Jens Panse, Karoline V. Gleixner, Massimo Triggiani, Elisabeth Aberer, Andreas Reiter, Nikolas von Bubnoff, Roberta Parente, Haifa Kathrin Al-Ali, Knut Brockow, Serena Merante, Olivier Hermine, Akif Selim Yavuz, Patrizia Bonadonna, Chiara Elena, Anja Illerhaus, Olivier Lortholary, Marek Niedoszytko, Hans Hägglund, Bjorn van Anrooij, David Fuchs, Hanneke C. Kluin-Nelemans, Dietger Niederwieser, Emir Hadzijusufovic, Luca Malcovati, Wolfgang R. Sperr, Marie-Anne Morren, Jason Gotlib, Michael Doubek, Mattias Mattsson, Francesca Caroppo, Alexander Zink, Rosemarie Greul, Cecelia Perkins, Vanessa E Kennedy, Massimiliano Bonifacio, Mohamad Jawhar, Joanna N G Oude Elberink, Peter Valent, Judit Várkonyi, Roberta Zanotti, Michel Arock, Anna Belloni Fortina, Khalid Shoumariyeh, Aleksandra Górska, Juliana Schwaab, Karin Hartmann, Vito Sabato, and Study Grp European Competence
- Subjects
Risk ,Pediatrics ,medicine.medical_specialty ,Prognostic variable ,Diagnostic criteria ,DISORDERS ,DIAGNOSTIC-CRITERIA ,International Cooperation ,Mast cell activation syndrome ,Disease ,World Health Organization ,CLASSIFICATION ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Immunology and Allergy ,Registries ,030212 general & internal medicine ,Precision Medicine ,Systemic mastocytosis ,Competence (human resources) ,Mastocytosis ,Prognosis ,Therapy ,WHO classification ,Information Services ,MUTATIONS ,business.industry ,Cutaneous Mastocytosis ,Network on ,KIT ,SYSTEMIC MASTOCYTOSIS ,ADULTS ,Mast cell leukemia ,medicine.disease ,DELINEATION ,Europe ,030228 respiratory system ,MAST-CELLS ,Human medicine ,medicine.symptom ,business ,CELL ACTIVATION SYNDROMES - Abstract
Mastocytosis is a unique hematologic neoplasm with complex biology and pathology and a variable clinical course. The disease can essentially be divided into cutaneous mastocytosis (CM) and systemic mastocytosis (SM). In adults, SM is diagnosed in most cases and manifests as either indolent or advanced disease. Patients with advanced SM have an unfavorable prognosis with reduced survival. However, so far, little is known about the prevalence of various categories of SM and about prognostic factors. In an attempt to learn more about the behavior and evolution of various forms of CM and SM, the European Competence Network on Mastocytosis (ECNM) initiated a mastocytosis registry in 2012. In this article, the set up and start phase of this registry are described. Until 2018, more than 3000 patients from 12 countries and 25 centers have been enrolled. In a majority of all patients, robust follow-up data and relevant clinical end points are available. Using this data set, a series of registry projects have been launched, with the aim to validate previously identified diagnostic and prognostic variables and to identify new disease-related and patient-related parameters in various forms of mastocytosis. Moreover, the core data set of the registry will be useful to establish multiparametric scoring systems through which prognostication and individualized management of patients with mastocytosis should improve in the foreseeable future. (C) 2019 American Academy of Allergy, Asthma & Immunology
- Published
- 2019
- Full Text
- View/download PDF
96. Impact of spliceosome mutations on RNA splicing in myelodysplasia: dysregulated genes/pathways and clinical associations
- Author
-
Jacqueline Boultwood, Emmanouela Repapi, Christopher J. Smith, Richard N. Armstrong, Eva Hellström-Lindberg, Thomas Luft, Sally Killick, Stephen S. Taylor, Marco L. Hennrich, Anne-Claude Gavin, Eshita Sharma, Helen Lockstone, Luca Malcovati, Andrea Sanchi, Angela Hamblin, Paresh Vyas, Andrea Pellagatti, Anna Schuh, Hamid Dolatshad, Stephen Smith, Violetta Steeples, Aleksandar Radujkovic, Patrick Horn, Mario Cazzola, John Broxholme, Swagata Roy, Anthony D. Ho, Elli Papaemmanuil, Shalini Singh, Aristoteles Giagounidis, Pellagatti, Andrea [0000-0002-6122-0221], Armstrong, Richard N [0000-0001-7744-5890], Boultwood, Jacqueline [0000-0002-4330-2928], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Spliceosome ,Splicing Factor U2AF/genetics ,DNA Repair ,RNA Splicing ,Immunology ,Serine-Arginine Splicing Factors/genetics ,Biology ,medicine.disease_cause ,Biochemistry ,Cohort Studies ,03 medical and health sciences ,Splicing factor ,Phosphoproteins/genetics ,hemic and lymphatic diseases ,medicine ,Humans ,RNA Splicing Factors/genetics ,Spliceosomes/genetics ,Gene ,Myelodysplastic Syndromes/epidemiology/genetics ,Regulation of gene expression ,Gene knockdown ,Mutation ,Serine-Arginine Splicing Factors ,Cell Biology ,Hematology ,Phosphoproteins ,Splicing Factor U2AF ,Survival Analysis ,Phenotype ,030104 developmental biology ,Gene Expression Regulation ,Myelodysplastic Syndromes ,RNA splicing ,Spliceosomes ,Cancer research ,RNA Splicing Factors - Abstract
SF3B1, SRSF2, and U2AF1 are the most frequently mutated splicing factor genes in the myelodysplastic syndromes (MDS). We have performed a comprehensive and systematic analysis to determine the effect of these commonly mutated splicing factors on pre-mRNA splicing in the bone marrow stem/progenitor cells and in the erythroid and myeloid precursors in splicing factor mutant MDS. Using RNA-seq, we determined the aberrantly spliced genes and dysregulated pathways in CD34+ cells of 84 patients with MDS. Splicing factor mutations result in different alterations in splicing and largely affect different genes, but these converge in common dysregulated pathways and cellular processes, focused on RNA splicing, protein synthesis, and mitochondrial dysfunction, suggesting common mechanisms of action in MDS. Many of these dysregulated pathways and cellular processes can be linked to the known disease pathophysiology associated with splicing factor mutations in MDS, whereas several others have not been previously associated with MDS, such as sirtuin signaling. We identified aberrantly spliced events associated with clinical variables, and isoforms that independently predict survival in MDS and implicate dysregulation of focal adhesion and extracellular exosomes as drivers of poor survival. Aberrantly spliced genes and dysregulated pathways were identified in the MDS-affected lineages in splicing factor mutant MDS. Functional studies demonstrated that knockdown of the mitosis regulators SEPT2 and AKAP8, aberrantly spliced target genes of SF3B1 and SRSF2 mutations, respectively, led to impaired erythroid cell growth and differentiation. This study illuminates the effect of the common spliceosome mutations on the MDS phenotype and provides novel insights into disease pathophysiology.
- Published
- 2018
- Full Text
- View/download PDF
97. Shiozawa Y, Malcovati L, Gallì A, et al. Gene expression and risk of leukemic transformation in myelodysplasia. Blood. 2017;130(24):2642-2653
- Author
-
LUCA MALCOVATI
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2018
- Full Text
- View/download PDF
98. Autoantibodies Against Type I IFNs in Patients with Ph-Negative Myeloproliferative Neoplasms
- Author
-
Emanuela Sant’Antonio, Alessandro Borghesi, Ilaria Carola Casetti, Luca Malcovati, Paul Bastard, Virginia Valeria Ferretti, Adrian Gervais, Elisa Rumi, Jean-Laurent Casanova, Luca Arcaini, Daniele Vanni, Jérémie Rosain, Oscar Borsani, Chiara Trotti, and Daniela Pietra
- Subjects
business.industry ,Immunology ,Ph Negative ,Autoantibody ,Medicine ,In patient ,Cell Biology ,Hematology ,business ,Biochemistry ,631.Myeloproliferative Syndromes and Chronic Myeloid Leukemia: Basic and Translational - Abstract
Background. The classic Ph-negative myeloproliferative neoplasms (MPN) are a group of clonal haematopoietic disorders, including polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis (MF), whose shared and diverse phenotypic signatures are caused by a dysregulated JAK/STAT signal transduction because of acquired somatic mutations. It has been demonstrated that autoimmune diseases and MPN can be associated (Kristinsson et al., Haematologica. 2010 Jul;95(7):1216-20.), suggesting a common background of immune dysregulation (Barosi, Curr Hematol Malig Rep. 2014 Dec;9(4):331-9). SARS-CoV-2 infection displays extreme inter-individual clinical variability, ranging from silent infection to lethal disease. It has been described that at least 10% of patients with life-threatening coronavirus disease 2019 (COVID-19) pneumonia have neutralizing autoantibodies (AAbs) against type I IFNs, that precede SARS-CoV-2 infection (Bastard et al., Science. 2020 Oct 23;370(6515):eabd4585). In this study we searched for AAbs against type I IFNs in a cohort of MPN patients to evaluate the prevalence of these AAbs in the MPN population and to check for clinical correlations, including severity of COVID-19. Methods. Plasma samples from consecutively referred MPN patients were prospectively collected between November 2020 and June 2021 and frozen at -30°C immediately after collection. Levels of AAbs against type I IFN subtypes including IFNs alpha, beta and omega were measured using the enzyme-linked immunosorbent assay (ELISA) and a neutralization assay, as previously reported (Bastard et al., Science. 2020 Oct 23;370(6515):eabd4585; Moreews et al., Sci Immunol. 2021 May 25;6(59):eabh1516). Results. We included a total of 219 MPN patients (101 ET, 76 PV, 36 MF and 6 MPN unclassificable). Neutralizing AAbs to type I IFNs were detected in 29 patients (13.2%, 95%CI: 9.1% - 18.5%). Comparing patients with and without AAbs we observed a significant difference in terms of distribution of MPN diagnosis (P = 0.029) and driver mutations (P = 0.019), while we did not observe a difference in terms of age, sex, and treatment (Table 1). Overall, 29 patients (13%) got SARS-CoV-2 infection and 8 of them (28%) required hospitalization due to severe COVID-19. AAbs against type I IFNs were detected in 4 of the 29 SARS-CoV-2 infected patients. A higher rate of hospitalization for severe COVID-19 was observed in patients with AAbs to type I IFNs (2 of 4 patients, 50%) compared to those without these AAbs (6 of 25 patients, 24%), although the difference did not reach a statistical significance (P = 0.300). Conclusions. In this study, we detected a prevalence of AAbs against type I IFNs which is much higher in our MPN cohort (13%) than in the general population (2-3%). We also found a correlation between the presence of AAbs to type I IFNs and both the hematological diagnosis and the driver mutation. Despite a comparable prevalence of SARS-CoV-2 infection between MPN patients with or without AAbs to type I IFNs, we observed a different rate of hospitalization due to severe COVID-19 which is almost twice in those with AAbs against type I IFNs compared to those without these AAbs. However, this difference did not reach a statistical significance, probably because of the low number of SARS-CoV-2 infection in the subgroup of patients with AAbs against type I IFNs. Thus, further studies to analyse the prevalence of AAbs against type I IFNs in patients with MPN, their association with other forms of auto-immunity and severe COVID-19 are warranted. Figure 1 Figure 1. Disclosures Arcaini: Gilead Sciences: Research Funding; Bayer, Celgene, Gilead Sciences, Roche, Sandoz, Janssen-Cilag, VERASTEM: Consultancy; Celgene, Roche, Janssen-Cilag, Gilead: Other: Travel expenses; Celgene: Speakers Bureau. Rumi: Novartis, Abbvie: Consultancy.
- Published
- 2021
99. Modeling Clonal Progression in SF3B1-Mutant Myelodysplastic Syndrome
- Author
-
Luca Malcovati, Gabriele Todisco, Sergei Doulatov, J. Philip Creamer, Martina Sarchi, Anna Gallì, and Courtnee Clough
- Subjects
hemic and lymphatic diseases ,Immunology ,Mutant ,Cancer research ,Cell Biology ,Hematology ,Biology ,Biochemistry - Abstract
SF3B1-mutant myelodysplastic syndrome (MDS) has recently been proposed as a distinct disorder characterized by ring sideroblasts, ineffective erythropoiesis and good prognosis. Selected co-occurring genetic abnormalities were reported associated with significantly worse outcome and suggested as exclusion criteria for the proposed entity. However, it remains unclear how a limited spectrum of co-occurring drivers affects SF3B1-mutant MDS biology to determine evolution from a relatively indolent condition to high risk malignancy. To gain a better insight into the clonal progression of SF3B1-mutant MDS, we analyzed SF3B1 co-mutations in a cohort of 176 SF3B1-mutated patients diagnosed with a myeloid neoplasm. RUNX1 and STAG2 were the only co-mutated genes found significantly associated with advanced disease phenotype (i.e. MDS with excess blasts and secondary acute myeloid leukemia) (OR=18.36 (2.18-862.91), P=0.001, and OR= Inf (3.57-Inf), P RUNX1 and STAG2 control hematopoietic stem and progenitor cell self-renewal and differentiation by regulation of gene expression. To explore the biological impact of RUNX1 or STAG2 loss in the context of SF3B1-mutant MDS, we disrupted RUNX1 or STAG2 using CRISPR/Cas9 in an induced pluripotent stem cell (iPSC) model of MDS-RS that we previously established by reprogramming of bone marrow cells from SF3B1-mutant individuals. This patient-derived system allows hematopoietic progenitor expansion through doxycycline-mediated expression of 5 transcription factors (5F-HPC) and multilineage differentiation upon doxycycline withdrawal. We asked how RUNX1 or STAG2 disruption affected SF3B1-mutant HPCs self-renewal and differentiation in our model of MDS-RS, and sought to define the underlying transcriptional changes. RUNX1-edited cells maintained significantly higher proportion of CD34 + HPCs, suggesting that RUNX1 mutation increases self-renewal capacity of SF3B1-mutant progenitors. Consistently, SF3B1/RUNX1 double-mutant 5F-HPCs showed positive enrichment of HSC-specific gene signatures. This was accompanied by broad upregulation of inflammatory programs, recapitulating the activated gene signatures we identified in SF3B1/RUNX1 co-mutated patients in our cohort. RUNX1 disruption promoted myeloid skewing at the expense of erythroid differentiation in SF3B1-mutant cells. Consistent with this, granulocyte-monocyte progenitors (GMP) and myelo-erythroid transcriptional programs were positively and negatively enriched, respectively. By contrast, in SF3B1/STAG2 double-mutant 5F-HPCs, both erythroid and myeloid populations were reduced compared to SF3B1 single mutant controls, suggesting a block in both myeloid and erythroid differentiation, despite the presence of pro-differentiation signals. This was supported by a profound down-regulation of genes involved in the response to external stimulus and suppression of GMP specific transcriptional signature. In summary, we identified RUNX1 and STAG2 mutations as main drivers of disease progression in SF3B1-mutant patients, and generated extensive cell line panels to interrogate their functional interaction with mutant SF3B1. By applying CRISPR/Cas9 editing to our MDS-RS model, we could overcome limited availability of primary MDS samples to show that RUNX1 or STAG2 co-mutations drive progression through distinct biological mechanisms in SF3B1-mutant HPCs. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
100. Management of MDS with Isolated Del(5q) Patients in the European MDS (EUMDS) Registry, a Unique Prospective Real-World Dataset
- Author
-
Moshe Mittelman, Reinhard Stauder, Luca Malcovati, Jaroslav Cermak, Raphael Itzykson, Ulrich Germing, Guillermo Sanz, Agnès Guerci-Bresler, Saskia Langemeijer, Ioannis Kotsianidis, Mette Holm, Pierre Fenaux, Howard S. Oster, Eva Hellstrom Lindberg, Alexandra Smith, David G. Bowen, Corine van Marrewijk, Theo de Witte, Adele Taylor, Dominic Culligan, Argiris Symeonidis, and Krzysztof Madry
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
'MDS with isolated del(5q)' is a subtype of Myelodysplastic Syndrome (MDS) with characteristic morphological and genomic features. Clinically, MDS with isolated del(5q) is uniquely responsive to immunomodulatory agents, with lenalidomide (LEN) approved for treating anemia in this indication. Erythropoiesis Stimulating Agents (ESA) and red cell transfusion (RBCT) are other options for anemia management. The EUMDS Registry is an international prospective registry of newly diagnosed patients (within 100 days of diagnosis) with IPSS Low/Int-1-risk MDS (plus higher-risk MDS from 2017) recruiting since 1 st April 2008, currently at 146 sites in 17 countries. We describe the largest prospective Registry series of 'MDS with del(5q)' patients, with data from diagnosis. Methods Comparative analyses of baseline characteristics for categorical variables compared different groups by chi 2 test, and for continuous variables by Kruskal-Wallis test. p values are given with Bonferonni correction applied. Results Baseline characteristics Of 2469 EUMDS patients, 197 (8%) met the WHO 2016 classification criteria for 'MDS with isolated del5q'. Median follow up is 4.3 yrs. 77% were female (34% for all EUMDS patients), and median age was 73y. In comparison to EUMDS patients lacking del(5q), patients with MDS with isolated del(5q) were lower risk at diagnosis (IPSS and IPSS-R, p Interventional management To date, 168/197 patients received active intervention and the following sequence of interventions was observed, in order of frequency of initial intervention, RBCT>ESA>LEN: RBCT (n=96), - then ESA (42) then LEN (21) - then LEN (32) then ESA (5) ESA (n=55), - then RBCT (30) then LEN (13) - then LEN (9) then RBCT (6) LEN (n=17), - then ESA (3) then RBCT (2) - then RBCT (2) then ESA (2) Ninety-two patients have received LEN. In comparison with those not (yet) treated with LEN, LEN treated patients were younger (mean 70y vs 75y, p Adverse events were considered but proved too difficult to evaluate from the Registry data. For example, anticoagulant treatment data was collected, but not the specific indication. Outcome No difference in outcome (OS or PFS) was noted for patients treated with LEN vs. not (Figure, OS - adjusted Hazard Ratio, age and sex, 95% Confidence Interval: 0.67 [95% CI:0.35-1.29], lenalidomide treatment as a time-dependent variable). Conclusion EUMDS is uniquely placed to describe 'real-world' management of patients with 'MDS with isolated del(5q)' from diagnosis. To date, the largest (retrospective) case series reported had a median age 5 years younger than EUMDS, most likely representing referral bias(1). EUMDS demonstrates that most patients with MDS with isolated del(5q) require interventional therapy, and that more than half of transfusion dependent patients have received LEN. Further analysis of interventions and response will be presented. 1. Germing U, Lauseker M, Hildebrandt B, Symeonidis A, Cermak J, Fenaux P, et al. Survival, prognostic factors and rates of leukemic transformation in 381 untreated patients with MDS and del(5q): A multicenter study. Leukemia 2012;26(6):1286-92. Figure 1 Figure 1. Disclosures Fenaux: Takeda: Honoraria, Research Funding; Abbvie: Honoraria, Research Funding; JAZZ: Honoraria, Research Funding; Celgene/BMS: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Syros Pharmaceuticals: Honoraria. Symeonidis: Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GSK: Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; MSD: Consultancy, Research Funding; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Demo: Research Funding; WinMedica: Research Funding; Astellas: Consultancy, Research Funding; Sanofi/Genzyme: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Kotsianidis: Novartis Hellas: Consultancy, Other: NONE, Research Funding, Speakers Bureau; Abbvie: Consultancy, Other: NONE, Research Funding, Speakers Bureau; Bristol Hellas: Consultancy, Other: NONE, Research Funding, Speakers Bureau; Janssen Hellas: Consultancy, Other: NONE, Speakers Bureau; Genesis: Consultancy, Other: NONE; Astellas: Other: NONE, Research Funding, Speakers Bureau. Mittelman: Janssen · Roche · Novartis · Takeda · Medison / Amgen · Neopharm / Celgene / BMS · Abbvie · Gilead: Research Funding; Novartis · Takeda · Fibrogen · Celgene / BMS · Onconova · Geron: Other: Clini; Onconova · Novartis · Takeda · Silence: Membership on an entity's Board of Directors or advisory committees; MDS HUB: Consultancy; Celgene / BMS · Novartis: Speakers Bureau. Sanz: Helsinn Healthcare: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Roche: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, and expenses; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Boehringer Ingelheim: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, and expenses, Speakers Bureau; Gilead Sciences: Other: Travel, accommodations, and expenses; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, and expenses, Research Funding. Stauder: Celgene/BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel support; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees. Germing: Janssen: Honoraria; Celgene: Honoraria; Novartis: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Other: advisory activity, Research Funding; Jazz Pharmaceuticals: Honoraria. van Marrewijk: EUMDS Registry: Current Employment; Novartis: Other: Educational Grants; BV Oncology Europe: Other: Educational Grants; Amgen Limited: Other: Educational Grants; Celgene International: Other: educational grants; Janssen Pharmaceuticals: Other: educational grants; Takeda Pharmaceuticals: Other: educational grants; MDS-RIGHT: Other: project budgets. de Witte: Amgen: Research Funding; Celgene: Research Funding; Takeda: Research Funding; Novartis: Research Funding; Janssen: Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.