Classification and Personalized Prognostic Assessment on the Basis of Clinical and Genomic Features in Myelodysplastic Syndromes

Matteo Bersanelli, Erica Travaglino, Manja Meggendorfer, Tommaso Matteuzzi, Claudia Sala, Ettore Mosca, Chiara Chiereghin, Noemi Di Nanni, Matteo Gnocchi, Matteo Zampini, Marianna Rossi, Giulia Maggioni, Alberto Termanini, Emanuele Angelucci, Massimo Bernardi, Lorenza Borin, Benedetto Bruno, Francesca Bonifazi, Valeria Santini, Andrea Bacigalupo, Maria Teresa Voso, Esther Oliva, Marta Riva, Marta Ubezio, Lucio Morabito, Alessia Campagna, Claudia Saitta, Victor Savevski, Enrico Giampieri, Daniel Remondini, Francesco Passamonti, Fabio Ciceri, Niccolò Bolli, Alessandro Rambaldi, Wolfgang Kern, Shahram Kordasti, Francesc Sole, Laura Palomo, Guillermo Sanz, Armando Santoro, Uwe Platzbecker, Pierre Fenaux, Luciano Milanesi, Torsten Haferlach, Gastone Castellani, Matteo G Della Porta, Matteo Bersanelli, Erica Travaglino, Manja Meggendorfer, Tommaso Matteuzzi, Claudia Sala, Ettore Mosca, Chiara Chiereghin, Noemi Di Nanni, Matteo Gnocchi, Matteo Zampini, Marianna Rossi, Giulia Maggioni, Alberto Termanini, Emanuele Angelucci, Massimo Bernardi, Lorenza Borin, Benedetto Bruno, Francesca Bonifazi, Valeria Santini, Andrea Bacigalupo, Maria Teresa Voso, Esther Oliva, Marta Riva, Marta Ubezio, Lucio Morabito, Alessia Campagna, Claudia Saitta, Victor Savevski, Enrico Giampieri, Daniel Remondini, Francesco Passamonti, Fabio Ciceri, Niccolò Bolli, Alessandro Rambaldi, Wolfgang Kern, Shahram Kordasti, Francesc Sole, Laura Palomo, Guillermo Sanz, Armando Santoro, Uwe Platzbecker, Pierre Fenaux, Luciano Milanesi, Torsten Haferlach, Gastone Castellani, Matteo G Della Porta

Abstract

Purpose: Recurrently mutated genes and chromosomal abnormalities have been identified in myelodysplastic syndromes (MDS). We aim to integrate these genomic features into disease classification and prognostication.

Methods: We retrospectively enrolled 2,043 patients. Using Bayesian networks and Dirichlet processes, we combined mutations in 47 genes with cytogenetic abnormalities to identify genetic associations and subgroups. Random-effects Cox proportional hazards multistate modeling was used for developing prognostic models. An independent validation on 318 cases was performed.

Results: We identify eight MDS groups (clusters) according to specific genomic features. In five groups, dominant genomic features include splicing gene mutations (SF3B1, SRSF2, and U2AF1) that occur early in disease history, determine specific phenotypes, and drive disease evolution. These groups display different prognosis (groups with SF3B1 mutations being associated with better survival). Specific co-mutation patterns account for clinical heterogeneity within SF3B1- and SRSF2-related MDS. MDS with complex karyotype and/or TP53 gene abnormalities and MDS with acute leukemia-like mutations show poorest prognosis. MDS with 5q deletion are clustered into two distinct groups according to the number of mutated genes and/or presence of TP53 mutations. By integrating 63 clinical and genomic variables, we define a novel prognostic model that generates personally tailored predictions of survival. The predicted and observed outcomes correlate well in internal cross-validation and in an independent external cohort. This model substantially improves predictive accuracy of currently available prognostic tools. We have created a Web portal that allows outcome predictions to be generated for user-defined constellations of genomic and clinical features.

Conclusion: Genomic landscape in MDS reveals distinct subgroups associated with specific clinical features and discrete patterns of evolution, providing a proof of concept for next-generation disease classification and prognosis.

Conflict of interest statement

Manja MeggendorferEmployment: MLL Munich Leukemia Laboratory Marianna RossiConsulting or Advisory Role: Pfizer, Celgene, IQvia, Janssen Emanuele AngelucciHonoraria: Celgene, Vertex Pharmaceuticals Incorporated (MA) and CRISPR Therapeutics AG (CH)Consulting or Advisory Role: Novartis, Bluebird BioTravel, Accommodations, Expenses: Janssen-Cilag Massimo BernardiHonoraria: CelgeneConsulting or Advisory Role: PfizerTravel, Accommodations, Expenses: Medac, Amgen, Sanofi, Jazz Pharmaceuticals, BioTest, Abbvie, Takeda Lorenza BorinLeadership: CelgeneSpeakers' Bureau: GenzymeTravel, Accommodations, Expenses: Genzyme Benedetto BrunoHonoraria: Jazz Pharmaceuticals, Novartis, AmgenResearch Funding: Amgen Valeria SantiniHonoraria: Celgene/Bristol-Myers Squibb, Novartis, Janssen-CilagConsulting or Advisory Role: Celgene/Bristol-Myers Squibb, Novartis, Menarini, Takeda, PfizerResearch Funding: CelgeneTravel, Accommodations, Expenses: Janssen-Cilag, Celgene Andrea BacigalupoHonoraria: Pfizer, Therakos, Novartis, Sanofi, Jazz Pharmaceuticals, Riemser, Merck Sharp & Dohme, Janssen-Cilag, Gilead Sciences, Kiadis Pharma, Astellas PharmaConsulting or Advisory Role: Novartis, Kiadis Pharma, Gilead Sciences, Astellas PharmaSpeakers' Bureau: Pfizer, Therakos, Novartis, Sanofi, Riemser, Merck Sharp & Dohme, Adienne, Jazz PharmaceuticalsTravel, Accommodations, Expenses: Sanofi, Therakos, Jazz Pharmaceuticals Maria Teresa VosoHonoraria: Celgene/Jazz, AbbvieConsulting or Advisory Role: Celgene/JazzSpeakers' Bureau: CelgeneResearch Funding: Celgene Esther OlivaHonoraria: Celgene, Novartis, Amgen, Alexion PharmaceuticalsConsulting or Advisory Role: Amgen, Celgene, NovartisSpeakers' Bureau: Celgene, NovartisPatents, Royalties, Other Intellectual Property: Royalties for QOL-E instrument Francesco PassamontiSpeakers' Bureau: Novartis, AOP Orphan Pharmaceuticals Niccolò BolliConsulting or Advisory Role: JanssenSpeakers' Bureau: Celgene, Amgen Alessandro RambaldiHonoraria: Amgen, OmerosConsulting or Advisory Role: Amgen, Omeros, Novartis, Astellas Pharma, Jazz PharmaceuticalsTravel, Accommodations, Expenses: Celgene Wolfgang KernEmployment: MLL Munich Leukemia LaboratoryLeadership: MLL Munich Leukemia LaboratoryStock and Other Ownership Interests: MLL Munich Leukemia Laboratory Shahram KordastiHonoraria: Beckman Coulter, GWT-TUD, Alexion PharmaceuticalsConsulting or Advisory Role: Syneos HealthResearch Funding: Celgene, Novartis Guillermo SanzHonoraria: CelgeneConsulting or Advisory Role: Abbvie, Celgene, Helsinn Healthcare, Janssen, Roche, Amgen, Boehringer Ingelheim, Novartis, TakedaSpeakers' Bureau: TakedaResearch Funding: CelgeneTravel, Accommodations, Expenses: Celgene, Takeda, Gilead Sciences, Roche Pharma AG Armando SantoroConsulting or Advisory Role: Bristol-Myers Squibb, Servier, Gilead Sciences, Pfizer, Eisai, Bayer AG, MSD, Sanofi, ArQuleSpeakers' Bureau: Takeda, Roche, Abbvie, Amgen, Celgene, AstraZeneca, ArQule, Lilly, Sandoz, Novartis, Bristol-Myers Squibb, Servier, Gilead Sciences, Pfizer, Eisai, Bayer AG, MSD Uwe PlatzbeckerHonoraria: Celgene/JazzConsulting or Advisory Role: Celgene/JazzResearch Funding: Amgen, Janssen, Novartis, BerGenBio, CelgenePatents, Royalties, Other Intellectual Property: part of a patent for a TFR-2 antibody (Rauner et al. Nature Metabolics 2019)Travel, Accommodations, Expenses: Celgene Pierre FenauxHonoraria: CelgeneResearch Funding: Celgene Torsten HaferlachEmployment: MLL Munich Leukemia LaboratoryLeadership: MLL Munich Leukemia LaboratoryConsulting or Advisory Role: IlluminaNo other potential conflicts of interest were reported.

Figures

FIG 1.
FIG 1.
(A) Frequency of mutations and chromosomal abnormalities in the EuroMDS cohort (N = 2,043), stratified according to the type of mutation (missense, nonsense, affecting a splice site, or other). Insertions and deletions (del) were categorized according to whether they resulted in a shift in the codon reading frame (by either 1 or 2 base pairs [bp]) or were in frame. Splicing factor genes were the most frequently mutated (49%), followed by DNA methylation–related genes (37.9%), chromatin and histone modifier genes (31.3%), signaling genes (28.5%), transcription regulation genes (24%), tumor suppressor genes (11.1%), and cohesin complex genes (7.6%). (B) Frequency of recurrently mutated genes and chromosomal abnormalities in the EuroMDS cohort, broken down by MDS subtype according to 2016 WHO criteria. (C) VAF of driver mutations in the EuroMDS cohort, broken down by gene and gene function (boxplots reporting median, 25-75 percentiles, and ranges); VAF of X-linked genes (ATRX, BCOR, BCORL1, PHF6, PIGA, SMC1A, STAG2, UTX, and ZRSR2, highlighted by asterisk in the figure plot) was halved in male patients. (D) Relationship between the number of genomic abnormalities (mutations and chromosomal abnormalities) and outcome (overall survival). MDS, myelodysplastic syndromes; MDS 5q-, MDS with isolated deletion of long arm of chromosome 5; MDS-EB1, MDS with excess of blasts, type 1; MDS-EB2, MDS with excess of blasts, type 2; MDS-MLD, MDS with multilineage dysplasia; MDS-RS-MLD, MDS with ring sideroblasts and multilineage dysplasia; MDS-RS-SLD, MDS with ring sideroblasts and single-lineage dysplasia; MDS-SLD, MDS with single-lineage dysplasia; VAF, variant allele frequencies.
FIG 2.
FIG 2.
(A) Probability of overall survival after allogeneic transplantation in the EuroMDS cohort. Patients were stratified according to specific genomic features. A total of 424 cases with complete information about transplant procedures and clinical outcome entered the analysis. (B) Comparison of probability of survival among different genomic-based MDS groups (P values of log-rank test were reported). AML, acute myeloid leukemia; MDS, myelodysplastic syndromes.
FIG 3.
FIG 3.
Fraction of explained variation that was attributable to different prognostic factors for overall survival.
FIG 4.
FIG 4.
Personalized prediction of overall survival using a multistate prognostic model including clinical and genomic features and their interactions in two patients from the EuroMDS cohort (labeled as patient A and patient B), both classified as MDS with multilineage dysplasia according to 2016 WHO classification and belonging to low-risk group according to age-adjusted revised version of International Prognostic Scoring System (IPSS-R). Using currently available prognostication, both patients are predicted to have an indolent clinical course without significant risk of disease evolution and death (in the EuroMDS cohort, Kaplan-Meier curves show a median survival of 79 months for low-risk age-adjusted IPSS-R). When looking at mutational profile, driver mutations involved different splicing factor genes in these patients: patient A carries SF3B1 mutation, whereas patient B presents SRSF2 mutation. We then calculated expected survival by using the novel genomic-based prognostic model (exponential survival curves are reported in the figure). Patient A was classified into genomic-based group 6, and patient B was classified into group 5. Accordingly, the estimation of life expectancy is now significantly different in these two patients, as underlined by the slope of the two exponential curves. The model predicts a better probability of survival for patient A (with SF3B1 mutation) with respect to patient B (with SRSF2 mutation), thus reflecting more precisely the observed clinical outcome. In fact, patient B died 16 months after the diagnosis as a result of leukemic evolution, whereas patient A was still alive without evidence of disease progression after 60 months of follow-up. IPSS-R fails to capture such a difference in clinical outcome. The interpretation of the predicted survival curves by genomic-based predictive model is meaningful also considering that we are in the context of a cohort of elderly patients: patient A (age 78 years) has a 30% survival probability at the age of 80, whereas patient B (age 73 years) has a 30% survival probability at the age of 74.
FIG A1.
FIG A1.
Genomic groups in EuroMDS cohort (N = 2,043) and their relationship with WHO category (defined according to 2016 classification criteria) and overall survival. According to a Bayesian clustering algorithm (Dirichlet processes), patients are classified into eight distinct genomic groups on the basis of the presence or specific mutations and/or chromosomal abnormalities: Group 0, MDS without specific genomic profile; Group 1, MDS with SF3B1 mutations and co-existing mutations in other genes (ASXL1 and RUNX1); Group 2, MDS with TP53 mutations and/or complex karyotype; Group 3, MDS with SRSF2 and concomitant TET2 mutations; Group 4, MDS with U2AF1 mutations associated with deletion of chromosome 20q and/or abnormalities of chromosome 7; Group 5, MDS with SRSF2 mutations with co-existing mutations in other genes (ASXL1, RUNX1, IDH2, and EZH2); Group 6, MDS with isolated SF3B1 mutations (or associated with mutations of TET2 and/or JAK/STAT pathways genes); Group 7, MDS with AML-like mutation patterns (DNMT3A, NPM1, FLT3, IDH1, and RUNX1 genes). These genomic MDS groups significantly differ in WHO MDS categories distribution and in cumulative probability of survival. AML, acute myeloid leukemia; BM, bone marrow; MDS, myelodysplastic syndromes; MDS-EB1, MDS with excess of blasts, type 1; MDS-EB2, MDS with excess of blasts, type 2; MDS-MLD, MDS with multilineage dysplasia; MDS-RS-MLD, MDS with ring sideroblasts and multilineage dysplasia; MDS-RS-SLD, MDS with ring sideroblasts and single-lineage dysplasia; MDS-SLD, MDS with single-lineage dysplasia; PB, peripheral blood; WHO, World Health Organization.
FIG A2.
FIG A2.
Extrapolation of genomic landscape of MDS genomic groups through Bayesian Networks, applied to the whole MDS cohort. The size of each node accounts for the number of correspondent genomic or cytogenetic alterations. The color of each link reflects odds ratio (shades of brown represent mutual exclusivity while shades of green color degree co-occurrence). The thickness of edges grows with increasing significance of mutual exclusivity/co-occurrence between alterations. MDS, myelodysplastic syndromes.
FIG A3.
FIG A3.
Wide-ranging genomic heterogeneity of 2016 WHO categories within MDS genomic groups. MDS, myelodysplastic syndromes; MDS-EB1, MDS with excess of blasts, type 1; MDS-EB2, MDS with excess of blasts, type 2; MDS-MLD, MDS with multilineage dysplasia; MDS-RS-MLD, MDS with ring sideroblasts and multilineage dysplasia; MDS-RS-SLD, MDS with ring sideroblasts and single-lineage dysplasia; MDS-SLD, MDS with single-lineage dysplasia.
FIG A4.
FIG A4.
Diagram to correctly classify MDS patients into the appropriate genomic group according to individual profile. AML, acute myeloid leukemia; MDS, myelodysplastic syndromes.

References

    1. Adès L, Itzykson R, Fenaux P: Myelodysplastic syndromes. Lancet 383:2239-2252, 2014
    1. Arber DA Orazi A Hasserjian R, et al. : The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127:2391-2405, 2016
    1. Della Porta MG Travaglino E Boveri E, et al. : Minimal morphological criteria for defining bone marrow dysplasia: A basis for clinical implementation of WHO classification of myelodysplastic syndromes. Leukemia 29:66-75, 2015
    1. Senent L Arenillas L Luno E, et al. : Reproducibility of the World Health Organization 2008 criteria for myelodysplastic syndromes. Haematologica 98:568-575, 2013
    1. Malcovati L Della Porta MG Pascutto C, et al. : Prognostic factors and life expectancy in myelodysplastic syndromes classified according to WHO criteria: A basis for clinical decision making. J Clin Oncol 23:7594-7603, 2005
    1. Malcovati L Hellström-Lindberg E Bowen D, et al. : Diagnosis and treatment of primary myelodysplastic syndromes in adults: Recommendations from the European LeukemiaNet. Blood 122:2943-2964, 2013
    1. Greenberg PL Tuechler H Schanz J, et al. : Revised international prognostic scoring system for myelodysplastic syndromes. Blood 120:2454-2465, 2012
    1. Della Porta MG Tuechler H Malcovati L, et al. : Validation of WHO classification-based prognostic scoring system (WPSS) for myelodysplastic syndromes and comparison with the revised International Prognostic Scoring System (IPSS-R). A study of the International Working Group for prognosis in myelodysplasia. Leukemia 29:1502-1513, 2015
    1. Cazzola M, Della Porta MG, Malcovati L: The genetic basis of myelodysplasia and its clinical relevance. Blood 122:4021-4034, 2013
    1. Papaemmanuil E Cazzola M Boultwood J, et al. : Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med 365:1384-1395, 2011
    1. Yoshida K Sanada M Shiraishi Y, et al. : Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478:64-69, 2011
    1. Papaemmanuil E Gerstung M Malcovati L, et al. : Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122:3616-3627, 2013
    1. Schanz J Tüchler H Solé F, et al. : New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge. J Clin Oncol 30:820-829, 2012
    1. Papaemmanuil E Gerstung M Bullinger L, et al. : Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 374:2209-2221, 2016
    1. Gerstung M Papaemmanuil E Martincorena I, et al. : Precision oncology for acute myeloid leukemia using a knowledge bank approach. Nat Genet 49:332-340, 2017
    1. Grinfeld J Nangalia J Baxter EJ, et al. : Classification and personalized prognosis in myeloproliferative neoplasms. N Engl J Med 379:1416-1430, 2018
    1. Grinfeld J Nangalia J Baxter EJ, et al. . R package for Hierarchical Dirichlet Process.
    1. Harrell FE, Lee KL, Mark DB: Multivariable prognostic models: Issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat Med 15:361-387, 1996
    1. Perperoglou A: Cox models with dynamic ridge penalties on time-varying effects of the covariates Stat Med 33:170-180, 2014
    1. Grinfeld J Nangalia J Baxter EJ, et al. . EUROMDS Project: Personalized prediction of clinical outcome in patients with myelodysplastic syndrome according to genomic and clinical features.
    1. Pellagatti A Armstrong RN Steeples V, et al. : Impact of spliceosome mutations on RNA splicing in myelodysplasia: Dysregulated genes/pathways and clinical associations. Blood 132:1225-1240, 2018
    1. Malcovati L Papaemmanuil E Bowen DT, et al. : Clinical significance of SF3B1 mutations in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms. Blood 118:6239-6246, 2011
    1. Klampfl T Gisslinger H Harutyunyan AS, et al. : Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 369:2379-2390, 2013
    1. Reilly B Tanaka TN Diep D, et al. : DNA methylation identifies genetically and prognostically distinct subtypes of myelodysplastic syndromes. Blood Adv 3:2845-2858, 2019
    1. Masaki S Ikeda S Hata A, et al. : Myelodysplastic syndrome-associated SRSF2 mutations cause splicing changes by altering binding motif sequences. Front Genet 10:338, 2019
    1. Liang Y Tebaldi T Rejeski K, et al. : SRSF2 mutations drive oncogenesis by activating a global program of aberrant alternative splicing in hematopoietic cells. Leukemia 32:2659-2671, 2018
    1. Haferlach T Nagata Y Grossmann V, et al. : Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28:241-247, 2014
    1. Bacher U Haferlach T Schnittger S, et al. : Investigation of 305 patients with myelodysplastic syndromes and 20q deletion for associated cytogenetic and molecular genetic lesions and their prognostic impact. Br J Haematol 164:822-833, 2014
    1. Haase D Stevenson KE Neuberg D, et al. : TP53 mutation status divides myelodysplastic syndromes with complex karyotypes into distinct prognostic subgroups. Leukemia 33:1747-1758, 2019
    1. Kulasekararaj AG Jiang J Smith AE, et al. : Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome. Blood 124:2698-2704, 2014
    1. Della Porta MG Gallì A Bacigalupo A, et al. : Clinical effects of driver somatic mutations on the outcomes of patients with myelodysplastic syndromes treated with allogeneic hematopoietic stem-cell transplantation. J Clin Oncol 34:3627-3637, 2016
    1. Lindsley RC Saber W Mar BG, et al. : Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. N Engl J Med 376:536-547, 2017

Source: PubMed

3
Sottoscrivi