Accepted Manuscript Clinical Significance of DNA Variants in Chronic Myeloid Neoplasms (CMNs): A Report of the Association for Molecular Pathology Rebecca F. McClure, Mark D. Ewalt, Jennifer Crow, Robyn L. Temple-Smolkin, Mrudula Pullambhatla, Rachel Sargent, Annette S. Kim PII:
S1525-1578(17)30409-9
DOI:
10.1016/j.jmoldx.2018.07.002
Reference:
JMDI 722
To appear in:
The Journal of Molecular Diagnostics
Received Date: 22 August 2017 Revised Date:
7 June 2018
Accepted Date: 19 July 2018
Please cite this article as: McClure RF, Ewalt MD, Crow J, Temple-Smolkin RL, Pullambhatla M, Sargent R, Kim AS, Clinical Significance of DNA Variants in Chronic Myeloid Neoplasms (CMNs): A Report of the Association for Molecular Pathology, The Journal of Molecular Diagnostics (2018), doi: 10.1016/ j.jmoldx.2018.07.002. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
Clinical Significance of DNA Variants in Chronic Myeloid Neoplasms (CMNs): A Report of the Association for Molecular Pathology
RI PT
Rebecca F. McClure,*† Mark D. Ewalt,*‡ Jennifer Crow,*§ Robyn L. Temple-Smolkin,¶ Mrudula Pullambhatla,¶ Rachel Sargent,*ǁ and Annette S. Kim*,**
SC
From the The Chronic Myeloid Neoplasms Working Group of the Clinical Practice Committee,*
Association for Molecular Pathology,¶ Bethesda, Maryland; the Department of Pathology,† Health
M AN U
Sciences North/Horizon Santé-Nord, Sudbury, Ontario, Canada; the Department of Pathology,‡ University of Colorado School of Medicine, Aurora, Colorado; the Huguley Memorial Medical Center,§ Burleson, Texas; the University of Pennsylvania Perelman School of Medicine,ǁ Philadelphia, Pennsylvania; and the Department of Pathology, Brigham and Women’s Hospital, Harvard Medical
TE D
School, Boston, Massachusetts
Short running title: DNA variants in Chronic Myeloid Neoplasms
EP
The AMP 2015 -2017 Clinical Practice Committee consisted of Marina N. Nikiforova (2016 Chair), Antonia Sepulveda (2017 Chair), Monica J. Basehore, Mark Boguski, Susan Butler-Wu, Christopher
AC C
Coldren, Linda Cook, Jennifer Crow, Birgit Funke, Meera R. Hameed, Lawrence J. Jennings, Arivarasan Karunamurthy, Annette S. Kim, Bryan Krock, Mary Lowery-Nordberg, Melissa Miller, Keyur Patel, Jess Friedrich Peterson, Benjamin Pinsky, Carolyn S. Richards, Somak Roy, Mark J. Routbort, Kandelaria Rumilla, Ryan Schmidt, and David S. Viswanatha.
Footnote: R.F.McC. and M.D.E. contributed equally.
ACCEPTED MANUSCRIPT
Disclosures: A.K. received consulting or advisory fees from Aushon Biosystems, and Papgene, Inc., as well as speaker fees, consulting or advisory fees from LabCorp, Inc. M.D.E. has received speaker fees/honoraria from Invivoscribe. R.S. has received consulting or advisory fees from Janssen Global
RI PT
Services LLC and The Double Hit Lymphoma Foundation, and provided voluntary consulting or advisory services to Curis.
Standard of practice is not defined by this article and there may be alternatives. See Disclaimer for
M AN U
SC
further details.
Funding: Supported by the Association for Molecular Pathology.
Corresponding author:
TE D
Annette S. Kim, MD, PhD 75 Francis Street, Thorn 613A Boston, MA 02115
EP
United States
AC C
Email:
[email protected]
ACCEPTED MANUSCRIPT
ABSTRACT To address the clinical relevance of small DNA variants in chronic myeloid neoplasms (CMNs), an
RI PT
Association for Molecular Pathology (AMP) Working Group comprehensively reviewed published literature, summarized key findings that support clinical utility, and defined critical gene inclusions for high-throughput sequencing testing panels. This review highlights the biological complexity of CMNs (including myelodysplastic syndromes, myeloproliferative neoplasms, entities with overlapping features
SC
(myelodysplastic syndromes/myeloproliferative neoplasms), and systemic mastocytosis), the genetic heterogeneity within diagnostic categories, and similarities between apparently disparate diagnostic
M AN U
entities. The founding variant’s hematopoietic differentiation compartment, specific genes and variants present, order of variant appearance, individual subclone dynamics, and therapeutic intervention all contribute to the clinicopathologic features of CMNs. Selection and efficacy of targeted therapies are increasingly based on DNA variant profiles present at various time points; therefore, high-throughput
TE D
sequencing remains critical for patient management. The following genes are a minimum recommended list to provide relevant clinical information for the management of most CMNs: ASXL1, BCOR, BCORL1, CALR, CBL, CEBPA, CSF3R, DNMT3A, ETV6, EZH2, FLT3, IDH1, IDH2, JAK2, KIT, KRAS, MPL, NF1, NPM1,
EP
NRAS, PHF6, PPM1D, PTPN11, RAD21, RUNX1, SETBP1, SF3B1, SMC3, SRSF2, STAG2, TET2, TP53, U2AF1, and ZRSR2. This list is not comprehensive for all myeloid neoplasms and will evolve as insights into effects
AC C
of combinations of relevant biomarkers on specific clinicopathologic characteristics of CMNs accumulate.
ACCEPTED MANUSCRIPT
INTRODUCTION Chronic myeloid neoplasms (CMNs) are a heterogeneous group of hematopoietic disorders that
RI PT
are currently classified using predominantly non-specific clinicopathologic features. We use the term CMNs to include myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPNs), those myeloid neoplasms with overlapping features (MDS/MPNs), and systemic mastocytosis, thereby
distinguishing these chronic entities from acute myeloid leukemia (AML) that has been sufficiently
SC
reviewed in numerous other papers. This heterogeneity of CMNs complicates the ability to make an accurate diagnosis, provide reliable prognostic information, and select appropriate therapy. Single
M AN U
genetic variants already feature prominently in the diagnosis and management of a few MPNs, notably BCR/ABL1 in chronic myelogenous leukemia (CML), FIP1L1/PDGFRA in one type of myeloid/lymphoid neoplasm with eosinophilia, KIT variants in systemic mastocytosis (SM), and JAK2, CALR, and MPL variants in the classic MPNs, including polycythemia vera (PV), essential thrombocythemia (ET), and
TE D
primary myelofibrosis (PMF). Introduction of high-throughput sequencing technology, however, has generated a recent explosion of literature pertaining to DNA variants and their relevance with respect to diagnosis, prognosis, and the therapeutic management of CMNs. For most adult non-CML CMNs, which
EP
is the focus of this review, the cataloging of specific variants is well underway and has yielded new insights into which cellular processes are disrupted within the myeloid neoplasms as a group, and
AC C
highlighted the genomic complexity underlying current diagnostic categories. In the 2016 World Health Organization (WHO), most diagnostic categories for CMNs lack a single variant as a common driver for the group.1,2 In contrast, each neoplasm contains one or more variants in genes that have been identified as altered in myeloid neoplasms as a whole (so-called "myeloid genes"). It has recently been discovered that variants in myeloid genes can be present in hematopoietic cells of individuals showing no clinical features of a myeloid neoplasm.3–5 This phenomenon is known as clonal hematopoiesis of indeterminate potential (CHIP) in individuals who lack any evidence of a myeloid neoplasm, and clonal
ACCEPTED MANUSCRIPT
cytopenia(s) of undetermined significance (CCUS), in individuals who have one or more cytopenia but lack sufficient clinicopathologic features to fulfill WHO criteria for a myeloid neoplasm. The incidence of CHIP and CCUS increases with age and both are associated with an increased risk of a subsequent
RI PT
hematopoietic neoplasm. Although these categories are not formally recognized in the current WHO classification, they are included in this review, as they represent pre-malignant conditions similar to others that are routinely considered worthy of identification and monitoring (eg, monoclonal
SC
gammopathy of undetermined significance and clonal B-cell lymphocytosis).
Initial variant characterization studies in CMNs seemed to merely underscore the heterogeneity
M AN U
already known to exist, without providing much practical clarification as to how the variants might be integrated into clinical practice. However, recent investigations, particularly those evaluating clonal architecture in individual CMNs, have provided a more comprehensive picture of clonal evolution. These evaluations are already providing clinically useful genomic biomarker information and promise to not
TE D
only clarify the underlying biology of this heterogeneous group of neoplasms but also to allow for more accurate, and perhaps even truly individualized, diagnoses, prognoses, and therapies. The goal of the Association for Molecular Pathology (AMP) CMN Working Group is to review and
EP
summarize the current state of the literature regarding small DNA variants in adult, non-CML CMNs, and to provide education and guidance regarding the practical clinical relevance of variants in this group of
AC C
neoplasms. Although it is clear that relevant literature continues to accumulate, it is the opinion of the authors that a “current” collation of genomic biomarker information in CMNs, would be beneficial for those involved in the clinical laboratory evaluation and management of such patients.
MATERIALS AND METHODS From November 2015 through November 2016, the AMP CMN Working Group performed a review of articles (>600 published in the English language literature) pertaining to DNA variants in adult
ACCEPTED MANUSCRIPT
CMNs with a focus on small somatic variants that might have sufficient clinical relevance to be included in testing panels for the management of these neoplasms. Essentially all of the studies were retrospective association studies, not clinical trials. Diseases primarily driven by a single recurrent
RI PT
translocation were excluded from this study, such as CML and neoplasms associated with eosinophils and abnormalities of PDGFRA, PDGFRB, or FGFR1. Aplastic anemia, a disease that shows
clinicopathologic overlap with MDS, and juvenile myelomonocytic leukemia (JMML), a pediatric disease
excluded due to their distinct pathogenesis from other CMNs.
SC
that has some overlapping features with adult chronic myelomonocytic leukemia (CMML), were also
M AN U
A master database was created to relate genes with reported variants to WHO disease categories and to discrete facts felt to be of diagnostic, prognostic, or therapeutic relevance. The database matrix included the clinically relevant fact, WHO disease category, gene name, gene relevance in cell biological processes, gene associations (genes with variants identified together or appearing
TE D
mutually exclusive), specific variant when applicable, incidence (number of patients in the study with the feature being evaluated), total number of patients in the study, and data source. Only gene symbols are used in this text and readers are referred to the HUGO Gene Nomenclature Committee
EP
(http://www.genenames.org, last accessed 4/14/17) for full gene designations. A review of relevant basic cell processes, focusing on the normal role of each gene in the
AC C
database, was performed and is summarized graphically in Figures 1, 2, and 36 (also Supplemental Figures S17,8 and S29) to provide an aid for data analysis and discussion. From the database, Supplemental Tables S1, S2, S3, S4, S5.10-140 were created to summarize the most clinically relevant data in a form that could be more easily viewed. In the text, only genes that were found in greater than 10% of cases were listed, but genes mutated at lower frequencies in specific diseases are noted in the Supplemental Tables S1, S2, S3, S4, and S5 and organized by cell process. However, these latter tables are not meant to be comprehensive, and it is possible that infrequent drivers may have been missed by
ACCEPTED MANUSCRIPT
this study. Prognostic data was further analyzed to be presented as a heatmap (Figure 4) representing a synthesized, overall view of the clinical value of variants in each gene. To create the heatmap, key
RI PT
prognostic matrix points [progression, including fibrosis, transformation to acute leukemia, leukemiafree survival (LFS), and overall survival (OS)] were given a score from 1 to 3 as follows: Unclear- no convincing reports OR contradictory reports OR <5 patients reported (labeled as “unclear”
SC
prognostically); Score 1 - ≤2 papers OR multiple contradictory reports OR <25 patients reported; Score 2 - ≥2 papers AND no contradictory reports AND ≥25 patients reported; Score 3 - ≥4 papers AND no
RESULTS COMMON PATHWAYS ALTERED IN CMNs
M AN U
contradictory reports AND ≥100 patients reported.
TE D
The majority of genes commonly containing variants in CMNs may be classified into several key pathways that span the breadth of cellular functions. The four most significant pathways are: i) receptor kinase signaling transduction (Figure 1 and Supplemental Table S1); ii) transcription via transcription
EP
factors (Figure 1 and Supplemental Table S2); iii) epigenetic modification (Figure 2 and Supplemental Table S3); and iv) RNA splicing (Figure 3 and Supplemental Table S4).
AC C
Activity through these key pathways, which are all critically implicated in CMNs, regulates cell cycle progression, apoptosis, and protein degradation. Other pathways with fewer components containing gene variants are shown in Supplemental Figures S1 and S2 and Supplemental Table S5. DNA variants may occur from several types of sequence alterations (single nucleotide changes,
insertions, deletions, and inversions) resulting in a variety of mutations (missense, nonsense, frameshift, and splice site alteration) that lead to altered RNA and/or protein levels and/or functions. Variants are frequently described as showing gain of function (GOF), loss of function (LOF), or alteration of
ACCEPTED MANUSCRIPT
function (AOF), based on the final functional result for the protein encoded. Missense and in-frame insertion/deletion variants are typically associated with GOF or AOF while nonsense and frameshift variants are typically associated with LOF or AOF. In many genes (eg, CALR, JAK2, MPL, SF3B1, SRSF2,
RI PT
and U2AF1), there are variant hotspots, whereas in other genes (eg, TP53), variants may be found
throughout the gene. Variant locations for each gene and the most common corresponding changes in protein function are denoted in Supplemental Table S6. It should be noted that not all somatic variants
SC
identified in CMNs are pathogenic drivers.
M AN U
CATEGORIZATION OF VARIANTS BY WHO DISEASE CLASSIFICATIONS
DNA variants reported in WHO categories of CMNs were further cataloged based on variant diagnostic specificity, prognostic utility, and therapeutic implications (Supplemental Tables S1, S2, S3, S4, S5). Although gene sets vary between studies, there is general consensus regarding the most
TE D
commonly identified genes containing variants implicated in CMNs and those genes are included here. The broad range of reported incidences for variants in most genes likely resulted from study differences with respect to the number of cases, groupings of cases, sequencing targets (focused panels versus
EP
whole exome), sequencing analytic sensitivity, sample type, and processing. All diagnostic categories have reported variants in genes representing multiple cellular pathways. Though some variants appear
AC C
to have prognostic utility for specific categories, others show utility across many/all diagnostic categories. However, for most reported gene variants, there was either minimal or absent available prognostic information, conflicting prognostic information, or an absence of prognostic significance. Studies also varied widely with respect to which prognostic features were evaluated and the subgroupings of patients reported. The most commonly reported prognostic endpoints were risk for progression (fibrosis and/or acute leukemia, almost exclusively AML), leukemia-free survival (LFS), and overall survival (OS). Most articles reported each particular prognostic feature as a comparison between
ACCEPTED MANUSCRIPT
the diagnostic group with the variant and the diagnostic group without the variant. Unless otherwise specified, these methods of reporting were assumed for the purposes of this review. For ease of visualization this complex and often incomplete data, genes with variants reported as having prognostic
RI PT
significance were weighted for clinical relevance based on a scoring system that was designed to reflect the strength of the prognostic correlation and presented in a heatmap (Figure 4).
SC
Clonal hematopoiesis (CHIP and CCUS)
With the recent description of CHIP and CCUS,3–5,141 the list of genes that may be clinically
M AN U
relevant for myeloid neoplasms has expanded.The most common CHIP genes (accounting for 63% to 78% of somatic variants found in clonal hematopoiesis) are also commonly found in myeloid neoplasms (DNMT3A, ASXL1, TET2, JAK2, PPM1D, SF3B1, and TP53).3–5,141 These genes predominantly define the clinical implications of CHIP. In addition, clonal hematopoiesis may also be seen with a large number of
TE D
other genes less frequently, including SRSF2, TP53, CBL, U2AF1, IDH2, ATM, TET2, GNAS, GNB1, BCOR, CUX1, SETD2, SETBP1, and BCORL1. Overall, when compared with age-matched controls, the presence of clonal hematopoiesis at or greater than 2% variant allele fraction (VAF) has been associated with an
EP
increased risk of a subsequent hematopoietic neoplasm (hazard ratio 11 to 12), death (hazard ratio 1.4), and an overall rate of progression of 0.5% to 1% per year.4,142,143 Interestingly, these patients also have
AC C
an increased risk of cardiovascular disease.4,144 Highly sensitive sequencing methods, such as can be achieved by molecular barcoding, have detected very small clones sizes (VAF 0.03% t0 2%) that reveal clonal hematopoiesis may be found in 95% of 50- to 70-year–olds, although the clinical significance of these smaller clones has not been studied to date.145 Variants in spliceosome genes appear to arise almost exclusively in individuals >70 yo and these variants do not appear to provide a proliferative advantage to normal hematopoietic stem and progenitor cells.146 Studies have suggested that higher VAF (>10%), and an increased number of variants (>2), presence of spliceosome gene variants, and
ACCEPTED MANUSCRIPT
comutation patterns involving TET2, DNMT3A, or ASXL1 may identify CHIP patients at increased risk of subsequent myeloid neoplasm.147 The clinical relevance of most individual variants has not yet been
RI PT
determined due to the low frequency of any individual variant in the available studies.
Myelodysplastic syndrome (MDS)
Reported series of DNA variants in MDS typically lump together many of the WHO MDS
SC
subcategories and even include entities from the mixed MDS/MPN categories, such as CMML. Thus, conclusive data for individual MDS categories is difficult to obtain. As such, the following summarizes
M AN U
the data for the inclusive category of MDS.
Genes reported to show variants in ≥10% of cases in at least one series included: SF3B1 (10% to 33%),12,22,24,25,31,84,90,108,130-132 TET2 (13% to 37%),124 ASXL1 (5% to 46%),22-25,84,89,99–105 TP53 (5% to 18%),22,23,25,31 U2AF1 (5% to 17%),22,24,91,103,131,132,135,136 SRSF2 (12% to 33%),12,22,24,25,90-92,103,105,131,132 STAG2 (5% to 15%),24,31,138 RUNX1 (8% to 20%),22–25,83,84 DNMT3A (3% to 13%),22,24,25,84,88,89,100,101,103-105 EZH2 (3%
TE D
to 11%),22-25,84,89,100,105,112,113 ZRSR2 (3% to 11%),24,89,91,92,103,131,132,135 IDH1/2 (4% to 12%),23,24,89,104,114,115,117 cohesins as a group (8% to 15%),24,25,89,92,133,137,138 KIT (1% to 10%),24,25,31 NRAS (3% to 10%),22,23,25,26,31 and
EP
KRAS (<1% to 10%).22-25,31 Genes from all major cell process categories were represented. Although no single gene or gene category aligned exclusively with MDS or any of its subcategories, SF3B1 variants are
AC C
highly correlated with the dysplastic feature of ring sideroblasts (RS). This association is so robust that the WHO 2016 revision requires only 5% RS in the presence of a pathologic SF3B1 variant for a diagnosis of MDS-RS compared to the at least 15% RS required for cases without an SF3B1 variant.2 Variants in some genes appear to have prognostic utility for patients with MDS (Figure 4 and
Supplemental Tables S1, S2, S3, S4, and S5). Variants in the following genes are the most significant poor prognostic markers: ASXL1 (increased progression, decreased LFS and OS, poor overall),3,23-25,84,102– 104
CBL (decreased OS, poor overall),23,24,26 BCOR (decreased OS),24,105 cohesins as a group (increased
ACCEPTED MANUSCRIPT
progression, decreased OS, poor overall),24,92,133 ETV6 (decreased OS,23–25 poor overall), EZH2 (decreased OS, poor overall),22–25,84,89,105,100,112,113 FLT3 (increased progression),40,41 KRAS (increased progression, poor overall),24,41 NRAS (decreased OS, poor overall),22–25 PHF6 (poor overall),24 PPM1D (increased risk
RI PT
of therapy-related myeloid neoplasms),82 PTPN11 (decreased OS),79 RUNX1 (increased progression, decreased LFs and OS, poor overall),22–25,83 SETBP1 (increased progression, decreased OS, poor overall), 86–88
SRSF2 (increased progression, decreased LFS and OS, poor overall),22,26,91,103,132,135 STAG2 (increased
25,79,84,139,140
SC
progression, decreased OS),24,31,92,133 TP53 (increased progression, decreased OS, poor overall),22–
and U2AF1 (decreased OS, poor overall).22,26,136,137 In addition, patients with more pathogenic
M AN U
variants fair worse than patients with fewer variants.148 Only SF3B1 variants have been associated with better prognosis (increased LFS and OS so long as not associated with variants in RUNX1 or TP53),12,25,84,104,130,132 the effect being independent of the categorization of MDS-RS.12 The following genes had ambiguous data with respect to variant status and prognosis: DNMT3A,22,24,31,104,110 IDH1,23,24,100,103,105,114-118IDH2,24,105,114,115,117,118 IDH1/2,104,117 NPM1,22-24, 40 TET2,23,31,40,42,3,101,105,107,124–126 and
TE D
ZRSR2.103,135 Variants in the remaining genes either had no impact on prognosis or no data identified. Haferlach et al recently proposed a new prognostic predictor for MDS based only on the variant status
EP
of 14 myeloid genes (ASXL1, CBL, ETV6, EZH2, KRAS, LAMB4, NF1, NPM1, NRAS, PRPF8, RUNX1, STAG2, TET2, and TP53).24 This was shown to outperform the Revised International Prognostic Scoring System
AC C
(IPSS-R). In the transplant setting, TP53 (especially truncating variants) and RAS variants are associated with high rates of relapse and poor survival post transplantation.82 TP53 and PPM1D variants are commonly found in therapy-related MDS (38% and 15% of cases, respectively) and are associated with poor prognosis. In therapy-related MDS lacking a TP53 mutation, the prognosis is similar to nontherapy–related MDS.82 Specific therapeutic responses in MDS have been associated with variants in the following genes: STAG2 and RAD21 (better response to DNA methyl transferase (DNMT) inhibitors),133 SF3B1 (increased response to DNMT inhibitors),104 TET2 (increased response to DNMT inhibitors when
ACCEPTED MANUSCRIPT
ASXL1 wildtype),101 and TP53 (decreased response to lenalidomide).149 However, small molecule therapeutics that specifically target proteins that are altered due to known gene variants are not yet commonly used in MDS patients.
RI PT
Clonal architecture studies in individual MDS patients have provided interesting insights into the pathogenesis and dynamic nature of MDS clones.24,25,60,89,148,150 Mossner et al reported the
reconstruction of variant hierarchies in serial samples from 30 cases, including many pre- and post-
SC
therapy samplings.148 The results showed that each patient's clone is unique with respect to founding variants and to those appearing throughout the disease course. In all cases, variants in genes involved in
M AN U
methylation and/or splicing were identified as the earliest events in 90% of the cases. Interestingly, the remaining 10% of the cases had TP53 variants initially identified, followed by the appearance of variants in methylation (especially ASXL1, TET2, and DNMT3A) or splicing genes and/or del(5q). Variants in genes involved in signaling pathways were not identified early, but rather later in clonal progression, as was
TE D
the case for large variants, detected using classic cytogenetic techniques. This study also clearly confirmed that founding clones frequently involve lymphocytes (both B-cells and T-cells) further highlighting that clonal initiation occurs in pluripotent hematopoietic stem cells, and underscoring the
EP
importance of using non-hematopoietic cells as internal germline controls for these types of studies. This study also demonstrated that clonal progression may be linear, branching, or a combination of
AC C
both, occurring with or without therapy. Following therapy, relapses may arise from the original founding clone, from re-emergence of subclones that were insensitive to therapy, or as outgrowth of apparently new subclones. Although specific clinicopathologic features appeared to track with overall clonal burden (eg, hemoglobin, white blood cell count, platelet count) or specific subclones (eg., ring sideroblasts with SF3B1 variants), other distinct associations of clonal patterns with WHO subclassifications were not clearly identified. There was also a direct correlation between the complexity of clonal architecture and clone aggressiveness, with death frequently occurring shortly after the
ACCEPTED MANUSCRIPT
emergence of additional variant complexity. The data from this detailed clonal architecture study aligns well with reports of variant gene frequencies in MDS and apparent order of identification of variants in prior reports.24,108,132,136 Overall, the data from clonal architecture studies support the hypothesis that
RI PT
MDS likely arises from CHIP in essentially all cases and that the clinicopathologic features are dictated by the emergence of subsequent variants. Spliceosome component variants which, as mentioned above, are identified in CHIP essentially exclusively in individuals >70 years old, are also seen predominantly in
SC
MDS patients >70 years old.24,25 This suggests a specific component of age-induced development.
Spliceosome gene variants also appear associated with dysplastic features since they are enriched in
M AN U
CMNs with prominent dysplasia.
Myelodysplastic/myeloproliferative neoplasms (MDS/MPN)
Reporting on the clinical significance of DNA variants in each of these neoplasms is variable and
TE D
consists of predominantly small series or the inclusion of these cases within larger general groups. Thus, accurate data extraction is difficult. However, as might be expected from their clinicopathologic
EP
similarities to MDS and MPN, there is significant genetic overlap among these entities.
Chronic myelomonocytic leukemia (CMML)
AC C
Genes reported to show variants in ≥10% of cases in at least one series included: TET2 (22% to 61%),28,29,32–34,42,43,107,124,127 SRSF2 (28% to 52%),28,32–34,43,73,132 ASXL1 (22% to 60%),25,29-34,42,43,73,99,105-107 RUNX1 (7% to 37%),31,32,34,35,42,43,85 CBL (10% to 22%),27–34 CEBPA (4% to 20%),31,80,81 JAK2 (1% to 10%),28,29,31–34,43,44 KRAS (7% to 18%),25,28,29,31–34,43 NRAS (4% to 16%),28,29,31–34,43 SETBP1 (4% to 16%),3133,90–93
EZH2 (6% to 13%),25,28–34,106,112 DNMT3A (2% to 13%),25,29,31–34,105 KIT (<1% to 11%),34,69 IDH2 (0% to
11%),32–34,43,118 SF3B1 (4% to 10%),31,32,34,43,108 U2AF1 (5% to 15%),31–34 STAG2 (10%),31 ZRSFR2 (8% to 10%),31,32,34,43 and cohesins in general (6% to 10%).31,138 Genes from all major cell process categories
ACCEPTED MANUSCRIPT
were represented with no single gene or gene category aligned specifically with CMML or any of its subcategories. Variant identification in some genes appears to have prognostic utility for CMML patients (Figure 4 and Supplemental Tables S1, S2, S3, S4, and S5). Variants in the following genes are the most
RI PT
significant poor prognostic markers: ASXL1 (increased progression to AML, decreased LFS and OS, poor overall),28,31,34,42,43,96,99 CBL (decreased OS, poor overall),23,24,27,29,34 DNMT3A (decreased overall survival, poor overall),29,31 EZH2 (increased progression, decreased OS, poor overall),28–31,33,43,112,113 MPL
SC
(decreased OS),33 NPM1 (increased progression, decreased OS),31,81,98 NRAS (decreased OS, poor
overall),33,34 and SETBP1 (increased progression to AML).86,87,94,95 Variants in the following genes had
M AN U
ambiguous data with respect to prognosis: CEBPA,80,81 FLT3,39-41 IDH2,34,43,118 RUNX1,28,31,81 SETBP1 (overall survival),86,87,94-97 SF3B1, SRSF2,28,31,34 and TET2.12,22-24,28,31,34,42,43,90,101,105,107,124 Variants in the remaining genes either had no impact on prognosis or no data identified. No gene variants were identified as being predictive of response to therapies currently available for the treatment of CMML.
TE D
However, rare tumors containing activating FLT3 variants have been treated with FLT3 inhibitors.151,152 Other signaling pathway components such as JAK2, KRAS/NRAS, MAPK, and MEK as well as methylation pathway components IDH1/2 have been the focus of targeted therapy development.153
EP
Clonal architecture studies in CMML have shown very similar findings to those identified in MDS as discussed above: variants in genes involved in methylation or RNA splicing, in particular ASXL1, TET2,
AC C
and SRSF2, are identified early and variants in genes encoding transcription factors and signaling/other pathway components appear later in clonal progression.32 At this time, gene variants aligning specifically with monocytic differentiation, the hallmark feature of CMML, have not been clearly identified. It remains unclear whether prominent monocytic differentiation is due to a specific combination and/or order of acquisition of known myeloid genes, or whether the key driver variant(s) remain to be discovered in other genes. Of note, a study using whole-exome sequencing revealed greater than 10% of CMML cases contain variants in several genes not among those currently described as recurring variants
ACCEPTED MANUSCRIPT
in CMNs.33 CBL variants are enriched in CMML compared with other CMNs and it has been proposed that the presence of a CBL variant should suggest the diagnosis of CMML in the correct clinical context.73 Not surprisingly, when compared with MDS, CMML as a group shows a higher percentage of cases
RI PT
containing variants in signaling genes associated with proliferative features (eg, JAK2, MPL, KIT, KRAS, and NRAS).
SC
Atypical CML (aCML)
Genes reported to show variants in ≥10% of cases in at least one series included: ASXL1 (20% to
M AN U
66%),31,73 TET2 (30% to 41%),31,73 SRSF2 (40%),73 SETBP1 (24% to 32%),31,77,93,97,98 NRAS (27% to 30%),31,77 U2AF1 (13%)77, EZH2 (13% to 20%),31,77,112 and KRAS (10%).31 Genes from all major cell process categories were represented but no single gene or gene category aligned specifically with aCML. Variant identification in some genes appears to have prognostic utility for patients with aCML (Figure 4). Variants in SETBP1 are enriched in aCML and are the most significant poor prognostic markers
TE D
(decreased OS).86,93 Rare studies show EZH2 is also associated with inferior prognosis.31,112 The following genes were suggested to be associated with poor prognosis in at least one study: ASXL1,
EP
DNMT3A, IDH1/2, FLT3, and NPM1.31 No gene variants were identified as good prognostic markers. The remainder either had no impact on prognosis, contradictory data, or no data identified. No gene variants
AC C
were identified as having implications for targeted therapy.
MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T, formerly refractory anemia with ring sideroblasts and thrombocytosis, RARS-T) Genes reported to show variants in ≥10% of cases in at least one series included: SF3B1 (72% to 87%),12,31,45,46,130 JAK2 (50% to 100%),45–49 MPL (1% to 23%),31,41,45,46,48 EZH2 (12% to 25%),25,112 BCOR (24%),25 DNMT3A (12% to 17%),31,44 CALR (13% to 15%),11,12 TET2 (26%),47 and ASXL1 (10%).45,108 Variants
ACCEPTED MANUSCRIPT
in SF3B1 are associated with ring sideroblasts, as discussed above, explaining the high incidence of these variants identified in MDS/MPN-RS-T. However, as in other CMNs, variants in epigenetic genes, such as EZH2, BCOR, DNMT3A, TET2, and ASXL1, are also present in most cases and likely represent initial
RI PT
events. Acquisition of an SF3B1 variant may precede or follow the appearance of variants in JAK2, CALR, or MPL which give the clone proliferative features, including thrombocytosis.
SC
Myeloproliferative Neoplasms (MPNs) Essential thrombocythemia (ET)
M AN U
Genes reported to show variants in ≥10% of cases in at least one series included: JAK2 (2% to 75%),10,13,15,18,35,44,49-54 CALR (16% to 73%),10,11,13–17 ASXL1 (4% to 25%),31,35,61 TET2 (5% to 21% and 13% to 30% in post-ET-MF),13,35,123,127,128 MPL (1% to 8% and 11% in post-ET MF),10,11,13,16,17,36,44,49,52,67 and SF3B1 (2% to 3% and 10% in post-ET-MF).35,108 Approximately 90% of ET clones acquire a variant in one of JAK2 (~60%), CALR (~25%), or MPL (~5%) at some point, with these being virtually mutually exclusive in
TE D
any given cell/clone.15 No single gene or gene category aligned exclusively with ET. Variant identification in some genes appears to have prognostic utility for patients with ET (Figure 4 and Supplemental Tables
EP
S1, S2, S3, S4, and S5). Variants in the following genes are the most significant poor prognostic markers: ASXL1 (increased fibrosis),35 JAK2 (increased thrombosis, rare transformation to PV with very high VAF,
AC C
increased fibrosis),11,14,15,50,51,53,57 MPL (increased thrombosis, poorer OS),11,52 TET2 (increased progression to AML and poorer OS),13 and TP53 (particularly biallelic loss of function leading to rapid transformation to AML, decreased OS).13 In addition, patients with more variants carry a worse prognosis than patients with fewer variants.13 Variants in CALR are associated with increased platelet counts but decreased thrombosis.11,13,14,15,17,18 There is no clear association of JAK2, CALR, or MPL variant status with major bleeding events, risk of transformation to AML, or OS. The remainder of the gene variants reported either had no impact on prognosis or no data identified. However, lack of an
ACCEPTED MANUSCRIPT
identifiable variant in JAK2, CALR, or MPL (triple negative cases) appears to be a poor prognostic feature in ET.13 Evaluations of DNA variants, including single case clonal architecture studies,13 have provided
RI PT
new insight into the pathogenesis of ET. As for other CMNs, variants in genes coding for components of methylation or splicing are identified early in the majority of cases studied. Variants in genes coding for components of signaling and other pathways (including JAK2, MPL, and possibly CALR) typically appear
SC
secondarily but contribute to the proliferative clinicopathologic features.18
It has been suggested that ET with JAK2 variants and PV are a spectrum of the same disease,15 as
M AN U
both entities show a direct correlation between the JAK2 VAF and the level of erythrocytosis.50 Additionally, the rare cases of ET that progress to PV show an apparent, direct relationship to the JAK2 VAF at diagnosis.15 Rare neoplasms with low JAK2 VAF, that initially present as ET, can later lose the unmutated JAK2 allele (through loss of heterozygosity at chromosome 9p), resulting in increased JAK2
TE D
VAF, progression to a PV phenotype, and rapid progression to fibrosis.15 Very high JAK2 VAF (>75%, sometimes referred to as "homozygous") has only been described in ET with secondary fibrosis and PV.15,154 Reports of PV transforming to ET were not found and PV does not ever appear to develop from
EP
ET containing CALR or MPL variants.15
AC C
Polycythemia vera (PV)
Genes reported to show variants in ≥10% of cases in at least one series included: JAK2
(essentially 100% with good sensitivity assay and all exons evaluated),10,13,15,18,35,36,44,50-54,56,57 ASXL1 (7% and 22% to 50% in post-PV MF),35,109 TET2 (7% to 36% and 7% to 33% in post-PV MF).13,28,35,127,128 Although JAK2 variants are not specific for PV, essentially all PVs contain a JAK2 variant and do not contain variants in either CALR or MPL.10,11 Variant identification in some genes appears to have prognostic utility for patients with PV (Figure 4 and Supplemental Tables S1, S2, S3, S4, and S5).
ACCEPTED MANUSCRIPT
Variants in the following genes are the most significant poor prognostic markers: ASXL1 (increased fibrosis),35 JAK2 (very high VAF associated with increased thrombosis and fibrosis and poor overall prognosis),15,50,51,54,57–59 TET2 (increased progression to AML and poorer OS),13 and TP53 (particularly
RI PT
biallelic loss of function leading to rapid transformation to AML).13 In addition, patients with more
variants carry a worse prognosis than patients with fewer variants.13 Variants in the remaining genes either had no impact on prognosis or no data identified.
SC
Individual patient clonal hierarchy studies13 and others studies18 indicate that, as for other
CMNs, variants in genes coding for components of methylation or splicing can appear early in PV with
M AN U
the secondary appearance of the JAK2 variant, although “JAK2-first” cases (where mutations in JAK2 occur first and epigenetic or splicing second) are more likely to present as PV.155 Variants in genes of signaling and alternate cell processes may also appear secondarily. Erythrocytosis is the hallmark of PV
Primary myelofibrosis (PMF)
TE D
and is closely associated with a high level of JAK2 activity.15,50
Genes reported to show variants in ≥10% of cases in at least one series included: ASXL1 (12% to
EP
75%),20,35,61,76,106,109 JAK2 (25% to 79%),13,15,17,35,56,61CALR (23% to 60%),10,11,13,15,17,19 TET2 (10% to 50%),13,35,61,76,106,127,129 SRSF2 (9% to 17%),76,122 EZH2 (6% to 13%),18,36,76,106,112 and DNMT3A (3% to
AC C
7%).18,35,76,106,111 Among genes with variants present in <10% of cases, MPL (3% to 9%) is highlighted here, as it has been reported as a recurrent gene in PMF in many series.11,13,35,36,44,52,66,76,74,75 Approximately 90% of PMF clones are identified as having a variant in one of JAK2 (~60%), CALR (~25%), or MPL (~5%) at some point, with these being virtually mutually exclusive in the same cell/clone.10,11,15,156 No single gene or gene category aligned specifically with the diagnosis of PMF. Variant identification in some genes appears to have prognostic utility for patients with PMF (Figure 4 and Supplemental Tables S1, S2, S3, S4, and S5). Variants in the following genes are the most significant poor prognostic markers:
ACCEPTED MANUSCRIPT
ASXL1 (increased fibrosis, increased progression, decreased OS, poor overall),14,19,20,31,76,104 EZH2 (increased progression, decreased LFS and OS),14,36,76,112 IDH1 (decreased LFS, decreased OS),76 JAK2 (need for splenectomy, increased progression, poor overall),32,55,57,58 MPL (when compared with PMF-
RI PT
CALR and PMF-JAK2 – increased risk of transfusion dependency, poorer OS),11,52,60,74 NRAS (poor
overall),56 and SRSF2 (increased progression, decreased LFS and OS, poor overall).14,66,116 As in the other CMNs, patients with more variants carry a worse prognosis than patients with fewer variants,13 and
SC
PMF lacking detectable variants in any of the main three genes (JAK2, CALR, or MPL) has a particularly poor overall prognosis.11,16,56 The following are the most significant good prognostic markers: CALR
M AN U
(lower risk of thrombosis11 and increased OS11,18,20 when compared with those with variants in JAK2 and/or MPL). The following had ambiguous data with respect to prognosis: IDH275,76,120 and MPL (when compared with PMF-JAK2).66,67 Variants in the remaining genes either had no impact on prognosis or no data identified.
TE D
Individual clonal hierarchy studies indicate that, as for other CMNs, variants in genes coding for components of methylation or splicing appear early in PMF in the majority of cases and variants in genes coding for components of signaling and alternate cell processes occur secondarily.13,76,157 Although there
EP
are few clinicopathologic features specifically associated with secondary gene variants in PMF, it appears that clones containing CALR variants are associated with higher platelet counts, but lower
AC C
thrombotic events and lower total white blood cell counts than clones containing JAK2 variants.11 Additional insight into the pathogenesis of ET, PV, and PMF has been gained by detailed analysis
of DNA variants with respect to clonal architecture. Regardless of JAK2, CALR, or MPL status, the final phenotype and natural history is affected by the presence and order of appearance of variants in a variety of other myeloid genes.155,158 For example, Ortmann et al demonstrated that although variants of JAK2, TET2, or both may appear in all of ET, PV and PMF, clones acquiring JAK2 p.V617F prior to emergence of a TET2 variant (JAK2-first) behave differently than those acquiring TET2 variants prior to
ACCEPTED MANUSCRIPT
emergence of JAK2 p.V617F (TET2-first).155 JAK2-1st stem and progenitor cells can undergo erythroid/megakaryocyte differentiation but do not themselves expand until they subsequently acquire a TET2 variant. This correlates to the observation that these patients are more likely to present as PV
RI PT
and have thrombocytosis. In contrast, TET2-1st stem and progenitor cells show clonal expansion, but do not undergo erythroid/megakaryocyte differentiation until they subsequently acquire a JAK2 variant. As such, these MPNs are not as likely to show erythrocytosis and/or thrombocytosis at initial presentation.
number of variants present (clone complexity).155
SC
As in MDS, the clinical aggressiveness of all MPN clones generally appears to be directly related to the
M AN U
Because it appears that signaling through various tyrosine kinase pathways is a common feature of these diseases, many small molecule therapeutics that target a variety of components in several key pathways are being utilized or evaluated (eg, inhibitors of JAK, PI3K, AKT, MTOR, TORC, histone deacetylase). It remains to be seen whether gene variant data will be informative for guiding therapy
TE D
further in classic MPNs. Several recent reviews of this topic are available.159,160
Chronic neutrophilic leukemia (CNL)
EP
When strict WHO criteria are applied, essentially 100% of these extremely rare cases have a variant in CSF3R.161–163 The p.T618I mutation is highly specific and correlates with sensitivity to
AC C
ruxolitinib.163,164 Clones containing the p.S783fs* mutation, and possibly other c-terminal truncating variants, show inhibition by dasatinib.164 SETBP1 variants are reported in the range of 15% to 55%, but some reports that have taken a more rigid classification of aCML and CNL have suggested that the percentage may be closer to 1%.1,73,161,162
Chronic eosinophilic leukemia (CEL)
ACCEPTED MANUSCRIPT
Chronic eosinophilic leukemia, not otherwise specified (CEL-NOS), is a clonal proliferation of eosinophils that does not contain translocations of PDGFRA, PDGFRB, or FGFR1. In addition to cytogenetic aberrations, clonality may be demonstrated by detection of small, somatic DNA variants in
RI PT
myeloid genes, the most common being: ASXL1 (43%), TET2 (36%), EZH2 (29%), SETBP1 (22%), CBL (14%), NOTCH1 (14%), and rarely DNMT3A, NRAS, JAK2 (non-p.V617F), and GATA2.165 DNA variant
evaluation now allows for a diagnosis of CEL in many cases that would have previously been considered
SC
idiopathic hypereosinophilic syndrome.1,2 Further studies will be required to clarify what underlying
M AN U
pattern of clonal architecture and progression of variants gives this CMN its clinicopathologic features.
Systemic Mastocytosis (SM)
Genes reported to show variants in ≥10% in at least one series included: KIT (essentially 100%),38,68,70,71 TET2 (20% to 47%),37,38,72,129 SRSF2 (36% to 43%),38,68 ASXL1 (12% to 29%),37,38,68 RUNX1 (23%),38,68 JAK2 (5% to 16%),38,68 KRAS/NRAS (14%),38,68 CBL (4% to 13%),37,38 DNMT3A (12%),37 EZH2 (5%
TE D
to 10%),38,68 and KRAS (10%).68 For the following genes, the highest incidence was reported in systemic mastocytosis with an associated hematopoietic neoplasm (SM-AHN): ASXL1, CBL, JAK2, KRAS/NRAS, and
EP
TET2. Genes from all major cell process categories were represented, but no single gene or gene category aligned exclusively with SM or any of its subcategories. However, the outgrowth of mature
AC C
mast cells seen in mastocytosis is strongly associated with KIT variants and essentially all cases have a KIT variant in the mature mast cell component, with variable presence in other hematopoietic compartments.69,71 It should be noted that increased mast cells may be seen in other hematologic malignancies as either reactive (eg, in lymphoplasmacytic lymphoma) or as part of the neoplastic clone due to increased KIT activation without mutation (eg, in FIP1L1/PDGFRA myeloid/lymphoid neoplasm with eosinophilia). Variant identification in some genes appears to have prognostic utility for patients with SM. Variants in the following genes were the most significant poor prognostic markers: ASXL1
ACCEPTED MANUSCRIPT
(decreased OS, especially if present with variants in one or more of RUNX1, SRSF2, DNMT3A),37,38,68 DNMT3A (decreased OS, especially if present with variants of one or more of TET2 or ASXL1),37 KIT (poor overall if present in multiple lineages),70,71 RUNX1 (decreased OS, especially if present with variants in
RI PT
one or more of ASXL1 and SRSF2),38 and SRSF2 (decreased OS, especially if present with variants in one or more of ASXL1 and RUNX1).38 A KIT variant identified only in mast cells is a good prognostic marker. Variants in the remaining genes either had no impact on prognosis or no data were identified.
SC
Tyrosine kinase inhibitors with inhibitory activity for KIT may be used for therapy in systemic mastocytosis, although each drug has a unique profile with respect to activity on protein derived from
M AN U
the wild-type gene and from KIT with the various known variants.166–168 For example, KIT p.D816V is associated with resistance to the tyrosine kinase inhibitor, imatinib, but has been associated with responses to other inhibitors including midostaurin.
Clonal architecture studies in SM have shown that SM-AHN clones, including those designated
TE D
as aggressive (ASM-AHN), have initiating variants in non-KIT genes.38,69 Similar to other CMNs, these variants found in SM-AHN are most common in genes found in the associated hematologic neoplasm involved in epigenetics (TET2, ASXL1, and EZH2) or RNA splicing (SRSF2). In these cases, the KIT variants
EP
appear to arise within a subclone in a similar manner to other CMNs where variants in signaling pathway genes appear secondarily.68 The non-KIT gene variant patterns identified in these cases parallel the
AC C
patterns seen in the associated non-SM CMNs (eg, SM-AHNs in which the AHN resembles CMML have clonal variant architecture similar to CMML in addition to the KIT variant). Interestingly, within the broader group of neoplasms classified as mastocytosis, the following features directly correlate with aggressiveness: presence and number of non-KIT gene variants, acquisition of the KIT variant within early hematopoietic compartments (HSC or early myeloid progenitor compartments) and higher KIT VAF within the compartment. Clinically indolent SM clones rarely contain non-KIT variants and only rarely contain the KIT variant in an early hematopoietic compartment (when presentat low VAF). The most
ACCEPTED MANUSCRIPT
indolent forms of mastocytosis (non-systemic/localized to skin and mucosa) appear to have KIT variants that are found only in mature mast cells, without variants identified in additional myeloid genes.69
RI PT
DISCUSSION
This review was initiated following AMP member discussions that yielded a consensus that the recent explosion of literature regarding the clinical relevance of small DNA variants in CMNs, driven by
SC
the use of high-throughput sequencing, was ripe for comprehensive review. This led to the formation of the AMP CMN Working Group. This group was charged with the challenging task of comprehensively
M AN U
reviewing the literature, summarizing key findings that support clinical utility, and defining the need for gene inclusion in high-throughput sequencing testing panels. It was evident, early in the process, that there was a need to clearly place the commonly reported genes within the context of their normal cell biology to clarify the discussion of the extensive literature. A second challenge was the ever-increasing
TE D
volume of detailed literature pertaining to the topic. Thus, the Working Group's summary of relevant published data attempts to pull findings into a readily accessible format that identifies clinically relevant biomarkers, provides insight into basic biology, and establishes easy access to key references. This
EP
exercise was helpful to identify a list of "myeloid genes" that appear to be the most important biomarkers based on our current understanding of CMNs. The exercise also highlighted not only the
AC C
complexity of the underlying biology of these neoplasms but also the genetic heterogeneity within diagnostic categories and the similarities between apparently disparate diagnostic entities. Recent meticulous studies evaluating the architecture of individual patient clones over time and through therapy have begun to provide clarity on the pathogenesis of CMNs. They have shown that each neoplasm is unique but that there is an overarching theme (Figure 5). In nearly all cases, the founding variant is in a gene involved in either epigenetic regulation or RNA splicing, the latter being particularly common in patients >70 years old. Variants in genes of other cell processes are often identified as later
ACCEPTED MANUSCRIPT
events. It is these later-appearing variants and their relative dominance within the total neoplastic population that appears to determine the heterogeneous clinicopathologic features currently used for sub-classification of CMNs. This relative dominance also appears to have the potential to drive
RI PT
progression toward more aggressive behavior. The hematopoietic differentiation compartment in which the founding variant is identified, the specific genes and specific variants that are present, the order of variant appearance, individual subclone dynamics, and therapeutic intervention, all contribute to the
SC
clinicopathologic features of any individual CMN at any point in time. Insight into the effect that
combinations of relevant biomarkers have on the specific clinicopathologic characteristics of CMNs
M AN U
continues to accumulate.
As more targeted therapies become available, their selection and efficacy will likely be based on the profile of DNA variants present in each patient's clone at various time points. Several inhibitors targeting elements of signaling pathways are already in clinical use. Therapies targeting spliceosome
TE D
components and epigenetic modulators will soon become available. Thus, sequencing for DNA variants, remains critical for patient management at all levels (diagnosis, prognosis, therapy, monitoring). For the time being, genomic diagnostic laboratories must be capable of identifying variants in a large list of
EP
myeloid genes, with optimal sensitivity, and be able to aid in diagnosis and reliably evaluate and report clonal architecture throughout the course of disease.
AC C
Based on this review, the following core 34-gene set (including the core CHIP gene set) is recommended to provide relevant clinical information for the management of most CMNs at this time (Supplemental Table S6): ASXL1, BCOR, BCORL1, CALR, CBL, CEBPA, CSF3R, DNMT3A, ETV6, EZH2, FLT3, IDH1, IDH2, JAK2, KIT, KRAS, MPL, NF1, NPM1, NRAS, PHF6, PPM1D, PTPN11, RAD21, RUNX1, SETBP1, SF3B1, SMC3, SRSF2, STAG2, TET2, TP53, U2AF1, and ZRSR2. For utilization of some CMN prognostic scoring systems, other genes may be required, and this list does not include all of the variants that may be seen in CHIP or other myeloid neoplasms, such as AML or aplastic anemia. As a result, laboratories
ACCEPTED MANUSCRIPT
endeavoring to develop a pan-myeloid panel should consider these recommendations as a minimum recommended list to provide relevant diagnostic and prognostic information in CMNs and enable
RI PT
monitoring of clonal architecture.
DISCLAIMER
The AMP Clinical Practice Guidelines and Reports are developed to be of assistance to laboratory and
SC
other healthcare professionals by providing guidance and recommendations for particular areas of practice. The Guidelines or Report should not be considered inclusive of all proper approaches or
M AN U
methods, or exclusive of others. The Guidelines or Report cannot guarantee any specific outcome, nor do they establish a standard of care. The Guidelines or Report are not intended to dictate the treatment of a particular patient. Treatment decisions must be made based on the independent judgment of healthcare providers and each patient’s individual circumstances. AMP makes no warranty, express or
TE D
implied, regarding the Guidelines or Report and specifically excludes any warranties of merchantability and fitness for a particular use or purpose. AMP shall not be liable for direct, indirect, special, incidental,
EP
or consequential damages related to the use of the information contained herein.
ACKNOWLEDGMENTS
AC C
We thank Norman Cyr for his expert graphic design contributions to this manuscript; Dr. R. Coleman Lindsley for helpful discussions; and multiple colleagues for their primary literature contributions to the field which have been referenced within reviews due to journal limitations.
ACCEPTED MANUSCRIPT
REFERENCES 1.
Swerdlow S, Campo E, Harris N, Jaffe E, Pileri S, Stein H, Thiele J: Classification of Tumours of
2.
RI PT
Haematopoietic and Lymphoid Tissues: Revised Fourth Edition. World Health Organization Press, 2017. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M,
Vardiman JW: The 2016 revision to the World Health Organization classification of myeloid neoplasms
3.
SC
and acute leukemia. Blood 2016, 127:2391–2405.
Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose S a, Bakhoum SF, Chambert K, Mick E,
M AN U
Neale BM, Fromer M, Purcell SM, Svantesson O, Landén M, Höglund M, Lehmann S, Gabriel SB, Moran JL, Lander ES, Sullivan PF, Sklar P, Grönberg H, Hultman CM, McCarroll SA, Rose SA, Bakhoum SF, Chambert K, Mick E, Neale BM, Fromer M, Purcell SM, Svantesson O, Sullivan PF, Sklar P, Grönberg H, Hultman CM, McCarroll SA: Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA
4.
TE D
Sequence. N Engl J Med 2014, 371:2477–2487.
Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman P V, Mar BG, Lindsley RC, Mermel CH,
Burtt N, Chavez A, Higgins JM, Moltchanov V, Kuo FC, Kluk MJ, Henderson B, Kinnunen L, Koistinen HA,
EP
Ladenvall C, Getz G, Correa A, Banahan BF, Gabriel S, Kathiresan S, Stringham HM, McCarthy MI, Boehnke M, Tuomilehto J, Haiman C, Groop L, Atzmon G, Wilson JG, Neuberg D, Altshuler D, Ebert BL:
2498. 5.
AC C
Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014, 371:2488–
Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, McMichael JF, Schmidt HK,
Yellapantula V, Miller CA, Ozenberger BA, Welch JS, Link DC, Walter MJ, Mardis ER, Dipersio JF, Chen F, Wilson RK, Ley TJ, Ding L: Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014, 20:1472–1478.
ACCEPTED MANUSCRIPT
6.
Wahl MC, Will CL, Lührmann R: The Spliceosome: Design Principles of a Dynamic RNP Machine.
Cell, Elsevier, 2018, 136:701–718. Singh VP, Gerton JL: Cohesin and human disease: lessons from mouse models. Curr Opin Cell
RI PT
7.
Biol 2015, 37:9–17. 8.
Losada A: Cohesin in cancer: chromosome segregation and beyond. Nat Rev Cancer 2014,
SC
14:389–393.
Morreale F, Walden H: Types of Ubiquitin Ligases. Cell 2016, 165:248.
10.
Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, Avezov E, Li J, Kollmann K, Kent
M AN U
9.
DG, Aziz A, Godfrey a L, Hinton J, Martincorena I, Van Loo P, Jones a V, Guglielmelli P, Tarpey P, Harding HP, Fitzpatrick JD, Goudie CT, Ortmann C a, Loughran SJ, Raine K, Jones DR, Butler a P, Teague JW, O’Meara S, McLaren S, Bianchi M, Silber Y, Dimitropoulou D, Bloxham D, Mudie L, Maddison M,
TE D
Robinson B, Keohane C, Maclean C, Hill K, Orchard K, Tauro S, Du M-Q, Greaves M, Bowen D, Huntly BJP, Harrison CN, Cross NCP, Ron D, Vannucchi a M, Papaemmanuil E, Campbell PJ, Green a R: Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013, 369:2391–2405. Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NCC, Berg T,
EP
11.
Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant’Antonio E, Ferretti V,
AC C
Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schönegger A, Bock C, Malcovati L, Pascutto C, SupertiFurga G, Cazzola M, Kralovics R: Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013, 369:2379–2390. 12.
Malcovati L, Karimi M, Papaemmanuil E, Ambaglio I, Jädersten M, Jansson M, Elena C, Gallì A,
Walldin G, Della Porta MG, Raaschou-Jensen K, Travaglino E, Kallenbach K, Pietra D, Ljungström V, Conte S, Boveri E, Invernizzi R, Rosenquist R, Campbell PJ, Cazzola M, Hellström Lindberg E: SF3B1 mutation
ACCEPTED MANUSCRIPT
identifies a distinct subset of myelodysplastic syndrome with ring sideroblasts. Blood 2015, 126:233– 241. Lundberg P, Karow A, Nienhold R, Looser R, Hao-shen H, Nissen I, Girsberger S, Lehmann T,
RI PT
13.
Passweg J, Stern M, Beisel C, Kralovics R, Skoda RC: Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood 2014, 123:2220–2228.
Rotunno G, Mannarelli C, Guglielmelli P, Pacilli A, Pancrazzi A, Pieri L, Fanelli T, Bosi A, Vannucchi
SC
14.
AM, Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative
M AN U
Investigators: Impact of calreticulin mutations on clinical and hematological phenotype and outcome in essential thrombocythemia. Blood 2014, 123:1552–1555. 15.
Rumi E, Pietra D, Ferretti V, Klampfl T, Harutyunyan AS, Milosevic JD, Them NCC, Berg T, Elena C,
Casetti IC, Milanesi C, Sant’Antonio E, Bellini M, Fugazza E, Renna MC, Boveri E, Astori C, Pascutto C, Kralovics R, Cazzola M, Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie
TE D
Mieloproliferative Investigators: JAK2 or CALR mutation status defines subtypes of essential
1551. 16.
EP
thrombocythemia with substantially different clinical course and outcomes. Blood 2014, 123:1544–
Al Assaf C, Van Obbergh F, Billiet J, Lierman E, Devos T, Graux C, Hervent A-SS, Emmerechts J,
AC C
Tousseyn T, De Paepe P, Papadopoulos P, Michaux L, Vandenberghe P: Analysis of phenotype and outcome in essential thrombocythemia with CALR or JAK2 mutations. Haematologica 2015, 100:893– 897. 17.
Andrikovics H, Krahling T, Balassa K, Halm G, Bors A, Koszarska M, Batai A, Dolgos J, Csomor J,
Egyed M, Sipos A, Remenyi P, Tordai A, Masszi T: Distinct clinical characteristics of myeloproliferative neoplasms with calreticulin mutations. Haematologica 2014, 99:1184–1190.
ACCEPTED MANUSCRIPT
18.
Cazzola M, Kralovics R: From Janus kinase 2 to calreticulin: the clinically relevant genomic
landscape of myeloproliferative neoplasms. Blood 2014, 123:3714–3719. Tefferi A, Guglielmelli P, Lasho TL, Rotunno G, Finke C, Mannarelli C, Belachew AA, Pancrazzi A,
RI PT
19.
Wassie EA, Ketterling RP, Hanson C a, Pardanani A, Vannucchi AM: CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia
20.
SC
2014, 28:1494–1500.
Tefferi A, Lasho TL, Finke CM, Knudson RA, Ketterling R, Hanson CH, Maffioli M, Caramazza D,
M AN U
Passamonti F, Pardanani A: CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia 2014, 28:1472–1477. 21.
Panagiota V, Thol F, Markus B, Fehse B, Alchalby H, Badbaran A, Lehmann U, Koenecke C,
Shahswar R, Chaturvedi A, Stadler M, Eder M, Göhring G, Koenigsmann M, Kloos A, Trummer A, Schroeder T, Kobbe G, Thiede C, Platzbecker U, Schlegelberger B, Kreipe H-H, Ganser A, Kröger N,
TE D
Heuser M: Prognostic effect of calreticulin mutations in patients with myelofibrosis after allogeneic hematopoietic stem cell transplantation. Leukemia 2014, 28:1552–1555. Bejar R, Stevenson KE, Caughey B a., Abdel-Wahab O, Steensma DP, Galili N, Raza A, Kantarjian
EP
22.
H, Levine RL, Neuberg D, Garcia-Manero G, Ebert BL: Validation of a prognostic model and the impact of
23.
AC C
mutations in patients with lower-risk myelodysplastic syndromes. J Clin Oncol 2012, 30:3376–3382. Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, Kantarjian H, Raza A,
Levine RL, Neuberg D, Ebert BL: Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med 2011, 364. 24.
Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, Schnittger S, Sanada M, Kon
A, Alpermann T, Yoshida K, Roller A, Nadarajah N, Shiraishi Y, Shiozawa Y, Chiba K, Tanaka H, Koeffler
ACCEPTED MANUSCRIPT
HP, Klein H-U, Dugas M, Aburatani H, Kohlmann A, Miyano S, Haferlach C, Kern W, Ogawa S: Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia, Nature Publishing Group,
25.
RI PT
2014, 28:241–247. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, Yoon CJ, Ellis P,
Wedge DC, Pellagatti A, Shlien A, Groves MJ, Forbes SA, Raine K, Hinton J, Mudie LJ, McLaren S, Hardy C, Latimer C, Della Porta MG, O’Meara S, Ambaglio I, Galli A, Butler AP, Walldin G, Teague JW, Quek L,
SC
Sternberg A, Gambacorti-Passerini C, Cross NCP, Green AR, Boultwood J, Vyas P, Hellstrom-Lindberg E, Bowen D, Cazzola M, Stratton MR, Campbell PJ: Clinical and biological implications of driver mutations in
26.
M AN U
myelodysplastic syndromes. Blood 2013, 122:3616–3627.
Bejar R: Clinical and genetic predictors of prognosis in myelodysplastic syndromes.
Haematologica, Ferrata Storti Foundation, 2014, 99:956–964. 27.
Grand FH, Hidalgo-Curtis CE, Ernst T, Zoi K, Zoi C, McGuire C, Kreil S, Jones A, Score J,
TE D
Metzgeroth G, Oscier D, Hall A, Brandts C, Serve H, Reiter A, Chase AJ, Cross NCP: Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. Blood
28.
EP
2009, 113:6182–6192.
Meggendorfer M, Roller A, Haferlach T, Eder C, Dicker F, Grossmann V, Kohlmann A, Alpermann
AC C
T, Yoshida K, Ogawa S, Koeffler HP, Kern W, Haferlach C, Schnittger S: SRSF2 mutations in 275 cases with chronic myelomonocytic leukemia (CMML). Blood 2012, 120:3080–3088. 29.
Jankowska AM, Makishima H, Tiu R V, Szpurka H, Huang Y, Traina F, Visconte V, Sugimoto Y,
Prince C, O’Keefe C, Hsi ED, List A, Sekeres M a, Rao A, McDevitt M a, Maciejewski JP: Mutational spectrum analysis of chronic myelomonocytic leukemia includes genes associated with epigenetic regulation: UTX, EZH2, and DNMT3A. Blood 2011, 118:3932–3941.
ACCEPTED MANUSCRIPT
30.
Grossmann V, Kohlmann A, Eder C, Haferlach C, Kern W, Cross NCP, Haferlach T, Schnittger S:
Molecular profiling of chronic myelomonocytic leukemia reveals diverse mutations in 80% of patients
31.
Zoi K, Cross NCP: Molecular pathogenesis of atypical CML, CMML and MDS/MPN-unclassifiable.
Int J Hematol, Springer Japan, 2015, 101:229–242.
Merlevede J, Droin N, Qin T, Meldi K, Yoshida K, Morabito M, Chautard E, Auboeuf D, Fenaux P,
SC
32.
RI PT
with TET2 and EZH2 being of high prognostic relevance. Leukemia 2011, 25:877–879.
Braun T, Itzykson R, de Botton S, Quesnel B, Commes T, Jourdan E, Vainchenker W, Bernard O, Pata-
M AN U
merci N, Gayevskiy V, Dinger ME, Cowley MJ, Selimoglu-Buet D, Meyer V, Artiguenave F, Deleuze J-F, Preudhomme C, Stratton MR, Alexandrov LB, Padron E, Ogawa S, Koscielny S, Figueroa M, Solary E: Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents. Nat Commun 2016, February 2:1–13. 33.
Mason C, Khorashad J, Tantravahi S, Kelley T, Zabriskie M, Yan D, Pomicter A, Reynolds K, Eiring
TE D
A, Kronenberg Z, Sherman R, Tyner JW, Dalley B, Dao K-H, Yandell M, Druker B, Gotlib J, O’Hare T, Deininger MWN: Age-related mutations and chronic myelomonocytic leukemia. Leukemia 2016,
34.
EP
January:1–8.
Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, Berthon C,
AC C
Adès L, Fenaux P, Beyne-Rauzy O, Vey N, Braun T, Haferlach T, Dreyfus F, Cross NCP, Preudhomme C, Bernard OA, Fontenay M, Vainchenker W, Schnittger S, Birnbaum D, Droin N, Solary E: Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol 2013, 31:2428–2436. 35.
Brecqueville M, Rey J, Bertucci F, Coppin E, Finetti P, Carbuccia N, Cervera N, Gelsi-Boyer V,
Arnoulet C, Gisserot O, Verrot D, Slama B, Vey N, Mozziconacci M, Birnbaum D, Murati A: Mutation Analysis of ASXL1, CBL, DNMT3A, IDH1, JAK2, MPL, NF1, SF3B1, SUZ12, and TET2 in Myeloproliferative Neoplasms. Genes Chromosomes Cancer 2012, 51:743–755.
ACCEPTED MANUSCRIPT
36.
Guglielmelli P, Biamonte F, Score J, Hidalgo-Curtis C, Cervantes F, Maffioli M, Fanelli T, Ernst T,
Winkelman N, Jones A V., Zoi K, Reiter A, Duncombe A, Villani L, Bosi A, Barosi G, Cross NCP, Vannucchi
37.
RI PT
AM: EZH2 mutational status predicts poor survival in myelofibrosis. Blood 2011, 118:5227–5234. Traina F, Visconte V, Jankowska AM, Makishima H, Keefe CLO, Elson P, Han Y, Hsieh FH, Sekeres
MA, Kalaycio M, Lichtin AE, Advani AS, Duong HK, Copelan E, Kapur R, Saad STO, Maciejewski JP, Tiu R V:
in Systemic Mastocytosis. PLoS ONE 2012,7(8): e43090.
Jawhar M, Schwaab J, Schnittger S, Meggendorfer M, Pfirrmann M, Sotlar K, Horny H-P,
M AN U
38.
SC
Single Nucleotide Polymorphism Array Lesions, TET2 , DNMT3A , ASXL1 and CBL Mutations Are Present
Metzgeroth G, Kluger S, Naumann N, Haferlach C, Haferlach T, Valent P, Hofmann W-K, Fabarius A, Cross NCP, Reiter A: Additional mutations in SRSF2, ASXL1 and/or RUNX1 identify a high-risk group of patients with KIT D816V+ advanced systemic mastocytosis. Leukemia 2016, 30(1):136-43. 39.
Daver N, Strati P, Jabbour E, Kadia T, Luthra R, Wang S, Patel K, Ravandi F, Cortes J, Qin Dong X,
TE D
Kantarjian H, Garcia-Manero G: FLT3 mutations in myelodysplastic syndrome and chronic myelomonocytic leukemia. Am J Hematol 2013, 88:56–59. Bains A, Luthra R, Medeiros LJ, Zuo Z: FLT3 and NPM1 mutations in myelodysplastic syndromes:
EP
40.
Frequency and potential value for predicting progression to acute myeloid leukemia. Am J Clin Pathol
41.
AC C
2011, 135:62–69.
Bejar R, Levine R, Ebert BL: Unraveling the molecular pathophysiology of myelodysplastic
syndromes. J Clin Oncol 2011, 29:504–515. 42.
Gelsi-Boyer V, Trouplin V, Roquain J, Adlade J, Carbuccia N, Esterni B, Finetti P, Murati A,
Arnoulet C, Zerazhi H, Fezoui H, Tadrist Z, Nezri M, Chaffanet M, Mozziconacci MJ, Vey N, Birnbaum D:
ACCEPTED MANUSCRIPT
ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia. Br J Haematol 2010, 151:365–375. Itzykson R, Kosmider O, Renneville A, Morabito M, Preudhomme C, Berthon C, Adès L, Fenaux P,
RI PT
43.
Platzbecker U, Gagey O, Rameau P, Meurice G, Oréar C, Delhommeau F, Bernard OA, Fontenay M,
Vainchenker W, Droin N, Solary E: Clonal architecture of chronic myelomonocytic leukemias. Blood
44.
SC
2013, 121:2186–2198.
Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA, Wadleigh M, Steensma DP, Elliott MA,
M AN U
Wolanskyj AP, Hogan WJ, McClure RF, Litzow MR, Gilliland DG, Tefferi A: MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 2006, 108:3472–3476. 45.
Visconte V, Makishima H, Jankowska A, Szpurka H, Traina F, Jerez a, O’Keefe C, Rogers HJ,
Sekeres MA, Maciejewski JP, Tiu R V: SF3B1, a splicing factor is frequently mutated in refractory anemia
46.
TE D
with ring sideroblasts. Leukemia 2012, 26:542–545.
Jeromin S, Haferlach T, Grossmann V, Alpermann T, Kowarsch A, Haferlach C, Kern W, Schnittger
S: High frequencies of SF3B1 and JAK2 mutations in refractory anemia with ring sideroblasts associated
EP
with marked thrombocytosis strengthen the assignment to the category of myelodysplastic/myeloproliferative neoplasms. Haematologica 2013, 98:15–17. Flach J, Dicker F, Schnittger S, Kohlmann A, Haferlach T, Haferlach C: Mutations of JAK2 and
AC C
47.
TET2, but not CBL are detectable in a high portion of patients with refractory anemia with ring sideroblasts and thrombocytosis. Haematologica 2010, 95:518–519. 48.
Broséus J, Alpermann T, Wulfert M, Florensa Brichs L, Jeromin S, Lippert E, Rozman M,
Lifermann F, Grossmann V, Haferlach T, Germing U, Luño E, Girodon F, Schnittger S, MPN and MPNrEuroNet (COST Action BM0902): Age, JAK2(V617F) and SF3B1 mutations are the main predicting factors
ACCEPTED MANUSCRIPT
for survival in refractory anaemia with ring sideroblasts and marked thrombocytosis. Leukemia 2013, 27:1826–1831. Broseus J, Florensa L, Zipperer E, Schnittger S, Malcovati L, Richebourg S, Lippert E, Cermak J,
RI PT
49.
Evans J, Mounier M, Raya JM, Bailly F, Gattermann N, Haferlach T, Garand R, Allou K, Besses C, Germing U, Haferlach C, Travaglino E, Luno E, Pinan MA, Arenillas L, Rozman M, Sirvent MLP, Favre B, Guy J, Alonso E, Ahwij N, Jerez A, Hermouet S, Maynadie M, Cazzola M, Girodon F: Clinical features and course
SC
of refractory anemia with ring sideroblasts associated with marked thrombocytosis. Haematologica
50.
M AN U
2012, 97:1036–1041.
Passamonti F, Rumi E, Pietra D, Della Porta MG, Boveri E, Pascutto C, Vanelli L, Arcaini L,
Burcheri S, Malcovati L, Lazzarino M, Cazzola M: Relation between JAK2 (V617F) mutation status, granulocyte activation, and constitutive mobilization of CD34+ cells into peripheral blood in myeloproliferative disorders. Blood 2006, 107:3676–3682.
Vannucchi AM, Antonioli E, Guglielmelli P, Rambaldi A, Barosi G, Marchioli R, Marfisi RM, Finazzi
TE D
51.
G, Guerini V, Fabris F, Randi ML, De Stefano V, Caberlon S, Tafuri A, Ruggeri M, Specchia G, Liso V, Rossi E, Pogliani E, Gugliotta L, Bosi A, Barbui T: Clinical profile of homozygous JAK2 617V>F mutation in
Beer PA, Campbell PJ, Scott LM, Bench AJ, Erber WN, Bareford D, Wilkins BS, Reilly JT,
AC C
52.
EP
patients with polycythemia vera or essential thrombocythemia. Blood 2007, 110:840–846.
Hasselbalch HC, Bowman R, Wheatley K, Buck G, Harrison CN, Green AR: MPL mutations in myeloproliferative disorders: Analysis of the PT-1 cohort. Blood 2008, 112:141–149. 53.
Carobbio A, Thiele J, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I,
Vannucchi AM, Antonioli E, Gisslinger H, Buxhofer-Ausch V, Finazzi G, Gangat N, Tefferi A, Barbui T: Risk factors for arterial and venous thrombosis in WHO-defined essential thrombocythemia: an international study of 891 patients. Blood 2011, 117:5857–5859.
ACCEPTED MANUSCRIPT
54.
Lippert E, Boissinot M, Kralovics R, Girodon F, Dobo I, Praloran V, Boiret-Dupré N, Skoda RC,
Hermouet S: The JAK2-V617F mutation is frequently present at diagnosis in patients with essential
55.
RI PT
thrombocythemia and polycythemia vera. Blood 2006, 108:1865–1867. Hultcrantz M, Kristinsson SY, Andersson TML, Landgren O, Eloranta S, Derolf ÅR, Dickman PW,
Björkholm M: Patterns of survival among patients with myeloproliferative neoplasms diagnosed in
56.
SC
Sweden from 1973 to 2008: A population-based study. J Clin Oncol 2012, 30:2995–3001.
Tenedini E, Bernardis I, Artusi V, Artuso L, Roncaglia E, Guglielmelli P, Pieri L, Bogani C, Biamonte
M AN U
F, Rotunno G, Mannarelli C, Bianchi E, Pancrazzi A, Fanelli T, Malagoli Tagliazucchi G, Ferrari S, Manfredini R, Vannucchi AM, Tagliafico E, AGIMM investigators: Targeted cancer exome sequencing reveals recurrent mutations in myeloproliferative neoplasms. Leukemia 2014, 28:1052–1059. 57.
Guglielmelli P, Barosi G, Pieri L, Antonioli E, Bosi A, Vannucchi AM: JAK2V617F mutational status
and allele burden have little influence on clinical phenotype and prognosis in patients with post-
146.
Passamonti F, Rumi E, Pietra D, Elena C, Boveri E, Arcaini L, Roncoroni E, Astori C, Merli M, Boggi
EP
58.
TE D
polycythemia vera and post-essential thrombocythemia myelofibrosis. Haematologica 2009, 94: 144-
S, Pascutto C, Lazzarino M, Cazzola M: A prospective study of 338 patients with polycythemia vera: the
AC C
impact of JAK2 (V617F) allele burden and leukocytosis on fibrotic or leukemic disease transformation and vascular complications. Leukemia, Macmillan Publishers Limited, 2010, 24:1574. 59.
Tefferi A, Lasho TL, Schwager SM, Strand JS, Elliott M, Mesa R, Li C-Y, Wadleigh M, Lee SJ,
Gilliland DG: The clinical phenotype of wild-type, heterozygous, and homozygous JAK2V617F in polycythemia vera. Cancer 2006, 106:631–635.
ACCEPTED MANUSCRIPT
60.
Malcovati L, Della Porta MG, Pietra D, Boveri E, Pellagatti A, Gallì A, Travaglino E, Brisci A, Rumi
E, Passamonti F, Invernizzi R, Cremonesi L, Boultwood J, Wainscoat JS, Hellström-Lindberg E, Cazzola M:
thrombocytosis. Blood 2009, 114:3538–3545. 61.
RI PT
Molecular and clinical features of refractory anemia with ringed sideroblasts associated with marked
Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adélaïde J, Rey J, Vainchenker W, Bernard
OA, Chaffanet M, Vey N, Birnbaum D, Mozziconacci MJ: Mutations of ASXL1 gene in myeloproliferative
Barosi G, Bergamaschi G, Marchetti M, Vannucchi AM, Guglielmelli P, Antonioli E, Massa M,
M AN U
62.
SC
neoplasms. Leukemia 2009, 23:2183–2186.
Rosti V, Campanelli R, Villani L, Viarengo G, Gattoni E, Gerli G, Specchia G, Tinelli C, Rambaldi A, Barbui T: JAK2 V617F mutational status predicts progression to large splenomegaly and leukemic transformation in primary myelofibrosis. Blood 2007, 110:4030 - 4036. 63.
Campbell PJ, Griesshammer M, Döhner K, Döhner H, Kusec R, Hasselbalch HC, Larsen TS,
TE D
Pallisgaard N, Giraudier S, Le Bousse-Kerdilès M-C, Desterke C, Guerton B, Dupriez B, Bordessoule D, Fenaux P, Kiladjian J-J, Viallard J-F, Brière J, Harrison CN, Green AR, Reilly JT: V617F mutation in JAK2 is
64.
EP
associated with poorer survival in idiopathic myelofibrosis. Blood 2006, 107:2098 - 2100. Guglielmelli P, Barosi G, Specchia G, Rambaldi A, Lo Coco F, Antonioli E, Pieri L, Pancrazzi A,
AC C
Ponziani V, Delaini F, Longo G, Ammatuna E, Liso V, Bosi A, Barbui T, Vannucchi AM: Identification of patients with poorer survival in primary myelofibrosis based on the burden of JAK2V617F mutated allele. Blood 2009, 114:1477 - 1483. 65.
Tefferi A, Lasho TL, Huang J, Finke C, Mesa RA, Li CY, Wu W, Hanson CA, Pardanani A: Low
JAK2V617F allele burden in primary myelofibrosis, compared to either a higher allele burden or unmutated status, is associated with inferior overall and leukemia-free survival. Leukemia2008, 22:756.
ACCEPTED MANUSCRIPT
66.
Pardanani A, Guglielmelli P, Lasho TL, Pancrazzi A, Finke CM, Vannucchi AM, Tefferi A: Primary
myelofibrosis with or without mutant MPL: comparison of survival and clinical features involving 603
67.
RI PT
patients. Leukemia 2011, 25:1834–1839. Rumi E, Pietra D, Guglielmelli P, Bordoni R, Casetti I, Milanesi C, Sant’Antonio E, Ferretti V,
Pancrazzi A, Rotunno G, Severgnini M, Pietrelli A, Astori C, Fugazza E, Pascutto C, Boveri E, Passamonti F, De Bellis G, Vannucchi A, Cazzola M, Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano
SC
Malattie Mieloproliferative: Acquired copy-neutral loss of heterozygosity of chromosome 1p as a
molecular event associated with marrow fibrosis in MPL-mutated myeloproliferative neoplasms. Blood
68.
M AN U
2013, 121:4388–4395.
Schwaab J, Schnittger S, Sotlar K, Walz C, Fabarius A, Pfirrmann M, Kohlmann A, Grossmann V,
Meggendorfer M, Horny HP, Valent P, Jawhar M, Teichmann M, Metzgeroth G, Erben P, Ernst T, Hochhaus A, Haferlach T, Hofmann WK, Cross NCP, Reiter A: Comprehensive mutational profiling in
69.
TE D
advanced systemic mastocytosis. Blood 2013, 122:2460–2466. Jawhar M, Schwaab J, Schnittger S, Sotlar K, Horny H, Metzgeroth G, Müller N, Schneider S,
Naumann N, Walz C, Haferlach T, Valent P, Hofmann W-K, Cross NCP, Fabarius A, Reiter A: Molecular
EP
profiling of myeloid progenitor cells in multi-mutated advanced systemic mastocytosis identifies KIT
70.
AC C
D816V as a distinct and late event. Leukemia 2015, 29:1115–1122. Escribano, Luis; Alvarez-Twose, Ivan; Sanchez-Munoz, Laura; Garcia-Montero, andres; Nunez,
Rosa; Almeida, J; Jara-Acevedo, Maria; Teodosio, Teodosio; Garcia-Cosio, Monica; Bellas, Carmen; Orfao A: Prognosis in adult indolent systemic mastocytosis: A long- term study of the Spanish Network on Mastocytosis in a series of 145 patients. J Allergy Clin Immunol 2009, 124:514–521. 71.
Garcia-montero AC, Jara-acevedo M, Teodosio C, Sanchez ML, Nunez R, Prados A, Aldanondo I,
Sanchez L, Dominguez M, Botana LM, Sanchez-jimenez F, Sotlar K, Almeida J, Escribano L, Orfao A:
ACCEPTED MANUSCRIPT
NEOPLASIA KIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: a prospective study of the Spanish Network on Mastocytosis ( REMA ) in a series of
72.
RI PT
113 patients. 2016, 108:2366–2373. Soucie E, Hanssens K, Mercher T, Georgin-lavialle S, Damaj G, Livideanu C, Chandesris MO, Acin
Y, Letard S, Sepulveda P De, Hermine O, Bernard OA, Dubreuil P: Brief report In aggressive forms of
73.
SC
mastocytosis, TET2 loss cooperates with c-KITD816V to transform mast cells. 2012, 120:4846–4849. Meggendorfer M, Haferlach T, Alpermann T, Jeromin S, Haferlach C, Kern W, Schnittger S:
M AN U
Specific molecular mutation patterns delineate chronic neutrophilic leukemia, atypical chronic myeloid leukemia, and chronic myelomonocytic leukemia. Haematologica 2014, 99:e244–e246. 74.
Guglielmelli P, Pancrazzi A, Bergamaschi G, Rosti V, Villani L, Antonioli E, Bosi A, Barosi G,
Vannucchi AM, GIMEMA--Italian Registry of Myelofibrosis, MPD Research Consortium: Anaemia
75.
TE D
characterises patients with myelofibrosis harbouring Mpl mutation. Br J Haematol 2007, 137:244–247. Tefferi A, Jimma T, Sulai NH, Lasho TL, Finke CM, Knudson RA, McClure RF, Pardanani A: IDH
mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical
76.
EP
evidence for leukemogenic collaboration with JAK2V617F. Leukemia 2012, 26:475–480. Vannucchi AM, Lasho TL, Guglielmelli P, Biamonte F, Pardanani A, Pereira A, Finke C, Score J,
AC C
Gangat N, Mannarelli C, Ketterling RP, Rotunno G, Knudson RA, Susini MC, Laborde RR, Spolverini A, Pancrazzi A, Pieri L, Manfredini R, Tagliafico E, Zini R, Jones A, Zoi K, Reiter A, Duncombe A, Pietra D, Rumi E, Cervantes F, Barosi G, Cazzola M, Cross NCP, Tefferi A: Mutations and prognosis in primary myelofibrosis. Leukemia 2013, 27:1861–1869. 77.
Gambacorti-Passerini CB, Donadoni C, Parmiani A, Pirola A, Redaelli S, Signore G, Piazza V,
Malcovati L, Fontana D, Spinelli R, Magistroni V, Gaipa G, Peronaci M, Morotti A, Panuzzo C, Saglio G,
ACCEPTED MANUSCRIPT
Usala E, Kim DW, Rea D, Zervakis K, Viniou N, Symeonidis A, Becker H, Boultwood J, Campiotti L, Carrabba M, Elli E, Bignell GR, Papaemmanuil E, Campbell PJ, Cazzola M, Piazza R: Recurrent ETNK1
78.
RI PT
mutations in atypical chronic myeloid leukemia. Blood 2015, 125:499-503. Loh ML, Martinelli S, Cordeddu V, Reynolds MG, Vattikuti S, Lee CM, Wulfert M, Germing U,
Haas P, Niemeyer C, Beran ME, Strom S, Lübbert M, Sorcini M, Estey EH, Gattermann N, Tartaglia M:
myelomonocytic leukemia. Leuk Res 2005, 29:459–462.
Rafael Bejar, Papaemmanuil E, Haferlach T, Garcia-Manero G, Maciejewski JP, Sekeres MA,
M AN U
79.
SC
Acquired PTPN11 mutations occur rarely in adult patients with myelodysplastic syndromes and chronic
Walter MJ, Graubert TA, Cazzola M, Malcovati L, Ogawa S, Fenaux P, Hellstrom-Lindberg E, Kern W, Boultwood J, Pellagatti A, Bowen D, Tauro S, Groves MJ, Vyas P, Quek L, Nazha A, Thol F, Heuser M, Shih L-Y, Padron E, Sallman D, Komrokji RS, List AF, Santini V, Fontenay M, Campbell PJ, Tüchler H, Stevenson K, Neuberg DS, Greenberg P, Ebert BL: Somatic Mutations in MDS Patients Are Associated with Clinical
TE D
Features and Predict Prognosis Independent of the IPSS-R: Analysis of Combined Datasets from the International Working Group for Prognosis in MDS-Molecular Committee. Blood 2015, 126:907. 80.
Shih L-Y, Huang C-F, Lin T-L, Wu J-H, Wang P-N, Dunn P, Kuo M-C, Tang T-C: Heterogeneous
EP
patterns of CEBPalpha mutation status in the progression of myelodysplastic syndrome and chronic
81.
AC C
myelomonocytic leukemia to acute myelogenous leukemia. Clin Cancer Res 2005, 11:1821–1826. Ernst T, Chase A, Zoi K, Waghorn K, Hidalgo-Curtis C, Score J, Jones A, Grand F, Reiter A,
Hochhaus A, Cross NCP: Transcription factor mutations in myelodysplastic/myeloproliferative neoplasms. Haematologica 2010, 95:1473–1480. 82.
Lindsley RC, Saber W, Mar BG, Redd R, Wang T, Haagenson MD, Grauman P V., Hu Z-H, Spellman
SR, Lee SJ, Verneris MR, Hsu K, Fleischhauer K, Cutler C, Antin JH, Neuberg D, Ebert BL: Prognostic
ACCEPTED MANUSCRIPT
Mutations in Myelodysplastic Syndrome after Stem-Cell Transplantation. N Engl J Med 2017, 376:536– 547. Dicker F, Haferlach C, Sundermann J, Wendland N, Weiss T, Kern W, Haferlach T, Schnittger S:
RI PT
83.
Mutation analysis for RUNX1, MLL-PTD, FLT3-ITD, NPM1 and NRAS in 269 patients with MDS or secondary AML. Leukemia 2010, 24:1528–1532.
Kuendgen A, Seidler J, Lauseker M, Haferlach T, Schnittger S, Kohlmann A, Hildebrandt B, Müller-
SC
84.
Thomas C, Betz B, Haas R, Gattermann N, Royer-Pokora B, Götze K, Germing U: Prognostic Impact Of
85.
M AN U
Molecular Mutations In 182 Patients With Myelodysplastic Syndromes. Blood 2013, 122:2758. Kuo M-C, Liang D-C, Huang C-F, Shih Y-S, Wu J-H, Lin T-L, Shih L-Y: RUNX1 mutations are frequent
in chronic myelomonocytic leukemia and mutations at the C-terminal region might predict acute myeloid leukemia transformation. Leukemia. 2009, pp. 1426–1431.
Makishima H, Yoshida K, Nguyen N, Przychodzen B, Sanada M, Okuno Y, Ng KP, Gudmundsson
TE D
86.
KO, Vishwakarma B a, Jerez A, Gomez-Segui I, Takahashi M, Shiraishi Y, Nagata Y, Guinta K, Mori H, Sekeres M a, Chiba K, Tanaka H, Muramatsu H, Sakaguchi H, Paquette RL, McDevitt M a, Kojima S,
EP
Saunthararajah Y, Miyano S, Shih L-Y, Du Y, Ogawa S, Maciejewski JP: Somatic SETBP1 mutations in myeloid malignancies. Nat Genet 2013, 45:942–946. Fernandez-Mercado M, Pellagatti A, Di Genua C, Larrayoz MJ, Winkelmann N, Aranaz P, Burns A,
AC C
87.
Schuh A, Calasanz MJ, Cross NCP, Boultwood J: Mutations in SETBP1 are recurrent in myelodysplastic syndromes and often coexist with cytogenetic markers associated with disease progression. Br J Haematol 2013, 163:235–239. 88.
Hou HA, Kuo YY, Tang JL, Chou WC, Yao M, Lai YJ, Lin CC, Chen CY, Liu CY, Tseng MH, Huang CF,
Chiang YC, Lee FY, Liu MC, Liu CW, Huang SY, Ko BS, Wu SJ, Tsay W, Chen YC, Tien HF: Clinical
ACCEPTED MANUSCRIPT
implications of the SETBP1 mutation in patients with primary myelodysplastic syndrome and its stability during disease progression. Am J Hematol 2014, 89:181–186. Walter MJ, Shen D, Shao J, Ding L, White BS, Kandoth C, Miller CA, Niu B, McLellan MD, Dees ND,
RI PT
89.
Fulton R, Elliot K, Heath S, Grillot M, Westervelt P, Link DC, DiPersio JF, Mardis E, Ley TJ, Wilson RK, Graubert TA: Clonal diversity of recurrently mutated genes in myelodysplastic syndromes. Leukemia
90.
SC
2013, 27:1275–1282.
Patnaik MM, Lasho TL, Hodnefield JM, Knudson R a., Ketterling RP, Garcia-Manero G, Steensma
M AN U
DP, Pardanani A, Hanson C a., Tefferi A: SF3B1 mutations are prevalent in myelodysplastic syndromes with ring sideroblasts but do not hold independent prognostic value. Blood 2012, 119:569–572. 91.
Hong JY, Seo J-Y, Kim S-H, Jung HA, Park S, Kim K, Jung CW, Kim JS, Park JS, Kim H-J, Jang JH:
Mutations in the Spliceosomal Machinery Genes SRSF2, U2AF1, and ZRSR2 and Response to Decitabine
92.
TE D
in Myelodysplastic Syndrome. Anticancer Res 2015, 35:3081–3089.
Lindsley RC, Mar BG, Mazzola E, Grauman P V., Shareef S, Allen SL, Pigneux A, Wetzler M, Stuart
RK, Erba HP, Damon LE, Powell BL, Lindeman N, Steensma DP, Wadleigh M, DeAngelo DJ, Neuberg D,
EP
Stone RM, Ebert BL: Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015, 125:1367–1376.
Piazza R, Valletta S, Winkelmann N, Redaelli S, Spinelli R, Pirola A, Antolini L, Mologni L,
AC C
93.
Donadoni C, Papaemmanuil E, Schnittger S, Kim D-W, Boultwood J, Rossi F, Gaipa G, Martini D, Greta P, Celle D, Francia P, Jang HG, Fantin V, Bignell GR, Magistroni V, Haferlach T, Pogliani EM, Campbell PJ, Chase AJ, Tapper WJ, Cross NCP, Gambacorti-Passerini C: Recurrent SETBP1 mutations in atypical chronic myeloid leukemia. Nat Genet 2013, 45:18–24.
ACCEPTED MANUSCRIPT
94.
Damm F, Itzykson R, Kosmider O, Droin N, Renneville A, Chesnais V, Gelsi-Boyer V, de Botton S,
Vey N, Preudhomme C, Clavert A, Delabesse E, Park S, Birnbaum D, Fontenay M, Bernard O, Solary E:
and secondary acute myeloid leukemias. Leukemia 2013, 27:1401–1403. 95.
RI PT
SETBP1 mutations in 658 patients with myelodysplastic syndromes, chronic myelomonocytic leukemia
Laborde RR, Patnaik MM, Lasho TL, Finke CM, Hanson CA, Knudson RA, Ketterling RP, Pardanani
A, Tefferi A: SETBP1 mutations in 415 patients with primary myelofibrosis or chronic myelomonocytic
Patnaik M, Itzykson R, Lasho T, Kosmider O, Finke C, Hanson C, Knudson R, Ketterling R, Tefferi
M AN U
96.
SC
leukemia: independent prognostic impact in CMML. Leukemia 2013, 27:2100–2102.
A, Solary E: ASXL1 and SETBP1 mutations and their prognostic contribution in chronic myelomonocytic leukemia: a two-center study of 466 patients. Leukemia 2014, 28:2206–2212. 97.
Meggendorfer M, Bacher U, Alpermann T, Haferlach C, Kern W, Gambacorti-Passerini C,
Haferlach T, Schnittger S: SETBP1 mutations occur in 9% of MDS/MPN and in 4% of MPN cases and are
TE D
strongly associated with atypical CML, monosomy 7, isochromosome i(17)(q10), ASXL1 and CBL mutations. Leukemia 2013, 27:1852–1860.
Caudill JSC, Sternberg AJ, Li CY, Tefferi A, Lasho TL, Steensma DP: C-terminal nucleophosmin
EP
98.
mutations are uncommon in chronic myeloid disorders. Br J Haematol 2006, 133:638–641. Gelsi-Boyer V, Trouplin V, Adélaïde J, Bonansea J, Cervera N, Carbuccia N, Lagarde A, Prebet T,
AC C
99.
Nezri M, Sainty D, Olschwang S, Xerri L, Chaffanet M, Mozziconacci MJ, Vey N, Birnbaum D: Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol 2009, 145:788–800. 100.
Nazha A, Narkhede M, Radivoyevitch T, Seastone DJ, Patel BJ, Gerds AT, Mukherjee S, Kalaycio
M, Advani A, Przychodzen B, Carraway HE, Maciejewski JP, Sekeres MA: Incorporation of molecular data
ACCEPTED MANUSCRIPT
into the Revised International Prognostic Scoring System in treated patients with myelodysplastic syndromes. Leukemia 2016, 30:2214-2220. Bejar R, Lord A, Stevenson K, Bar-Natan M, Pérez-Ladaga A, Zaneveld J, Wang H, Caughey B,
RI PT
101.
Stojanov P, Getz G, Garcia-Manero G, Kantarjian H, Chen R, Stone RM, Neuberg D, Steensma DP, Ebert BL: TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients.
102.
SC
Blood 2014, 124:2705–2712.
Thol F, Friesen I, Damm F, Yun H, Weissinger EM, Krauter J, Wagner K, Chaturvedi A, Sharma A,
M AN U
Wichmann M, Göhring G, Schumann C, Bug G, Ottmann O, Hofmann WK, Schlegelberger B, Heuser M, Ganser A: Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes. J Clin Oncol 2011, 29:2499–2506. 103.
Thol F, Kade S, Schlarmann C, Löffeld P, Morgan M, Krauter J, Wlodarski MW, Kölking B,
Wichmann M, Görlich K, Göhring G, Bug G, Ottmann O, Niemeyer CM, Hofmann W-KK, Schlegelberger B,
TE D
Ganser A, Heuser M: Frequency and prognostic impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with myelodysplastic syndromes. Blood 2012, 119:3578–3584. Traina F, Visconte V, Elson P, Tabarroki A, Jankowska a M, Hasrouni E, Sugimoto Y, Szpurka H,
EP
104.
Makishima H, O’Keefe CL, Sekeres M a, Advani a S, Kalaycio M, Copelan E a, Saunthararajah Y, Olalla
AC C
Saad ST, Maciejewski JP, Tiu R V: Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia 2014, 28:78–87. 105.
Damm F, Chesnais V, Nagata Y, Yoshida K, Scourzic L, Okuno Y, Itzykson R, Sanada M, Shiraishi Y,
Gelsi-Boyer V, Renneville A, Miyano S, Mori H, Shih L-Y, Park S, Dreyfus F, Guerci-Bresler A, Solary E, Rose C, Cheze S, Prébet T, Vey N, Legentil M, Duffourd Y, de Botton S, Preudhomme C, Birnbaum D, Bernard OA, Ogawa S, Fontenay M, Kosmider O: BCOR and BCORL1 mutations in myelodysplastic syndromes and related disorders. Blood 2013, 122:3169–3177.
ACCEPTED MANUSCRIPT
106.
Abdel-Wahab O, Pardanani A, Patel J, Wadleigh M, Lasho T, Heguy A, Beran M, Gilliland DG,
Levine RL, Tefferi A: Concomitant analysis of EZH2 and ASXL1 mutations in myelofibrosis, chronic myelomonocytic leukemia and blast-phase myeloproliferative neoplasms. Leukemia 2011, 25:1200–
107.
RI PT
1202.
Kosmider O, Gelsi-Boyer V, Ciudad M, Racoeur C, Jooste V, Vey N, Quesnel B, Fenaux P, Bastie J-
N, Beyne-Rauzy O, Stamatoulas A, Dreyfus F, Ifrah N, de Botton S, Vainchenker W, Bernard OA,
SC
Birnbaum D, Fontenay M, Solary E, Groupe Francophone des Myélodysplasies: TET2 gene mutation is a
108.
M AN U
frequent and adverse event in chronic myelomonocytic leukemia. Haematologica 2009, 94:1676–1681. Papaemmanuil E, Cazzola M, Boultwood J, Malcovati L, Vyas P, Bowen D, Pellagatti A, Wainscoat
JS, Hellstrom-Lindberg E, Gambacorti-Passerini C, Godfrey AL, Rapado I, Cvejic A, Rance R, McGee C, Ellis P, Mudie LJ, Stephens PJ, McLaren S, Massie CE, Tarpey PS, Varela I, Nik-Zainal S, Davies HR, Shlien A, Jones D, Raine K, Hinton J, Butler AP, Teague JW, Baxter EJ, Score J, Galli A, Della Porta MG, Travaglino E,
TE D
Groves M, Tauro S, Munshi NC, Anderson KC, El-Naggar A, Fischer A, Mustonen V, Warren AJ, Cross NCP, Green AR, Futreal PA, Stratton MR, Campbell PJ: Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med 2011, 365:1384–1395.
Ricci C, Spinelli O, Salmoiraghi S, Finazzi G, Carobbio A, Rambaldi A: ASXL1 mutations in primary
EP
109.
110.
AC C
and secondary myelofibrosis. Br J Haematol 2012, 156:404–407. Walter MJ, Ding L, Shen D, Shao J, Grillot M, McLellan M, Fulton R, Schmidt H, Kalicki-Veizer J,
O’Laughlin M, Kandoth C, Baty J, Westervelt P, DiPersio JF, Mardis ER, Wilson RK, Ley TJ, Graubert T a: Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia 2011, 25:1153– 1158.
ACCEPTED MANUSCRIPT
111.
Stegelmann F, Bullinger L, Schlenk RF, Paschka P, Griesshammer M, Blersch C, Kuhn S, Schauer S,
Döhner H, Döhner K: DNMT3A mutations in myeloproliferative neoplasms. Leukemia 2011, 25:1217–
112.
RI PT
1219. Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C, Jones A V, Waghorn K, Zoi K, Ross FM,
Reiter A, Hochhaus A, Drexler HG, Duncombe A, Cervantes F, Oscier D, Boultwood J, Grand FH, Cross NCP: Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet
Nikoloski G, Langemeijer SMC, Kuiper RP, Knops R, Massop M, Tönnissen ERLTM, van der
M AN U
113.
SC
2010, 42:722–726.
Heijden A, Scheele TN, Vandenberghe P, de Witte T, van der Reijden B a, Jansen JH: Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet, 2010, 42:665– 667. 114.
Patnaik MM, Hanson CA, Hodnefield JM, Lasho TL, Finke CM, Knudson RA, Ketterling RP,
TE D
Pardanani A, Tefferi A: Differential prognostic effect of IDH1 versus IDH2 mutations in myelodysplastic syndromes: a Mayo Clinic study of 277 patients. Leukemia 2012, 26:101–105. DiNardo CD, Jabbour E, Ravandi F, Takahashi K, Daver N, Routbort M, Patel KP, Brandt M, Pierce
EP
115.
S, Kantarjian H, Garcia-Manero G: IDH1 and IDH2 mutations in myelodysplastic syndromes and role in
116.
AC C
disease progression. Leukemia 2016, 30:980–984. Thol F, Weissinger EM, Krauter J, Wagner K, Damm F, Wichmann M, Göhring G, Schumann C,
Bug G, Ottmann O, Hofmann W-K, Schlegelberger B, Ganser A, Heuser M: IDH1 mutations in patients with myelodysplastic syndromes are associated with an unfavorable prognosis. Haematologica 2010, 95:1668–1674.
ACCEPTED MANUSCRIPT
117.
Jin J, Hu C, Yu M, Chen F, Ye L, Yin X, Zhuang Z, Tong H: Prognostic value of isocitrate
dehydrogenase mutations in myelodysplastic syndromes: a retrospective cohort study and meta-
118.
RI PT
analysis. PLoS One 2014, 9:e100206. Kosmider O, Slama L, Dreyfus F, Quesnel B, Slama B, Vey N, Lacombe C, Solary E, Birnbaum D,
Bernard OA, Fontenay M: Mutations of IDH1 and IDH2 genes in early and accelerated phases of
119.
SC
myelodysplastic syndromes and MDS / myeloproliferative neoplasms. Leukemia 2010, 24:1094–1096. Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, Pieri L, Finke CM,
M AN U
Kilpivaara O, Wadleigh M, Mai M, McClure RF, Gilliland DG, Levine RL, Pardanani A, Vannucchi AM: IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia 2010, 24:1302–1309. 120.
Yonal-Hindilerden I, Daglar-Aday A, Hindilerden F, Akadam-Teker B, Yilmaz C, Nalcaci M, Yavuz
AS, Sargin D: The Clinical Significance of IDH Mutations in Essential Thrombocythemia and Primary
121.
TE D
Myelofibrosis. J Clin Med Res 2016, 8:29–39.
Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A: IDH1 and IDH2 mutation analysis
122.
EP
in chronic- and blast-phase myeloproliferative neoplasms. Leukemia 2010, 24:1146–1151. Lasho TL, Jimma T, Finke CM, Patnaik M, Hanson CA, Ketterling RP, Pardanani A, Tefferi A: SRSF2
AC C
mutations in primary myelofibrosis: significant clustering with IDH mutations and independent association with inferior overall and leukemia-free survival. Blood 2012, 120:4168–4171. 123.
Pardanani A, Lasho T, Finke C, Oh ST, Gotlib J, Tefferi A: LNK mutation studies in blast-phase
myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia 2010, 24:1713–1718.
ACCEPTED MANUSCRIPT
124.
Smith AE, Mohamedali AM, Kulasekararaj A, Lim Z, Gäken J, Lea NC, Przychodzen B, Mian SA,
Nasser EE, Shooter C, Westwood NB, Strupp C, Gattermann N, Maciejewski JP, Germing U, Mufti GJ: Next-generation sequencing of the TET2 gene in 355 MDS and CMML patients reveals low-abundance
125.
RI PT
mutant clones with early origins, but indicates no definite prognostic value. Blood 2010, 116:3923–3932. Kosmider O, Gelsi-Boyer V, Cheok M, Grabar S, Della-Valle V, Picard F, Viguié F, Quesnel B,
Beyne-Rauzy O, Solary E, Vey N, Hunault-Berger M, Fenaux P, Mansat-De Mas V, Delabesse E, Guardiola
SC
P, Lacombe C, Vainchenker W, Preudhomme C, Dreyfus F, Bernard OA, Birnbaum D, Fontenay M: TET2 mutation is an independent favorable prognostic factor in myelodysplastic syndromes (MDSs). Blood
126.
M AN U
2009, 114:3285–3291.
Grossmann V, Tiacci E, Holmes AB, Kohlmann A, Martelli MP, Kern W, Spanhol-Rosseto A, Klein
H-U, Dugas M, Schindela S, Trifonov V, Schnittger S, Haferlach C, Bassan R, Wells VA, Spinelli O, Chan J, Rossi R, Baldoni S, De Carolis L, Goetze K, Serve H, Peceny R, Kreuzer K-A, Oruzio D, Specchia G, Di
TE D
Raimondo F, Fabbiano F, Sborgia M, Liso A, Farinelli L, Rambaldi A, Pasqualucci L, Rabadan R, Haferlach T, Falini B: Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood 2011, 118:6153–6163. Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Massé A, Kosmider O, Le Couedic J-
EP
127.
AC C
P, Robert F, Alberdi A, Lécluse Y, Plo I, Dreyfus FJ, Marzac C, Casadevall N, Lacombe C, Romana SP, Dessen P, Soulier J, Viguié F, Fontenay M, Vainchenker W, Bernard OA: Mutation in TET2 in myeloid cancers. N Engl J Med 2009, 360:2289–2301. 128.
Tefferi A, Pardanani A, Lim K-H, Abdel-Wahab O, Lasho TL, Patel J, Gangat N, Finke CM,
Schwager S, Mullally A, Li C-Y, Hanson CA, Mesa R, Bernard O, Delhommeau F, Vainchenker W, Gilliland DG, Levine RL: TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009, 23:905–911.
ACCEPTED MANUSCRIPT
129.
Tefferi A, Levine RL, Lim K-H, Abdel-Wahab O, Lasho TL, Patel J, Finke CM, Mullally A, Li C-Y,
Pardanani A, Gilliland DG: Frequent TET2 mutations in systemic mastocytosis: clinical, KITD816V and
130.
RI PT
FIP1L1-PDGFRA correlates. Leukemia 2009, 23:900–904. Papaemmanuil E, Malcovati L, Cazzola M, Hellstrom-Lindberg E, Bowen D, Boultwood JB, Green
AR, Futreal PA, Stratton MR, Campbell PJ: Identification of Novel Somatic Mutations in SF3B1, a Gene
Other Common Cancers. Eur. J. Cancer. 2011, p. 7.
Damm F, Thol F, Kosmider O, Kade S, Löffeld P, Dreyfus F, Stamatoullas-Bastard A, Tanguy-
M AN U
131.
SC
Encoding a Core Component of RNA Splicing Machinery, in Myelodysplasia with Ring Sideroblasts and
Schmidt A, Beyne-Rauzy O, de Botton S, Guerci-Bresler A, Göhring G, Schlegelberger B, Ganser A, Bernard OA, Fontenay M, Heuser M: SF3B1 mutations in myelodysplastic syndromes: clinical associations and prognostic implications. Leukemia. 2012, pp. 1137–1140. 132.
Makishima H, Visconte V, Sakaguchi H, Jankowska AM, Kar SA, Jerez A, Przychodzen B, Bupathi
TE D
M, Guinta K, Afable MG, Sekeres MA, Padgett RA, Tiu R V., Maciejewski JP: Mutations in the spliceosome machinery, a novel and ubiquitous pathway in leukemogenesis. Blood 2012, 119:3203–3210. Thota S, Viny AD, Makishima H, Spitzer B, Radivoyevitch T, Przychodzen B, Sekeres MA, Levine
EP
133.
RL, Maciejewski JP: Genetic alterations of the cohesin complex genes in myeloid malignancies. Blood
134.
AC C
2014, 124:1790–1798.
Lasho TL, Finke CM, Hanson CA, Jimma T, Knudson RA, Ketterling RP, Pardanani A, Tefferi A:
SF3B1 mutations in primary myelofibrosis: clinical, histopathology and genetic correlates among 155 patients. Leukemia 2011, 26:1135. 135.
Damm F, Kosmider O, Gelsi-Boyer V, Renneville A, Carbuccia N, Hidalgo-Curtis C, Della Valle V,
Couronné L, Scourzic L, Chesnais V, Guerci-Bresler A, Slama B, Beyne-Rauzy O, Schmidt-Tanguy A,
ACCEPTED MANUSCRIPT
Stamatoullas-Bastard A, Dreyfus F, Prébet T, de Botton S, Vey N, Morgan M a, Cross NCP, Preudhomme C, Birnbaum D, Bernard O a, Fontenay M, Groupe Francophone des Myélodysplasies: Mutations
myelodysplastic syndromes. Blood 2012, 119:3211–3218. 136.
RI PT
affecting mRNA splicing define distinct clinical phenotypes and correlate with patient outcome in
Graubert T a, Shen D, Ding L, Okeyo-Owuor T, Lunn CL, Shao J, Krysiak K, Harris CC, Koboldt DC,
Larson DE, McLellan MD, Dooling DJ, Abbott RM, Fulton RS, Schmidt H, Kalicki-Veizer J, O’Laughlin M,
SC
Grillot M, Baty J, Heath S, Frater JL, Nasim T, Link DC, Tomasson MH, Westervelt P, DiPersio JF, Mardis
syndromes. Nat Genet 2011, 44:53–57. 137.
M AN U
ER, Ley TJ, Wilson RK, Walter MJ: Recurrent mutations in the U2AF1 splicing factor in myelodysplastic
Wu S-JJ, Tang J-LL, Lin C-TT, Kuo Y-YY, Li L-YY, Tseng M-HH, Huang C-FF, Lai Y-JJ, Lee F-YY, Liu M-
CC, Liu C-WW, Hou H-AA, Chen C-YY, Chou W-CC, Yao M, Huang S-YY, Ko B-SS, Tsay W, Tien H-FF: Clinical implications of U2AF1 mutation in patients in myelodysplastic syndrome and its stability during disease
138.
TE D
progression. Am J Hematol 2013, 88:E277-82.
Kon A, Shih L-Y, Minamino M, Sanada M, Shiraishi Y, Nagata Y, Yoshida K, Okuno Y, Bando M,
Nakato R, Ishikawa S, Sato-Otsubo A, Nagae G, Nishimoto A, Haferlach C, Nowak D, Sato Y, Alpermann T,
EP
Nagasaki M, Shimamura T, Tanaka H, Chiba K, Yamamoto R, Yamaguchi T, Otsu M, Obara N, Sakata-
AC C
Yanagimoto M, Nakamaki T, Ishiyama K, Nolte F, Hofmann W-K, Miyawaki S, Chiba S, Mori H, Nakauchi H, Koeffler HP, Aburatani H, Haferlach T, Shirahige K, Miyano S, Ogawa S: Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms. Nat Genet 2013, 45:1232–1237. 139.
Jädersten M, Saft L, Smith A, Kulasekararaj A, Pomplun S, Göhring G, Hedlund A, Hast R,
Schlegelberger B, Porwit A, Hellström-Lindberg E, Mufti GJ: TP53 Mutations in Low-Risk Myelodysplastic Syndromes With del(5q) Predict Disease Progression. J Clin Oncol2011, 29:1971–1979.
ACCEPTED MANUSCRIPT
140.
Kita-Sasai Y, Horiike S, Misawa S, Kaneko H, Kobayashi M, Nakao M, Nakagawa H, Fujii H,
Taniwaki M: International prognostic scoring system and TP53 mutations are independent prognostic
141.
RI PT
indicators for patients with myelodysplastic syndrome. Br J Haematol 2001, 115:309–312. Mckerrell T, Park N, Moreno T, Grove CS, Ponstingl H, Stephens J, Group USS, Crawley C, Craig J,
Scott M, Hodkinson C, Baxter J, Rad R, Forsyth D, Quail M, Zeggini E, Ouwehand W, Varela I, Vassiliou GS: Leukemia-Associated Somatic Mutations Drive Distinct Patterns of Age-Related Clonal Hemopoiesis. Cell
Lindberg J, Ph D, Rose S a, Bakhoum SF, Ph D, Chambert K, Mick E, Neale BM, Ph D, Fromer M,
M AN U
142.
SC
Rep 2015, 10:1239–1245.
Ph D, Purcell SM, Ph D, Svantesson O, Sullivan PF, Sklar P, Ph D, Grönberg H, Ph D, Hultman CM, Ph D, Mccarroll S a, Ph D: Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N Engl J Med 2014, 371:2477–2487. 143.
Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, Ebert BL: Clonal
2015, pp. 9–16.
Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S,
EP
144.
TE D
hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood
Ardissino D, Baber U, Mehran R, Fuster V, Danesh J, Frossard P, Saleheen D, Melander O, Sukhova GK,
AC C
Neuberg D, Libby P, Kathiresan S, Ebert BL: Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med 2017, 377:111–121. 145.
Young AL, Challen GA, Birmann BM, Druley TE: Clonal haematopoiesis harbouring AML-
associated mutations is ubiquitous in healthy adults. Nat Commun 2016, 7:12484.
ACCEPTED MANUSCRIPT
146.
Matsunawa M, Yamamato R, Sanada M, Sato-Otsubo A, Shiozawa Y, Yoshida K, Otsu M, Shiraishi
Y, Miyano S, Isono K: Haploinsufficiencyof Sf3b1 leads to compromised stem cell function but not
147.
RI PT
myelodysplasia. Leukemia 2014, 28:1844–1850. Malcovati L, Gallì A, Travaglino E, Ambaglio I, Rizzo E, Molteni E, Elena C, Ferretti VV, Catricalà S,
Bono E, Todisco G, Bianchessi A, Rumi E, Zibellini S, Pietra D, Boveri E, Camaschella C, Toniolo D,
Papaemmanuil E, Ogawa S, Cazzola M: Clinical significance of somatic mutation in unexplained blood
Mossner M, Jann J, Wittig J, Nolte F, Fey S, Nowak V, Obl J, Pressler J, Palme I, Xanthopoulos C,
M AN U
148.
SC
cytopenia. Blood, American Society of Hematology, 2017, 129:3371–3378.
Boch T, Metzgeroth G, Henning R, Witt SH, Dukal H, Klein C, Schmitt S, Gelß P, Platzbecker U, Balaian E, Fabarius A, Blum H, Schulze TJ, Meggendorfer M, Haferlach C, Trumpp A, Wolf-Karsten H, Medyouf H, Nowak D: Mutational hierarchies in myelodysplastic syndromes dynamically adapt and evolve upon therapy response and failure. Blood 2016, 128:1246–1259.
Greenberg PL: The multifaceted nature of myelodysplastic syndromes: Clinical, molecular, and
TE D
149.
biological prognostic features. J Natl Compr Cancer Netw 2013, 11:877–885. Chesnais V, Renneville A, Toma A, Lambert J, Passet M, Dumont F, Chevret S, Lejeune J,
EP
150.
Raimbault A, Stamatoullas A, Rose C, Beyne-Rauzy O, Delaunay J, Solary E, Fenaux P, Dreyfus F,
AC C
Preudhomme C, Kosmider O, Fontenay M: Effect of lenalidomide treatment on clonal architecture of myelodysplastic syndromes without 5q deletion. Blood 2016, 127:749–760. 151.
Gu J, Wang Z, Xiao M, Mao X, Zhu L, Wang Y, Huang W: Chronic myelomonocytic leukemia with
double-mutations in DNMT3A and FLT3-ITD treated with decitabine and sorafenib. Cancer Biol Ther 2017, 4047:00–00.
ACCEPTED MANUSCRIPT
152.
Kosmider O, Chapuis N, Kaltenbach S, Coriat R, Boudou Rouquette P, Willems L, Chesnais V,
Radford-Weiss I, Bardet V, Mayeux P, Tamburini J, Fontenay M, Bouscary D: Sustained Leukemia-Free State and Molecular Response to Sorafenib in a Patient With Chronic Myelomonocytic Leukemia in
RI PT
Transformation Driven by Homozygous FLT3-ITD Malignant Hematopoiesis. Clin Lymphoma, Myeloma Leukemia 2018, 13:347–350.
Alfonso A, Montalban-Bravo G, Garcia-Manero G: Current management of patients with chronic
myelomonocytic leukemia. Curr Opin Oncol 2017, 29:79–87.
Scott LM, Scott MA, Campbell PJ, Green AR: Progenitors homozygous for the V617F mutation
M AN U
154.
SC
153.
occur in most patients with polycythemia vera, but not essential thrombocythemia. Blood 2006, 108:2435–2437. 155.
Ortmann CA, Kent DG, Nangalia J, Silber Y, Wedge DC, Grinfeld J, Baxter EJ, Massie CE,
Papaemmanuil E, Menon S, Godfrey AL, Dimitropoulou D, Guglielmelli P, Bellosillo B, Besses C, Döhner K,
TE D
Harrison CN, Vassiliou GS, Vannucchi A, Campbell PJ, Green AR: Effect of mutation order on myeloproliferative neoplasms. N Engl J Med 2015, 372:601–612. Campbell PJ, Scott LM, Buck G, Wheatley K, East CL, Marsden JT, Duffy A, Boyd EM, Bench AJ,
EP
156.
Scott MA, Vassiliou GS, Milligan DW, Smith SR, Erber WN, Bareford D, Wilkins BS, Reilly JT, Harrison CN,
AC C
Green AR, United Kingdom Myeloproliferative Disorders Study Group, Medical Research Council Adult Leukaemia Working Party, Australasian Leukaemia and Lymphoma Group: Definition of subtypes of essential thrombocythaemia and relation to polycythaemia vera based on JAK2 V617F mutation status: a prospective study. Lancet (London, England) 2005, 366:1945–1953. 157.
Lehmann U, Bartels S, Hasemeier B, Geffers R, Schlue J, Busche G, Hussein K, Kreipe H: SRSF2
mutation is present in the hypercellular and prefibrotic stage of primary myelofibrosis. Blood 2015, 121:4011–4012.
ACCEPTED MANUSCRIPT
158.
Chen E, Beer PA, Godfrey AL, Ortmann CA, Li J, Costa-Pereira AP, Ingle CE, Dermitzakis ET,
Campbell PJ, Green AR: Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1
159.
RI PT
signaling. Cancer Cell 2010, 18:524–535. Pandey R, Kapur R: Targeting phosphatidylinositol-3-kinase pathway for the treatment of
Philadelphia-negative myeloproliferative neoplasms. Mol Cancer, Molecular Cancer, 2015, 14:118. Geyer HL, Mesa RA: Therapy for myeloproliferative neoplasms: when, which agent, and how?
Hematol Am Soc Hematol Educ Progr 2014, 2014:277–286.
Gotlib J, Maxson JE, George TI, Tyner JW: The new genetics of chronic neutrophilic leukemia and
M AN U
161.
SC
160.
atypical CML: Implications for diagnosis and treatment. Blood 2013, 122:1707–1711. 162.
Maxson JE, Tyner JW: Genomics of chronic neutrophilic leukemia. Blood 2016, 129:blood-2016-
10-695981.
Pardanani A, Lasho TL, Laborde RR, Elliott M, Hanson C a, Knudson RA, Ketterling RP, Maxson JE,
TE D
163.
Tyner JW, Tefferi a: CSF3R T618I is a highly prevalent and specific mutation in chronic neutrophilic leukemia. Leukemia 2013, 27:1870–1873.
Maxson JE, Gotlib J, Pollyea D a, Fleischman AG, Agarwal A, Eide C a, Bottomly D, Wilmot B,
EP
164.
AC C
McWeeney SK, Tognon CE, Pond JB, Collins RH, Goueli B, Oh ST, Deininger MW, Chang BH, Loriaux MM, Druker BJ, Tyner JW: Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML. N Engl J Med 2013, 368:1781–1790. 165.
Wang SA, Tam W, Tsai A, Arber D, Hasserjian R, Geyer J, George T, Czuchlewski D, Foucar K,
Rogers H, Hsi E, Bryan Rea B, Bagg A, Dal Cin P, Zhao C, Kelley TW, Verstovsek S, Bueso-Ramos C, Orazi A: Targeted next-generation sequencing identifies a subset of idiopathic hypereosinophilic syndrome with
ACCEPTED MANUSCRIPT
features similar to chronic eosinophilic leukemia, not otherwise specified. Mod Pathol 2016, 29:854– 864. Shah NP, Lee FY, Luo R, Jiang Y, Donker M, Akin C: Dasatinib (BMS-354825) inhibits KIT D816V ,
RI PT
166.
an imatinib-resistant activating mutation that triggers neoplastic growth in most patients with systemic mastocytosis. Blood 2006, 108:286–291.
Zermati Y, Sepulveda P De, Feger F, Letard S, Kersual J, Casteran N, Bieche Y, Vivaud M,
SC
167.
Lortholary O, Hermine O, Dubreuil P: Effect of tyrosine kinase inhibitor STI571 on the kinase activity of
M AN U
wild-type and various mutated c-kit receptors found in mast cell neoplasms. Oncogene 2003, 22:660– 664. 168.
Verstovsek S: Advanced systemic mastocytosis: the impact of KIT mutations in diagnosis,
AC C
EP
TE D
treatment, and progression. Eur J Haematol 2012, 90:89–98.
ACCEPTED MANUSCRIPT
FIGURE LEGENDS Figure 1: Overview of cellular processes with components reported to have coding gene variants in
RI PT
chronic myeloid neoplasms (CMNs). External growth factors and cytokines bind and activate their respective transmembrane receptors, triggering activation of pathways (predominantly composed of kinase cascade proteins) leading to activated key intermediaries such as ERK, AKT1, TP53, and
mTORC1or2. These intermediaries interact with many targets located throughout the cell until signals
SC
reach effectors (such a transcription factors, mitochondrial proteins, structural and transport proteins) that ultimately facilitate the major cell functions of proliferation, differentiation, survival, or apoptosis.
M AN U
Signaling through JAK/STAT/ERK is most commonly used by the three main cytokine receptors known to be important for normal myeloid hematopoiesis; erythropoietin receptor (EPOR), predominantly for development of erythrocytes, thrombopoetin receptor (TPOR) (gene called MPL), predominantly for development of megakaryocytes, and colony stimulating factor 3 receptor (CSF3R), predominantly for
TE D
development of neutrophils. RUNX1 is a master hematopoietic transcriptional regulator, involved both in the formation of hematopoietic stem cells and differentiation, the latter with fine-tuning assistance by CEBPA. Core binding factor beta (CBFB) binds RUNX1 to enhance its DNA affinity but does not directly
EP
interact with the DNA. ETV6 is a strong transcriptional repressor, and is also implicated in interactions with corepressors that recruit histone deacetylases, such as NCOR2. TP53 is activated by diverse forms
AC C
of stress (cytokine deprivation, DNA damage, others) and is the main pro-apoptotic proponent in the cell’s continual balance of pro-apoptotic and anti-apoptotic factors. TP53 induces the expression of PPM1D (not shown), a phosphatase that negatively regulates the MAPK pathway and consequently p53mediated transcription and apoptosis. It initiates cell cycle arrest and activates pro-apoptotic members of the BCL2 family in the mitochondria through direct and indirect mechanisms, the latter through its role as a transcription factor, whereby it increases transcription of pro-apoptotic BCL2 family members and directly represses transcription of anti-apoptotic family members. The cohesion complex (SMC1,
ACCEPTED MANUSCRIPT
SMC3, RAD21, STAG) forms a ring-like structure that encircles DNA without directly binding it, providing sister chromatid cohesion during processes that require DNA looping such as transcription, meiosis, mitosis, recombination, and ribosomal biogenesis (Supplemental Figure S1). Nucleophosmin (NPM1)
RI PT
shuttles proteins between the nucleus and cytoplasm, playing a key role in ribosomal biogenesis, cell cycle regulation (such as maintaining the stability and function of p53), activation of cyclin-dependent kinases, and regulation of the mitotic spindle. Old or dysfunctional proteins may be disposed off through
SC
ubiquitin tagging (Supplemental Figure S2) followed by degradation, either in the proteasome or
through autophagy. The protein substrates affected by the CBL family include a wide range of tyrosine
M AN U
kinases acting in signal transduction pathways, including KIT, FLT3, and JAK2. SET and its binding partner, SETBP1, play poorly elucidated roles in hematopoiesis, although both are known to have oncogenic properties when functionally abnormal. Similarly, the role of PHF6 (not shown) is poorly understood but appears to act as a tumor suppressor, regulating transcription of signaling genes and rRNA. CALR is a
TE D
multi-compartmental protein (extracellular matrix, outer cell surface, cytosol, endoplasmic reticulum (ER), nucleus) that regulates a wide array of cellular processes including, among others, normal and abnormal protein movement through the ER, calcium signaling, and transcription modulation. However,
EP
variant CALR (CALRv) functions in a different manner than the wildtype, traveling with TPOR from the ER to the cell surface and activating the JAK/STAT pathway. For detailed information on the alterations
AC C
noted in this figure in myeloid neoplasia, see Supplemental Tables S1, S2, and S5.
Figure 2: Epigenetic modification of DNA and histones with an emphasis on components reported to have coding gene variants in chronic myeloid neoplasms (CMNs). Epigenetics broadly encompasses processes that impact gene expression without altering the underlying DNA sequence and includes methylation of both DNA and histones. DNA methylation is mediated by DNA methyltransferases (DNMTs) and occurs at cytosine residues of CpG dinucleotides located within CG-rich regions of gene
ACCEPTED MANUSCRIPT
promoters. DNMT1 is responsible for maintaining patterns of methylation during DNA replication whereas DNMT3A and DNMT3B are de novo methyltransferases, both of which are expressed in hematopoietic stem cells and involved in self-renewal and differentiation, though only DNMT3A is
RI PT
expressed in more mature elements. Cytosine methylation results in recruitment of transcriptional repressors and gene silencing. Cyotsine methylation marks can be lost during DNA replication or
actively removed by the TET family proteins, including TET2. TET enzyme activity is inhibited by 2-
SC
hydroxyglutarate (2-HG) which is generated by the variant form of isocitrate dehydrogenase (IDH), either from the cytosolic gene IDH1, or the mitochondrial homolog, IDH2. Histone methylation alters
M AN U
chromatin structure, directly affecting accessibility to transcriptional activators and repressors and allowing recruitment of additional epigenetic regulators such as DNMTs. The polycomb repressor complex 2 (PRC2) consists of four core members (JARID2, EED, SUZ12, and EZH1 or EXH2) and silences chromatin by methylating histone H3 with activity further augmented by ASXL1. BCOR is another
TE D
polycomb repressor complex (comprised of BCOR, BCORL1, and others), which represses transcription by ubiquitylating histone H2A. Opposing the action of the polycomb repressor complexes are a variety of histone deubiquitinases and histone demethylases, including KDM6A. For detailed information on the
EP
alterations noted in this figure in myeloid neoplasia, see Supplemental Table S3.
AC C
Figure 3: Spliceosome mechanisms with an emphasis on components reported to have coding gene variants in chronic myeloid neoplasms (CMNs). For genes containing more than one exon (>90% of all genes), excision of introns through splicing of mRNA is required prior to translation. The primary machinery required for this process is a complex of five small nuclear RNAs (snRNAs) coupled with over 150 small nuclear ribonucleic proteins (snRNPs), collectively termed the spliceosome.6 Two basic types of splice site sequences are known that recruit slightly different versions of spliceosome: The major splice sequence (used for approximately 99% of splices) recruits the major spliceosome (comprised of
ACCEPTED MANUSCRIPT
snRNPs U1, U2, U4, U5, U6). Intronic material to be excised is bookended by splice sites at their 5' and 3' ends (5'ss and 3'ss, respectively). The 5'ss is characterize by a GU sequence whereas the 3' by AG. Between these sites are two additional key recognition sequences, a conserved adenosine, the branch
RI PT
point site (BPS) located between the two ends, and a polypyrimidine tract (PPT) located just 5' of the 3'ss. The initial step in the splicing process is the recognition of the 5'ss by U1 snRNP. Simultaneously, there is recognition of the PPT by the serine-arginine (SR)-rich splicing factors SRSF1 or SFSR2 as well as
SC
the U2 auxiliary factor, U2AF2 (formerly U2AF65). In addition, U2AF1 (formerly U2AF35) recognizes the AG of the 3'ss along with ZRSR2, an SR factor with zinc finger activity. Together these form the early (E)
M AN U
complex. The U2 auxiliary factors guide the U2 snRNP to the BPS to form complex A. Binding of the U2 snRNA to the BPS, regulated by the SF3a (subunits 1 to 3) and SF3b (subunits 1 to 6) complexes, creates a bulge by a single base non-complementarity that exposes the 2'-hydroxyl of the conserved adenosine for the transesterification to the 5'-phosphate of the guanosine adjacent to the end of the 5' exon. The
TE D
complex U4/U5/U6 tri-snRNP is then recruited through interaction of U5 to the 3'ss, resulting in an initial B complex, and ultimately forming B* complex, the active spliceosome, with loss of U1 and U4. After the first catalytic step, the entirety is termed the C complex. The final transesterification results in
EP
fusion of the two exons and release of the intron in a lariat configuration. For detailed information on
AC C
the alterations noted in this figure in myeloid neoplasia, see Supplemental Table S4.
Figure 4: Heat map of incidence and prognostic significance of gene variants by chronic myeloid neoplasms (CMNs) entity (heatmap meant to be viewed in color). Commonly mutated genes in CMNs are listed with their overall incidence as the number in each box. The color-coding represents a synthesized, overall view of the clinical value of variants in each gene. Acccordingly, the figure is best viewed in color. To create the heatmap, key prognostic matrix points (progression, including fibrosis and transformation to acute leukemia, leukemia-free survival (LFS), overall survival (OS)) were given a score
ACCEPTED MANUSCRIPT
from 1 to 3 as follows: Unclear- no convincing reports OR contradictory reports OR <5 patients reported (labeled as “unclear” prognostically); Score 1 - ≤2 papers OR multiple contradictory reports OR <25 patients reported; Score 2 - ≥2 papers AND no contradictory reports AND ≥25 patients reported; Score 3
RI PT
- ≥4 papers AND no contradictory reports AND ≥100 patients reported. Of note, the papers used to generate the score could not all be referenced in this manuscript due to space limitations. *Several studies reviewed IDH1 and IDH2 mutations together and their conclusions are listed here. †The
SC
percentages for CMML and aCML may reflect the inclusion of cases of CNL whereas other references have suggested that CSF3R may be highly sensitive and specific for CNL. ‡These ranges for JAK2
M AN U
incidence in PV reflect the incidence of the JAK2 p.V617F mutation only. In addition, the prognostic designation for JAK2 in PV, ET, and PMF is related largely to high VAF JAK2 mutations, not the presence of a JAK2 mutation itself. §The incidence of KIT mutations in SM reflects the incidence in mast cells and not necessarily the incidence in all marrow compartments; particularly in SM with an associated
TE D
hematopoietic neoplasm. ¶SF3B1 mutations are found in 10% to 33% of cases of MDS overall, but are ǁ
present in 50% to 83% of cases of MDS with ring sideroblasts. PPM1D prognosis (shaded for MDS only) and frequency is only related to the high incidence of PPM1D mutations found in CHIP that is associated
EP
with therapy-related myeloid neoplasms, in general, after autologous stem cell transplantation. MDS - myelodysplastic syndrome; CMML - chronic myelomonocytic leukemia; aCML - atypical chronic
AC C
myelogenous leukemia; MDS/MPN-RS-T - myelodysplastic/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis; ET - essential thrombocythemia; PV - polycythemia vera; PMF primary myelofibrosis; SM - systemic mastocytosis.
Figure 5: Schematic of common changes acquired in the differentiation and progression of chronic myeloid neoplasms (CMNs). Although many and varied passenger variants are evident at the time of development of CMNs, the founder mutations in CMNs are enriched for variants in genes involved with
ACCEPTED MANUSCRIPT
epigenetic regulation and the spliceosome. TET2 and ASXL1 are particularly prevalent in the former category. In the latter category, the four main spliceosome genes (SRSF2, SF3B1, U2AF1, and ZRSR2) are commonly recurrent in MDS whereas only SRSF2 is particularly prevalent in all CMNs and SF3B1 in
RI PT
entities involving ring sideroblasts. Although this base appears to characterize MDS, in other CMNs additional secondary variants are recurrently identified and confer the disease-defining features of those entities, such as CSF3R in CNL or JAK2 and CALR in the non-CML MPNs. Finally, during disease
SC
progression, many CMNs acquire additional variants in TP53 or SETBP1. Specifically, in MDS, additional variants in signal transduction pathway members (such as NRAS, KRAS, FLT3, and KIT), myeloid
M AN U
transcription regulators (such as RUNX1 or ETV6), or additional epigenetic regulators (such as IDH1/2, DNMT3A, and EZH2) are common findings in more high-risk disease or secondary AML. In the non-CML MPNs, biallelic or LOH of the JAK2 lead to high VAFs in association with progressive polycythemia and fibrosis. x, variable number of passenger mutations; y, founder mutation(s); z, secondary mutation(s); n,
AC C
EP
TE D
progression nutation(s).
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
Signaling
Splicing
AC C
Other
PPM1Dll RUNX1 SETBP1 TP53 NPM1
7 3‐10 1‐2 10‐33¶ 12‐33 5‐17 3‐11 <1 2‐3 2‐3 15 8‐20 2‐4 5‐18 1‐4
20‐66
10 24
Rare 13‐20
12‐17 12‐25 0‐6
Rare 30‐41 3 8 † <5 4‐8
26 13‐15
50‐100
ET
PV
PMF
SM
4‐25
7
12‐75
21‐29
<1 <1 5‐21 16‐73 2
RI PT
MDS/MPN‐ RS‐T
3‐7 3 <1 2
7‐36 0
SC
10 22‐60 3‐7 2 2‐13 6‐13 <1‐3 1‐10 0‐11 22‐61 3 10‐22 † 0‐8 1‐10 <1‐11 7‐18 6 1‐7 4‐16 1 4‐10 28‐52 5‐15 8‐10 4‐20
aCML
3‐7 6‐13 0.5‐5 3‐5 0‐4 10‐50 23‐60 6
2‐75
23‐100‡
25‐79
1‐8
0
3‐9 7 3‐5
10
1‐23
27‐30 Rare
72‐87
1
2‐3
2‐7 9‐17
40 13
12 5‐10
0‐4 20‐29 4
5 100§ 10
5 3 36‐43 4‐5
4
3‐4
4
7‐37 4‐16 <1‐4 1‐6
EP
Transcription Factor
0‐2 3‐4 1‐10 <1‐10
CMML 6‐10
M AN U
Epigenetics
MDS 8‐15 <1 1 5‐15 5‐46 4‐6 <1‐3 3‐13 3‐11 1‐3 4‐12 3‐9 13‐37 8 2‐5
TE D
Cohesin
Gene Cohesins RAD21 SMC3 STAG2 ASXL1 BCOR BCORL1 DNMT3A EZH2 IDH1 IDH1/2* IDH2 TET2 CALR CBL CSF3R FLT3 JAK2 KIT KRAS MPL NF1 NRAS PTPN11 SF3B1 SRSF2 U2AF1 ZRSR2 CEBPA ETV6 PHF6
6 24‐32 9 <1
Good 3 2 1 Unclear
Bad
No Change
1
3 3
23 3