Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

JAK/STAT INHIBITION AND BEYOND IN Ph-NEGATIVE MPNs

Molecular prognostication in Ph-negative


MPNs in 2022

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


Alessandro Maria Vannucchi and Paola Guglielmelli
CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, Azienda Ospedaliero­Universitaria Careggi,
Florence, Italy

The application of genomic techniques, including cytogenetics and DNA sequencing, to decipher the molecular land-
scape of patients with myeloproliferative neoplasms (MPNs) has radically modified diagnostic approach and man-
agement through improved risk stratification. Three driver mutated genes (JAK2, MPL, CALR) are variably harbored
by >80% of patients and associated with clinical characteristics, as well as major disease-related complications and
different survival outcomes. Therefore, JAK2 V617F mutation is included in the revised International Prognosis Score
of Thrombosis for Essential Thrombocythemia score for prediction of thrombosis in patients with essential throm-
bocythemia and prefibrotic primary myelofibrosis, while a CALR type 1 mutated genotype constitutes a favorable
variable for survival in patients with myelofibrosis (MF). Novel, integrated clinical and cytogenetic/mutation scores
(Mutation-Enhanced International Prognostic Score System for Transplantation-Age Patients with Primary Myelofi-
brosis [MIPSS70/v2], genetically inspired prognostic scoring system [GIPSS], Myelofibrosis Secondary to PV and ET-
Prognostic Model [MYSEC-PM]) have been devised that guide selection of stem cell transplantation candidates with
MF or help predict the risk associated with the transplant procedure (Myelofibrosis Transplant Scoring System), with
greater performance compared with conventional scores based on hematologic and clinical variables only. On the
other hand, several clinical needs remain unmet despite the great amount of molecular information available nowa-
days. These include the prediction of evolution to acute leukemia in a clinically actionable time frame, the identifi-
cation of patients most likely to derive durable benefits from target agents, in primis JAK inhibitors, and, conversely,
the significance of molecular responses that develop in patients receiving interferon or some novel agents. Here, we
discuss briefly the significance and the role of genomic analysis for prognostication in patients with MPNs from a clini-
cian’s point of view, with the intent to provide how-to-use hints.

LEARNING OBJECTIVES
• To appreciate the variety of abnormalities in the cytogenetic and mutation profiles of patients with myeloprolifera­
tive neoplasms (MPNs)
• To be aware of the central role of molecular tests in the modern management of MPNs
• To learn how to use at best molecular information for risk­stratifying patients with MPNs
• To acknowledge the major clinical needs remaining unmet

Deep characterization of genomic abnormalities is essen­ mosomes/DNA sequence that deserve diagnostic and
tial for modern management of chronic myeloprolifera­ prognostic significance and can be identified through the
tive neoplasms (MPNs), including polycythemia vera (PV), application of methods available in specialized clinical lab­
essential thrombocythemia (ET), and primary myelofibrosis oratories. Therefore, this article is by no way an exhaustive
(PMF), as well as post­PV and post­ET myelofibrosis (MF) review of genomics in MPNs, nor does it discuss mechanis­
(collectively, secondary myelofibrosis [sMF]). PMF includes tic implications of genomic abnormalities.
an early/prefibrotic stage (pre­PMF) as well as an overt
fibrotic stage, as 2 distinct diagnostic entities.1 Genom­ (A few) Technical tips to know
ics informs diagnosis and risk assessment and supports Conventional methods to assess chromosomal abnormal­
therapy decision­making. Herein, we refer to genomic ities in MPNs are chromosomal banding (numerical and
abnormalities in MPNs to include only changes in chro­ structural changes) and fluorescence in situ hybridization

Molecular prognostication in MPN | 225


(that has the poten­tial to dis­cover also cyto­ge­net­i­cally cryp­tic Chromosomal abnor­mal­i­ties in MPNs
abnor­mal­i­ties), pref­er­a­bly using bone mar­row (BM) or, if unavail­ Chromosomal alter­ations occur at vary­ing fre­quen­cies and have
able, periph­ eral blood (PB) cells. Driver muta­ tions are rou­ var­i­able prog­nos­tic sig­nif­i­cance depending on the under­ly­ing
tinely assessed in PB sam­ples (pref­er­a­bly on gra­di­ent-puri­fied dis­ease (Table 1). Cytogenetic abnor­ mal­i­
ties in PV and ET at
granulocytes, which rep­ re­
sent the variably involved mye­ loid diag­no­sis are rare (5%-15%) but increase with dis­ease pro­gres­
clone and allow more repro­duc­ible mea­sure­ment of muta­tion sion to sMF (up to 80%).3 Most com­mon are dele­tion of the long
var­i­ant allele fre­quency [VAF] by elim­i­nat­ing con­tam­i­na­tion of arm of chro­mo­some 20, tri­somy 8, or tri­somy 9. In PV, an inter­
lym­phoid cells) by using dif­fer­ent poly­mer­ase chain reac­tion– me­di­ate- and high-risk kar­yo­type cat­e­gory was iden­ti­fied (Table
based tech­niques that, par­tic­u­larly for JAK2 V617F muta­tion, 1) influ­enc­ing over­all sur­vival (OS) and leu­ke­mia-free sur­vival.3,4
allow pre­cise cal­cu­la­tion of the ratio of mutated vs wild-type In ET, cyto­ge­netic abnor­mal­i­ties are infre­quent (7%) and asso­ci­
allele VAF; recommended assay sen­si­tiv­ity is ≤1%, in order to ated with shorter sur­vival (10-12 years vs 21 years with­out abnor­
iden­tify early dis­ease pre­sen­ta­tion and, on the other side, to mal­i­ties).5 However, abnor­mal kar­yo­type is not included in any
accu­rately mon­i­tor response to treat­ment (stem cell trans­plan­ inte­grated prog­nos­tic model for ET, while in asso­ci­a­tion with

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


ta­tion [SCT], inter­feron). Quantification of MPL and CALR VAF is advanced age, leu­ko­cy­to­sis, and venous throm­bo­sis, as well as
not rou­tinely performed owing to tech­ni­cal con­straints and lack advanced age and leu­ko­cy­to­sis, abnor­mal kar­yo­type is prognos­
of stan­dard­ized assays; how­ever, dis­tinc­tion of type 1 and type 2 tically infor­ma­tive for OS and leu­ke­mic trans­for­ma­tion, respec­
CALR muta­tion should be rou­tinely reported since it is prognos­ tively, in PV.6
tically infor­ma­tive. Next-gen­er­a­tion sequenc­ing (NGS)–based Conversely, spe­cific chro­mo­somal abnor­mal­i­ties are an inte­
meth­ods are cur­rently incor­po­rated in the workup of selected gral com­po­nent of prog­nos­tic scores for PMF, such as the DIPSS-
patients with MPNs. Usually, they are panel based, inter­ro­gat­ing plus, genetically inspired prognostic scoring system (GIPSS),
20 to 100+ most fre­quently mutated genes, with a con­ven­tional and Mutation-Enhanced International Prognostic Score System
detec­tion limit of 2% to 5%. Although NGS data can high­light for Transplantation-Age Patients with Primary Myelofibrosis
struc­tural changes and copy num­ber var­i­a­tions, for clin­i­cal pur­ (MIPSS70/v2) (discussed below). Patients with pre-PMF and overt
poses, only sin­gle-nucle­o­tide var­i­ants and inser­tions/dele­tions PMF have a sim­i­lar inci­dence and type of cyto­ge­netic abnor­
(indels) are reported. Ideally, attri­bu­tion of a sin­gle-nucle­o­tide mal­i­ties.7 An iso­lated 13q−, 20q−, and 9+ abnor­mal­ity deserves
var­i­ant to a somatic change requires paired anal­y­sis of a germ­line favor­able prog­nos­tic value, while unfa­vor­able abnor­mal­i­ties con­
DNA source, which in clin­i­cal prac­tice is unfea­si­ble and expen­ sist of a com­plex kar­yo­type (>3 abnor­mal­i­ties) or a sin­gle or 2
sive and is reserved for selected cases; there­fore, somatic def­i­ abnor­mal­i­ties, includ­ing +8, −7/7q−, i(17)q, −5/5q−, 12p−, inv(3),
ni­tion of a var­i­ant is based on com­par­a­tive inter­ro­ga­tion of large or 11q23 rearrangements or a monosomal kar­yo­type (MK). More
pop­u­la­tion data­bases to fil­ter out known pop­u­la­tion poly­mor­ recently, the prog­nos­tic value of cyto­ge­netic abnor­mal­i­ties was
phisms. A sec­ond level of com­plex­ity in interpreting NGS data refined in a series of 1002 patients, resulting in 3 risk categories8:
con­cerns the attri­bu­tion of path­o­ge­nic­ity to the reported var­i­ very high risk (VHR), favor­able, and unfa­vor­able kar­yo­type (see
ant (ie, var­i­ant anno­ta­tion), which requires bioinformatic exper­ Table 1 for details), with a median sur­vival of 1.2 years (haz­ard
tise and should be performed in accor­ dance with published ratio [HR], 3.8; 95% con­fid ­ ence inter­val [CI], 2.9-4.9), 2.9 years
guide­lines.2 (HR, 1.7; 95% CI, 1.4-2.0), and 4.4 years (ref­er­ence), respec­tively.

Table 1. Prognostically infor­ma­tive cyto­ge­netic abnor­mal­i­ties most com­monly encoun­tered in patients with MPNs

Essential Blast phase


Polycythemia vera Primary mye­lo­fi­bro­sis Post-PV/post–ET-MF
thrombocythemia post-MPNs
Prevalence, % 1-7 10-20 35-50 40-80 80-90
Chromosome • “Intermediate risk”: • “Unfavorable karyo”: com­plex • “Unfavorable” karyo: • Complex
abnor­mal­i­ties - Sole del(20q) or sin­gle or 2 abnor­mal­i­ties, abnor­mal­i­ties other karyo
with prog­nos­tic - Double abnor­mal­i­ties, includ­ing +8, −7/7q−, i(17)q, than iso­lated 13q− includ­ing
sig­nif­i­cance includ­ing +1q (median OS, −5/5q−, 12p−, inv(3), or 11q23 and 20q−, mono­somy more
86 months) rear (included in DIPSS-plus) fre­quently
• “High risk”: • “Favorable” karyo: nor­mal kar­ −5/del(5q),
- Complex kar­yo­type yo­type, sole abnor­mal­i­ties −7/del(7q),
(median OS, 9 months) of 20q−, 13q−, +9, chr 1 trans­ −17/del(17p)/
lo­ca­tion/dupli­ca­tion, or sex i(17q), and −18
chro­mo­some abnor­mal­ity
includ­ing −Y
• “VHR” karyo: sin­gle or mul­ti­ple
abnor­mal­i­ties of −7, inv (3),
I (17q), 12p−, 11q−, and
auto­so­mal tri­so­mies other
than +8 or +9
• “High-risk” karyo: all­ abnor­mal­i­
ties that are not VHR and favor­
able (included in MIPSS70/plus
v2.0 and GIPSS scores)

226 | Hematology 2022 | ASH Education Program


The unfa­vor­able and VHR kar­yo­type con­ferred a 2.0- and 4.4-fold V617F–neg­at­ive PV; muta­tions of the thrombopoietin recep­tor
increased risk to prog­ress to blast phase (BP), respec­tively. (MPL W515L/K/A) in 5% to 8% of patients with ET and PMF, includ­
An abnor­mal kar­yo­type was found in 34% of 376 patients with ing rare S505N; and exon 9 calreticulin muta­tions (CALR) in 20% to
sMF and had neg­at­ ive impact on median sur­vival: 10.1 years (95% 25% of ET and PMF. There are 2 pro­to­type CALR muta­tions: type
CI, 8.1-not reached) com­pared with 6.1 years (95% CI, 4.8-not 1 (52-bp dele­tion) and type 2 (5-bp inser­tion), with sev­eral other
reached) for nor­mal kar­yo­type.3 Shortened sur­vival was fur­ther type 1–like and type 2–like var­i­ants. Driver muta­tions rep­re­sent
affected by the pres­ence of a com­plex kar­yo­type (2.7 years), major diag­nos­tic cri­te­ria in the lat­est World Health Organization
com­plex kar­yo­type with­out MK (3.4 years), and MK (2.1 years). (WHO) clas­si­fi­ca­tion,1 as well as in the fifth edi­tion of the WHO
However, when added to the Myelofibrosis Secondary to PV and clas­si­fi­ca­tion12 and the International Consensus Classification of
ET-Prognostic Model (MYSEC-PM) strata (see below), abnor­mal Myeloid Neoplasms and Acute Leukemias.13 However, a driver
kar­yo­type did not add to sur­vival pre­dic­tion. muta­tion is miss­ing in occa­sional patients with PV and 10% to
Cytogenetic abnor­mal­i­ties con­sti­tute an impor­tant risk fac­tor 15% of those with ET and PMF (“tri­ple neg­a­tive”); this even­tu­al­ity
in the pro­gres­sion of MPNs to the accel­er­ated phase (AP)/BP and is acknowl­edged in cur­rent diag­nos­tic sys­tems that rec­om­mend

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


are reported in up to 90% of patients.9 The chro­mo­somal defects NGS to search uncom­mon (“noncanonical”) somatic var­i­ants in
most sig­nif­i­cantly asso­ci­ated with AP/BP are the com­plex kar­ JAK2 and MPL14 or to iden­tify a clonal marker. NGS may also dis­
yo­type (includ­ing −5/del(5q), −7/del(7q), −17/del(17p)/i(17q), and cover rare JAK2/MPL germline var­i­ants that under­lie hered­i­tary
−18), gains of chro­mo­some 1q (MDM4 gene locus, encoding a erythrocytosis or thrombocytosis mim­ick­ing MPNs.15,16
neg­a­tive reg­u­la­tor of TP53), or MK.10 Most patients with AP/BP Similar to those with acute leu­ke­mia and myelodysplastic
show loss of 17p13, which leads to a dele­tion of TP53.11 syn­dromes, patients with MPN may har­bor muta­tions in a vari­
ety of so-called mye­loid genes that include mainly epi­ge­netic
reg­u­la­tors, spliceosome com­po­nents, and onco­genes (Table 2
Mutations asso­ci­ated with MPNs lists the most fre­ quent).17 These muta­ tions may pre­ date or
MPN-asso­ci­ated driver muta­tions include JAK2 V617F muta­tion in fol­low the acqui­si­tion of the driver muta­tion and affect dis­ease
>95% of patients with PV and 60% with ET and PMF; JAK2 exon 12 phe­no­type.18 Not all­muta­tions have uni­form prog­nos­tic sig­nif­
indels (the most fre­quent is N542_E543del) in 1% to 3% of JAK2 i­cance. A set of so-called high muta­tion risk (HMR) genes,19,20

Table 2. Prognostically infor­ma­tive muta­tions of most fre­quent detec­tion in patients with MPN

Gene Polycythemia vera Essential thrombocythemia Primary mye­lo­fi­bro­sis “Secondary” mye­lo­fi­bro­sis


JAK2 • Higher risk of throm­bo­sis • Intermediate prog­no­sis and
V617F (included in revised IPSET) higher risk of throm­bo­sis
com­pared to patients with
CALR muta­tion
CALR • Lower risk of throm­bo­sis com­ • CALR type 1/like: improved • Improved OS com­pared
pared to JAK2 mutated sur­vival com­pared to JAK2 to JAK2 V617F and “tri­ple
V617F and “tri­ple neg­a­tive” neg­a­tive”
• Improved out­come after SCT • Improved out­come after SCT
• Absence of CALR type 1/like • Absence of CALR muta­tion
included in MIPSS70/plus v2.0 included in MYSEC-PM and
• Absence of CALR muta­tion MTSS scores
included in MYSEC-PM and
MTSS scores
MPL W515 • Intermediate prog­no­sis and
higher risk of throm­bo­sis
com­pared to patients with
CALR muta­tion
“Triple • Inferior OS and LFS com­pared
neg­a­tive” to JAK2/CALR muta­tion (espe­
cially in pre-PMF)
JAK2 • Similar rates of throm­bo­sis,
exon 12 evo­lu­tion to post–PV-MF
and BP, and sur­vival to
JAK2 V617F
ASXL1 • “Adverse var­i­ant” asso­ci­ • “HMR var­i­ant” asso­ci­ated with
ated with infe­rior OS and infe­rior OS/LFS, lower PFS fol­
MFS low­ing SCT
• Included in MIPSS-PV • Included in MIPSS70/plus v2.0
and GIPSS
EZH2 • “Adverse var­i­ant” asso­ci­ated • “HMR var­i­ant” asso­ci­ated with
with infe­rior OS/LFS infe­rior OS
• Included in MIPSS70/plus v2.0

Molecular prog­nos­ti­ca­tion in MPN | 227


Table 2. Prognostically infor­ma­tive muta­tions of most fre­quent detec­tion in patients with MPN (Continued )

Gene Polycythemia vera Essential thrombocythemia Primary mye­lo­fi­bro­sis “Secondary” mye­lo­fi­bro­sis


IDH1 • IDH1: “adverse var­i­ant” • IDH2: “adverse var­i­ant” asso­ci­ • “HMR var­i­ants” asso­ci­ated with • “Adverse var­i­ant” asso­ci­ated
IDH2 asso­ci­ated with infe­rior OS ated with infe­rior OS infe­rior OS/LFS and as lower with lower PFS fol­low­ing SCT
and LFS PFS fol­low­ing SCT
• Included in MIPSS70/plus v2.0
SRSF2 • “Adverse var­i­ant” asso­ci­ • “Adverse var­i­ant” asso­ci­ated • “HMR var­i­ant” asso­ci­ated with
ated with infe­rior OS with infe­rior OS/LFS infe­rior OS/LFS
• Included in MIPSS-PV • Included in MIPSS-ET • Included in MIPSS70/plus v2.0
and GIPSS
U2AF1 • “HMR var­i­ant” asso­ci­ated infe­
Q157 rior OS
• Included in MIPSS70/plus v2.0

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


and GIPSS
U2AF1 or • U2AF1: “adverse var­i­ant” asso­ • “Adverse var­i­ant” asso­ci­ated • “Adverse var­i­ant” asso­ci­ated
DNMT3A ci­ated with infe­rior OS/MFS with worse OS in SCT with worse OS in SCT
or CBL • Included in MIPSS-ET
RUNX1 • “Adverse var­i­ant” asso­ci­ • “Adverse var­i­ant” asso­ci­ated
ated with infe­rior OS and with infe­rior OS/LFS
LFS
TP53 • “Adverse var­i­ant” asso­ci­ • “Adverse var­i­ant” asso­ci­ated • “Adverse var­i­ant” asso­ci­ated
ated with infe­rior LFS with infe­rior OS/LFS with leu­ke­mic trans­for­ma­tion
• Included in MIPSS-ET
LNK • “Adverse var­i­ant” asso­ci­ated
(SH2B3) with infe­rior OS
SF3B1 • “Adverse var­i­ant” asso­ci­ated
with infe­rior OS and MFS
• Included in MIPSS-ET
RAS • “Adverse variant” associated
with inferior OS
• Associated with JAKi-refrac­to­
ri­ness
LFS, leu­ke­mia-free sur­vival; MFS, myelofibrosis-free survival; PFS, progression-free survival.

includ­ing ASXL1, EZH2, SRSF2, IDH1, IDH2, and U2AF1Q517, pre­ nuclei atypia, in tight clus­ters; and slightly reduced eryth­ro­poi­
dict infe­rior sur­vival in PMF, inde­pen­dent of each other and e­sis. Reticulin fibro­sis was grade 0/1. Cytogenetics showed a
other risk fac­tors, with >1 mutated gene har­bor­ing addi­tional nor­mal male kar­yo­type. A JAK2 V617F muta­tion with a VAF of
neg­a­tive weight.21 34% was detected in PB granulocytes. On the basis of those
find­ings, the patient received a WHO 2016 diag­no­sis of prefi­
brotic MF. The patient had a score of 2 according to the MPN-10
total symp­tom score (MPN-10).22
CLINICAL CASE
A 42-year-old man presented to an out­pa­tient clinic fol­low­ing
the ser­en­dip­i­tous dis­cov­ery of thrombocytosis (800 × 109/L) Molecular prog­nos­ti­ca­tion sys­tems to address the risk
in rou­ tine blood cell tests. Hemoglobin was in the lower of vas­cu­lar events
range (13.2  g/dL with nor­mal indexes), and leu­ko­cytes were Cardiovascular events are the main rea­son for mor­bid­ity and
10.3 × 109/L, with no imma­ ture cells in the blood smear. All mor­tal­ity in PV and ET; in pre-PMF, the rate of throm­bo­sis is sim­i­
other rou­tine tests were nor­mal. He was asymp­tom­atic, had no lar to ET, esti­mated at 1.99% patients/year. Patients with PV and
referred famil­ial his­tory for hema­to­logic malig­nan­cies, and had ET are con­ven­tion­ally risk-strat­i­fied for throm­bo­sis based on
no known generic car­dio­vas­cu­lar risk fac­tor; phys­i­cal exam­i­na­ 2 clin­i­cal cri­te­ria: age ≥60 years and throm­bo­sis his­tory.23 How­
tion was unre­mark­able, and spleen was not pal­pa­ble. An ultra­ ever, the dis­cov­ery that patients with ET who have a CALR muta­
sound scan con­firmed nor­mal spleen vol­ume and also ruled out tion have a sig­nif­i­cantly reduced rate of vas­cu­lar events led to the
splanch­nic vein throm­bo­sis. The patient under­went BM biopsy, devel­op­ment of an inte­grated score, the International Prognosis
which revealed hypercellularity with gran­u­lo­cytic pro­lif­er­a­tion Score of Thrombosis for Essential Thrombocythemia (IPSET),
with­out atypia or blast increase; marked expan­sion of the mega­ includ­ing the cur­rently recommended revised ver­sion (Table 3).24
kar­yo­cytic lin­e­age with mega­kar­yo­cytes of var­i­able size and The pos­i­tiv­ity of a JAK2 V617F muta­tion, in the absence of the

228 | Hematology 2022 | ASH Education Program


Table 3. Molecularly inte­grated risk scores in patients with ET and PV

Essential thrombocythemia Polycythemia vera


Characteristic
Revised IPSET MIPSS-ETa MIPSS-PVa
Age, y >60 >60 (4) >67 (2)
History of throm­bo­sis Yes — Yes (1)
JAK2 V617F Yes — —
White blood cell count — ≥11×109/L [1] ≥15×109/L (1)
Adverse muta­tion — SRSF2, SF3B1, U2AF1, TP53 [2] SRSF2 [3]
Male sex — Yes [1] —
Thrombosis risk group (rate % patients/y) Risk group [score]: median sur­vival (y)

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


Categories Very low: no throm­bo­sis his­tory, age ≤60y, JAK2WT Low [0-1]: 34.4 Low [0-1]: 24.0
Low: no throm­bo­sis his­tory, age ≤60y, JAK2 V617F Intermediate [2-5]: 14.1 Intermediate [2-3]: 13.1
Intermediate: no throm­bo­sis his­tory, age >60y, JAK2WT High [≥6]: 7.9 High [≥4]: 3.2
High: throm­bo­sis his­tory or age >60y with JAK2 V617F
Values within brack­ets indi­cate the var­i­able’s rank.
a

2 afore­men­tioned clin­i­cal cri­te­ria, qualifies a patient as low risk, national Prognostic Scoring System (IPSS) and Dynamic-IPSS
with aspi­rin as suggested treat­ment. IPSET predicted throm­bo­ (DIPSS), or the cyto­ge­net­ics-inte­grated DIPSS-plus con­tinue to
sis risk also in prefibrotic MF.25 The role of addi­tional muta­tions be largely used in prac­tice and, nota­bly, still are used for selec­
for throm­bo­sis in ET is largely unset­tled.26 In PV, no com­pel­ling tion of patients in clin­i­cal tri­als. Furthermore, the IPSS and DIPSS
evi­dence for driver/addi­tional muta­tions as being infor­ma­tive scores are rou­ tinely applied also to patients with pre-PMF,
for throm­ bo­sis risk has been reported yet; the dem­ on­
stra­ although it was dem­on­strated that they poorly dis­crim­i­nate
tion that a JAK2 V617F VAF ≥50% has an inde­pen­dent HR of 3.8 the inter­me­di­ate 1 and 2 risk categories.7,31 Such short­com­ings
(95% CI, 1.7-8.6) for venous throm­bo­sis27 might fos­ter devel­op­ might be addressed by more recently devel­oped, more infor­
ment of inte­grated scores. ma­tive, molec­u­lar-inte­grated scores.32
The abovementioned HMR genes con­fig­ure an adverse var­
i­able in inte­grated risk scores for PMF such as the Mutation-
Molecular prog­nos­ti­ca­tion sys­tems to address the risk Enhanced International Prognostic Score System for Trans­
of dying plantation-Age Patients With Primary Myelofibrosis (MIPSS70)31
According to a series of 3023 patients with MPNs, median OS is and MIPSS70v2.0, enriched with sex-adjusted hemo­ glo­bin
around 20, 15, and 5 years, respec­tively, for ET, PV, and PMF.28 In and revised kar­yo­type clas­si­fi­ca­tion.33 MIPSS70, orig­i­nally
ET, the con­ven­tional IPSET score uses age >60 years, leu­ko­cytes devel­ oped for patients of trans­ plant age but infor­ ma­tive
>11 × 109/L, and throm­bo­sis his­tory to dif­fer­en­ti­ate low-, inter­ age-inde­ pen­dently as well, includes 9 var­ i­
ables, 3 genetic
me­di­ate-, and high-risk patients with respec­tive median sur­ (HMR muta­tion, >1 HMR muta­tion, and absence of CALR type
vival not reached, 24.5 years, and 13.8 years.29 For PV, the 1/like muta­tion), 5 hema­to­logic-clin­i­cal fac­tors, and bone
International Working Group sur­vival model delin­eated 3 risk mar­row fibro­sis grade ≥2 (Table 4). Based on unique var­ia ­ ble-
groups with median sur­ viv­als of 10.9, 18.9, and 27.8 years, spe­cific HR weighted scores, 3-tiered MIPSS70 low-, inter­me­
based on older age, leu­ko­cy­to­sis, and venous throm­bo­sis.6 di­ate-, and high-risk categories, with cor­re­spond­ing median
More recently, a muta­tion-enhanced inter­na­tional prog­nos­tic sur­vival ranges of 27.7 years to “not reached,” 6.3 to 7.1 years,
sys­tem (MIPSS) for ET and PV was devised.4 Spliceosome muta­ and 2.3 to 3.1 years, were iden­ti­fied and val­i­dated in 2 inde­
tions adversely affected OS (SF3B1, SRSF2 in ET and SRSF2 pen­dent cohorts. Conversely, MIPSSv2 includes 5 risk cate­
in PV) and mye­lo­fi­bro­sis-free sur­vival (U2AF1, SF3B1 in ET); gories: VHR (median sur­vival, 1.8 years), high risk (4.1 years),
TP53 muta­tions predicted BP in ET. These adverse muta­tions inter­me­di­ate risk (7.7 years), low risk (16.4 years), and very low
occurred in 10% and 2% of patients with ET and PV, respec­ risk (median not reached) (Table 4). Improved per­for­mance of
tively. The inte­grated clin­i­cal-molec­u­lar model iden­ti­fied 3 risk MIPSS/v2 com­pared with con­ven­tional IPSS/DIPSS was dem­
categories with a median sur­vival of 8.3 to 34.3 years in ET and on­strated, with up to 40% of the patients being upgraded.
4.6 years to not reached in PV4 (Table 3). However, now­a­days, Of note, the MIPSS score was devel­oped for all­patients with
these scores are not used in clin­ i­
cal prac­
tice for ther­ apy PMF and includes fibro­sis grade (0-1 vs 2-3 cat­e­gory) as an
deci­sion-mak­ing. embed­ ded var­ i­able addressing the pre-PMF and overt-PMF
Conversely, prog­nos­tic scores deserve a cen­tral role in the cat­e­gory. A sep­a­rate model based only on molec­u­lar fac­tors,
man­age­ment of patients with MF spe­cif­i­cally concerning the GIPSS, incor­ po­ rated the 3-tiered kar­ yo­
type categories and
indi­ca­tion to SCT, the only poten­tially cura­tive option; how­ 4 muta­tions (ASXL1, SRSF2, and U2AF1Q157, plus absence of type
ever, the not neg­ li­
gi­
ble risk asso­ ci­ated with the trans­ plant 1/like CALR muta­tion) as inde­pen­dent risk fac­tors for sur­vival;
man­dates care­ful selec­tion of patients to make the pro­ce­dure risk categories were low (median sur­vival, 26.4 years), inter­me­
risk-effec­tive.30 Conventional clin­i­cal-only scores, such as Inter­ di­ate 1 (8.0 years), inter­me­di­ate 2 (4.2 years), and high (2 years

Molecular prog­nos­ti­ca­tion in MPN | 229


Table 4. Molecularly inte­grated risk scores in patients with PMF and sMF

MIPSS70/plus
DIPSS-plus MIPSS70 GIPSS MYSEC-PM MTSS
v2.0
Prognostic var­i­ables (points)
Age, y >65 (1) — — — 0.15 point/y ≥57 (1)
Constitutional Present (1) Present (1) Present (2) — Present (1) —
symp­toms
Hemoglobin, g/dL <10.0 (2) <10 (1) 8-9.9 F; 9-10.9M — <11 (2) —
(1)
<8 F, <9M (2)
Leukocyte >25.0 (1) >25.0 (2) — — >25.0 (1)

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


count,×109/L
Circulating blast ≥1% (1) ≥2% (1) ≥2% (1) — ≥3% (2) —
cells, %
Platelet <100 (1) <100 (2) — — <150 (1) <150 (1)
count,×109/L
RBC trans­fu­sion Yes (1) — — — — —
need
Karyotype Unfavorable (1) — High risk (3) High risk (1) — —
Very high risk (4) Very high risk (2)
BM fibro­sis grade — ≥MF2 (1) — — — —
Absence of CALR — Yes (1) Yes (2) Yes (1) Yes (2) Yes/absence of
MPL (2)
Molecular pro­file — HMR 1 mut (1) HMR+U2AF1 Q157 ASXL1 (1) — ASXL1 (1)
HMR ≥2 mut (2) 1 mut (2) SRSF2 (1)
HMR+U2AF1 U2AF1 Q157 (1)
Q157≥2 mut (3)
Karnofsky PS — — — — — < 90% (1)
HLA donor — — — — — MUD (2)
Risk group (score): Overall sur­vival in years
Categories Low (0):15y Low (0-1): 27.7y Very low (0): not Low (0): 26.4y Low (<11): not Low (0-2): 90%*
Intermediate 1 (1): Intermediate (2-4): reached Intermediate 1 (1): reached Intermediate (3-
6.7y 7.1y Low (1-2): 10.3y 8.0y Intermediate 1 4): 77%*
Intermediate 2 High (≥5): 2.3y Intermediate (3- Intermediate 2 (11-14): 9.3y High (5): 50%*
(2-3): 2.9y 4): 7.0y (2): 4.2y Intermediate 2 Very high (6-9):
High (≥4): 1.3y High (5-8): 3.5y High (3-6): 2.0y (14-16): 4.4y 34%*
Very high (≥9): High (≥16): 2.0y
1.8y
When/for whom During fol­low-up; At diag­no­sis; PMF At diag­no­sis; PMF At diag­no­sis; PMF At diag­no­sis; sMF MF patients
PMF patients patients patients patients planned for SCT
Website tool https:​­/​­/qxmd​ www​­.mipss70score​­.it www​ NA www​­.mysec-pm​ NA
.­ com​­/calculate​ ­.mipss70score​­.it ­.eu
­/calculator_315​­/
dipss​­-plus​­-score​
­-for​­-prognosis​­-in​
­-myelofibrosis
Molecular pro­file descrip­tion Karyotype descrip­tion
DIPSS-plus Unfavorable: com­plex kar­yo­type or sin­gle or 2 abnor­mal­i­ties, includ­ing +8,
−7/7q−, i(17q), −5/5q−, 12p−, inv(3), or 11q23 rearrangement
MIPSS70 Absence of CALR type 1 Not included
HRM: muta­tion in at least 1 of these
mutated genes: ASXL1, SRSF2, IDH1, IDH2,
EZH2
MIPSS70/plus v2.0 Absence of CALR type 1 High risk (HR): all­the abnor­mal­i­ties that are not VHR and favor­able (nor­mal kar­
HMR: muta­tion in at least 1 of these yo­type or sole abnor­mal­i­ties of 20q−, 13q−, +9, chro­mo­some 1 trans­lo­ca­tion/
mutated genes: ASXL1, SRSF2, IDH1, IDH2, dupli­ca­tion, or sex chro­mo­some abnor­mal­ity includ­ing −Y)
EZH2, U2AF1 Q157 Very high risk (VHR): includes sin­gle or mul­ti­ple abnor­mal­i­ties of −7, inv (3),
I (17q), 12p−, 11q−, and auto­so­mal tri­so­mies other than +8 or +9

230 | Hematology 2022 | ASH Education Program


Table 4. Molecularly inte­grated risk scores in patients with PMF and sMF (Continued )

MIPSS70/plus
DIPSS-plus MIPSS70 GIPSS MYSEC-PM MTSS
v2.0
Prognostic var­i­ables (points)
GIPSS Mutation in ASXL1, SRSF2, U2AF1 Q157 Same as MIPPS70/plus v2.0
MYSEC-PM Absence of CALR (all­types) Not included
MTSS Absence of CALR (all­types)/MPL Not included
Mutation in ASXL1
According to National Comprehensive Cancer Network guide­lines, in patients with MF, a lower-risk cat­e­gory includes the fol­low­ing com­bi­na­tion:
MIPSS70≤3, MIPSS70v2≤3, DIPSS-plus ≤1, DIPSS ≤2, and MYSEC-PM <14, while a higher-risk cat­e­gory includes MIPSS70≥4, MIPSS70v2≥4, DIPSS-plus >1,
DIPSS >2, and MYSEC-PM ≥14.

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


*5-year overall survival (learning-cohort).
HLA, human leukocyte antigen; NA, not available; PS, performance status; RBC, red blood cell.

[≥3 points]).34 For patients with sMF, the MYSEC-PM includes evo­lu­tion of pre-PMF to overt PM was made. The dis­ease was
5 clin­i­cal fac­tors plus an unmutated CALR geno­type, while the strat­i­fied as fol­lows:
role of “mye­loid” muta­tions is still debated.35 The sim­pli­fied DIPPS-plus: inter­me­di­ate 1 risk, due to con­sti­tu­tional symp­
National Comprehensive Cancer Network guide­ lines strat­ i­ toms and >1% PB blasts; the esti­mated sur­vival is about 7 years.
fi­ca­tion cri­te­ria include a lower and a higher risk cat­eg ­ ory, MIPSSv2: very high risk, due to BM fibro­sis ≥G2, con­sti­tu­
based on var­i­able scores of MIPSS and MIPSSv2, if geno­mics is tional symp­toms, absence of CALR type 1 muta­tion, and HMR
avail­­able, or DIPSS-plus and MYSEC-PM if molec­u­lar test­ing is cat­e­gory with ≥2 HMR mutated genes; the esti­mated sur­vival is
not avail­­able.36 <2 years with <5% like­li­hood of sur­vival at 10 years.
Mutations are included also in the Myelofibrosis Transplant The patient had become highly symp­tom­atic and had an
Scoring System (MTSS), an inte­grated score devel­oped for pre­ MPN-10 score of 44. An indi­ca­tion to hema­to­poi­etic SCT was
dicting prog­no­sis after SCT for PMF and sMF that includes age, posed after thor­ ough dis­
cus­sion with the patient, based
Karnofsky per­for­mance sta­tus, throm­bo­cy­to­pe­nia, leu­ko­cy­to­sis, on MIPSSv2. He had a fully HLA-matched sib­ling donor. The
human leukocyte antigen (HLA)-mismatched unre­lated donor, MTSS score yielded an inter­me­di­ate risk cat­e­gory, due to the
ASXL1 muta­tion, and non-CALR/MPL driver muta­tion geno­type. absence of a CALR/MPL muta­tion and the pres­ence of an ASXL1
The 5-year sur­vival was 83% (95% CI, 71%-95%), 64% (53%-75%), muta­tion; the esti­mated 5-year sur­vival was 77%. After SCT, the
37% (17%-57%), and 22% (4%-39%), respec­ tively, in the low-, patient cleared the JAK2 V617F muta­tion at 4 months and HMR
inter­me­di­ate-, high-, and very high-risk categories37 (Table 4). muta­tions (assayed at 1 year). He is alive, with no evi­dence of
dis­ease, after 3 years.

CLINICAL CASE (Con­t in­u ed) How we use molec­u­lar tests in patients with MPNs
The patient was cat­e­go­rized at diag­no­sis as IPSS low risk, with Our approach to the use of molec­u­lar tests for prog­nos­ti­ca­
projected OS >10 years. No cytoreductive ther­apy was insti­ tion pur­poses in MPNs is depicted in Figure 1. Driver muta­tions
tuted, except low-dose aspi­ rin since, based on the revised are step­wise inter­ro­gated in all­patients since, beyond being
IPSET score, he belonged to the low-risk cat­e­gory. For the next required for diag­no­sis, they are prognostically infor­ma­tive in ET
5 years, the dis­ease course was unevent­ful. Then, his hemo­ and pre-PMF for throm­bo­sis (revised IPSET) and in MF for sur­
glo­bin level began to down­trend (11.4 g/dL), mild leu­ko­cy­to­ vival (favor­able impact of CALR type 1/like). We use quan­ti­ta­tive
sis (18 × 109/L) appeared steadily, plate­lets were 550 × 1012/L, 1% assays for mea­sur­ing JAK2 V617F VAF at diag­no­sis and even­tu­
blasts and leukoerythroblastosis were detected in the blood ally doc­u­ment its increase at the time of evo­lu­tion to post-PV
smear, and lactate dehydrogenase was increased 1.8-fold and post-ET MF. Furthermore, serial mea­sure­ment of JAK2 V617F
above nor­mal. Spleen was pal­pa­ble at 4 cm from the left cos­ might be infor­ma­tive in patients with PV receiv­ing ropeginter­
tal mar­gin, and there was a recent onset of night sweats. The feron to doc­um ­ ent changes in the clone of mutated cells.38,39
patient consulted another insti­tu­tion, where a new diag­nos­tic Owing to the lim­ited appli­ca­bil­ity of sur­vival prog­nos­tic mod­
pro­ce­dure was performed. The bone mar­row biopsy spec­i­men els for deci­sion-mak­ing, together with finan­cial con­sid­er­ations,
revealed slightly reduced age-adjusted cel­lu­lar­ity, with marked we do not cur­rently sup­port rou­tine appli­ca­tion of NGS to inter­
increase of atyp­ i­
cal mega­ kar­
yo­cytes in paratrabecular clus­ ro­gate mye­loid muta­tions in patients with PV and ET, while we
ters; CD34+ cells were around 3%. Fibrosis was G2, with sparse per­form NGS rou­tinely in all­trans­plant-age patients with PMF
areas of G3 and ini­tial focal collagenization. PB blasts were 2%. and sMF at diag­no­sis. Although most expe­ri­ence was obtained
Cytogenetics showed iso­lated tri­somy 9. Targeted NGS panel until now in overt PMF, we cur­rently per­form NGS also in youn­ger
iden­ti­fied muta­tions in ASXL1 (VAF, 24%) and SRSF2 (VAF, 33%). patients with pre-PMF at diag­no­sis to cal­cu­late the MIPSS score,
The VAF of JAK2 V617F had increased to 75%. A diag­no­sis of as the presented clin­i­cal case would have advo­cated. Results

Molecular prog­nos­ti­ca­tion in MPN | 231


Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022
Figure 1. Flowchart on how we use genomic assays for diagnosis (A) and for prognostication in patients (B) with an established
diagnosis of MPN. Selected information (cytogenetics, JAK2 V617F and CALR mutations) collected at the time of diagnosis may also
inform risk scores at step B. NGS analysis for myeloid genes, if not available at the time of original investigation, is recommended in
patients with PMF and sMF who are transplant eligible. PET, post-ET; PPV, post-PV.

could be even­ tu­ally used also for assessing the trans­ plant- yo­
type and/or TP53 involve­ ment, although we acknowl­ edge
related risk (MTSS). There is still no indi­ca­tion if, and at what that results of the kar­yo­type inform clin­i­cal deci­sions only in
inter­vals, NGS should be repeated dur­ing the course of dis­ease; patients with PMF.
in prac­tice, now­ad ­ ays NGS is usu­ally performed coin­ci­dent with
some clin­i­cal and hema­to­logic evi­dence of dis­ease pro­gres­sion. Conclusions
In SCT-inel­i­gi­ble patients, inte­grated scores do not pro­vide clin­ The past 10 years have witnessed incred­i­ble advance­ments in
i­cally action­able infor­ma­tion, and we con­sider them not man­da­ molec­u­lar-based prog­nos­ti­ca­tion of MPNs. Yet, we are left with
tory if cost con­sid­er­ations limit their use. Cytogenetics should a num­ber of clin­i­cally rel­e­vant unmet needs, includ­ing the pre­
be ide­ally obtained in all­patients with MPNs at diag­no­sis and dic­tion of acute leu­ke­mia in a clin­i­cally action­able time frame,
dur­ing the clin­i­cal course, if indi­cated, to exclude com­plex kar­ the pre­dic­tion of response to JAKi (which might be adversely

232 | Hematology 2022 | ASH Education Program


asso­ci­ated with RAS/CBL muta­tions),40 and the short- and long- i­cal, clin­i­cal, and geno­mic data [published online ahead of print 29 June
term prog­nos­tic sig­nif­i­cance of molec­u­lar responses under 2022]. Blood.
14. Milosevic Feenstra JD, Nivarthi H, Gisslinger H, et al. Whole-exome
treat­ment.38,41,42 However, “Nothing hap­pens quite by chance. sequenc­ ing identifies novel MPL and JAK2 muta­ tions in tri­ple-neg­ a­
tive
It’s a ques­tion of accre­tion of infor­ma­tion and expe­ri­ence” (Jonas mye­lo­pro­lif­er­a­tive neo­plasms. Blood. 2016;127(3):325-332.
Salk). So, it may be expected that dis­cov­ery of new muta­tions, 15. Marty C, Saint-Martin C, Pecquet C, et al. Germ-line JAK2 muta­tions in the
epi­ge­netic mark­ers, and altered RNA expres­sion pro­file; appli­ kinase domain are respon­si­ble for hered­i­tary thrombocytosis and are resis­
tant to JAK2 and HSP90 inhib­i­tors. Blood. 2014;123(9):1372-1383.
ca­tion of a sin­ gle-cell geno­type; and exploi­ ta­
tion of arti­fi­
cial
16. Borowczyk M, Wojtaszewska M, Lewandowski K, et al. The JAK2 V617F
intel­li­gence to inter­pret geno­mic data might grad­u­ally result in muta­tional sta­tus and allele bur­den may be related with the risk of venous
novel, improved prog­nos­tic scores with a stron­ger impact on throm­bo­em­bolic events in patients with Philadelphia-neg­a­tive mye­lo­pro­
clin­i­cal deci­sion-mak­ing for patients with MPNs. lif­er­a­tive neo­plasms. Thromb Res. 2015;135(2):272-280.
17. Grinfeld J, Nangalia J, Baxter EJ, et al. Classification and per­son­al­ized prog­
no­sis in mye­lo­pro­lif­er­a­tive neo­plasms. N Engl J Med. 2018;379(15):1416-
Conflict-of-inter­est dis­clo­sure 1430.
Alessandro Maria Vannucchi has no rel­e­vant con­flict of inter­est. 18. Ortmann CA, Kent DG, Nangalia J, et al. Effect of muta­tion order on mye­lo­

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022


Paola Guglielmellihas has no rel­e­vant con­flict of inter­est. pro­lif­er­a­tive neo­plasms. N Engl J Med. 2015;372(7):601-612.
19. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prog­no­sis in
pri­mary mye­lo­fi­bro­sis. Leukemia. 2013;27(9):1861-1869.
Off-label drug use
20. Tefferi A, Finke CM, Lasho TL, et al. U2AF1 muta­ tion types in pri­ mary
Alessandro Maria Vannucchi: nothing to disclose. mye­lo­fi­bro­sis: phe­no­typic and prog­nos­tic dis­tinc­tions. Leukemia. 2018;
Paola Guglielmelli: nothing to disclose. 32(10):2274-2278.
21. Guglielmelli P, Lasho TL, Rotunno G, et al. The num­ber of prognostically
Correspondence det­ri­men­tal muta­tions and prog­no­sis in pri­mary mye­lo­fi­bro­sis: an inter­na­
tional study of 797 patients. Leukemia. 2014;28(9):1804-1810.
Alessandro Maria Vannucchi, CRIMM, Center Research and Inno­ 22. Emanuel RM, Dueck AC, Geyer HL, et al. Myeloproliferative neo­plasm (MPN)
vation of Myeloproliferative Neoplasms, University of Florence, symp­tom assess­ment form total symp­tom score: pro­spec­tive inter­na­tional
Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy; assess­ment of an abbre­vi­ated symp­tom bur­den scor­ing sys­tem among
e-mail: amvannucchi@unifi​­.it. patients with MPNs. J Clin Oncol. 2012;30(33):4098-4103.
23. Tefferi A, Barbui T. Polycythemia vera and essen­tial thrombocythemia: 2021
update on diag­no­sis, risk-strat­i­fi­ca­tion and man­age­ment. Am J Hematol.
References 2020;95(12):1599-1613.
1. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revi­sion to the World Health 24. Barbui T, Vannucchi AM, Buxhofer-Ausch V, et al. Practice-rel­e­vant revi­sion
Organization clas­si­fi­ca­tion of mye­loid neo­plasms and acute leu­ke­mia. of IPSET-throm­bo­sis based on 1019 patients with WHO-defined essen­tial
Blood. 2016;127(20):2391-2405. thrombocythemia. Blood Cancer J. 2015;5:e369.
2. Li MM, Datto M, Duncavage EJ, et al. Standards and guide­lines for the inter­ 25. Guglielmelli P, Carobbio A, Rumi E, et al. Validation of the IPSET score for
pre­ta­tion and reporting of sequence var­i­ants in can­cer: a joint con­sen­sus throm­bo­sis in patients with prefibrotic mye­lo­fi­bro­sis. Blood Cancer J.
rec­om­men­da­tion of the Association for Molecular Pathology, Amer­i­can 2020;10(2):21.
Society of Clinical Oncology, and College of Amer­i­can Pathologists. J Mol 26. Guglielmelli P, Gangat N, Coltro G, et al. Mutations and throm­bo­sis in
Diagn. 2017;19(1):4-23. essen­tial thrombocythemia. Blood Cancer J. 2021;11(4):77.
3. Mora B, Giorgino T, Guglielmelli P, et al. Value of cyto­ge­netic abnor­mal­i­ 27. Guglielmelli P, Loscocco GG, Mannarelli C, et al. JAK2V617F var­i­ant allele
ties in post-poly­cy­the­mia vera and post-essen­tial thrombocythemia mye­ fre­quency >50% identifies patients with poly­cy­the­mia vera at high risk for
lo­fi­bro­
sis: a study of the MYSEC pro­ ject. Haematologica. 2018;103(9): venous throm­bo­sis. Blood Cancer J. 2021;11(12):199.
e392-e394. 28. Szuber N, Mudireddy M, Nicolosi M, et al. 3023 Mayo Clinic patients with
4. Tefferi A, Guglielmelli P, Lasho TL, et al. Mutation-enhanced inter­na­tional mye­lo­pro­lif­er­a­tive neo­plasms: risk-strat­i­fied com­par­i­son of sur­vival and
prog­nos­tic sys­tems for essen­tial thrombocythaemia and polycythaemia out­ comes data among dis­ ease sub­ groups. Mayo Clin Proc. 2019;94(4):
vera. Br J Haematol. 2020;189(2):291-302. 599-610.
5. Gangat N, Jadoon Y, Szuber N, et al. Cytogenetic abnor­mal­i­ties in essen­tial 29. Passamonti F, Thiele J, Girodon F, et al. A prog­nos­tic model to pre­dict sur­
thrombocythemia: Clinical and molec­u­lar cor­re­lates and prog­nos­tic rel­e­ vival in 867 WHO-defined essen­tial thrombocythemia at diag­no­sis: a study
vance in 809 infor­ma­tive cases. Blood Cancer J. 2022;12(3):44. by the IWG-MRT. Blood. 2012;120(6):1197-1201.
6. Tefferi A, Rumi E, Finazzi G, et al. Survival and prog­ no­sis among 1545 30. Ali H, Bacigalupo A. 2021 Update on allo­ge­neic hema­to­poi­etic stem cell
patients with con­tem­po­rary poly­cy­the­mia vera: an inter­na­tional study. trans­plant for mye­lo­fi­bro­sis: a review of cur­rent data and appli­ca­tions on
Leukemia. 2013;27(9):1874-1881. risk strat­i­fi­ca­tion and man­age­ment. Am J Hematol. 2021;96(11):1532-1538.
7. Guglielmelli P, Pacilli A, Rotunno G, et al; AGIMM Group. Presentation and 31. Guglielmelli P, Lasho TL, Rotunno G, et al. MIPSS70: muta­tion-enhanced
out­come of patients with 2016 WHO diag­no­sis of prefibrotic and overt pri­ inter­na­tional prog­nos­tic score sys­tem for trans­plan­ta­tion-age patients
mary mye­lo­fi­bro­sis. Blood. 2017;129(24):3227-3236. with pri­mary mye­lo­fi­bro­sis. J Clin Oncol. 2018;36(4):310-318.
8. Tefferi A, Nicolosi M, Mudireddy M, et al. Revised cyto­ge­netic risk strat­i­fi­ca­ 32. Tefferi A. Primary mye­lo­fi­bro­sis: 2021 update on diag­no­sis, risk-strat­i­fi­ca­
tion in pri­mary mye­lo­fi­bro­sis: anal­y­sis based on 1002 infor­ma­tive patients. tion and man­age­ment. Am J Hematol. 2021;96(1):145-162.
Leukemia. 2018;32(5):1189-1199. 33. Tefferi A, Guglielmelli P, Lasho TL, et al. MIPSS70+ ver­sion 2.0: muta­tion and
9. Tang G, Hidalgo Lopez JE, Wang SA, et al. Characteristics and clin­i­cal sig­ kar­yo­type-enhanced inter­na­tional prog­nos­tic scor­ing sys­tem for pri­mary
nif­i­cance of cyto­ge­netic abnor­mal­i­ties in poly­cy­the­mia vera. Haematolog- mye­lo­fi­bro­sis. J Clin Oncol. 2018;36(17):1769-1770.
ica. 2017;102(9):1511-1518. 34. Tefferi A, Guglielmelli P, Nicolosi M, et al. GIPSS: genet­i­cally inspired prog­
10. Klampfl T, Harutyunyan A, Berg T, et al. Genome integ­rity of mye­lo­pro­lif­er­ nos­tic scor­ing sys­tem for pri­mary mye­lo­fi­bro­sis. Leukemia. 2018;32(7):1631-
a­tive neo­plasms in chronic phase and dur­ing dis­ease pro­gres­sion. Blood. 1642.
2011;118(1):167-176. 35. Passamonti F, Giorgino T, Mora B, et al. A clin­i­cal-molec­u­lar prog­nos­tic
11. Marcellino BK, Hoffman R, Tripodi J, et al. Advanced forms of MPNs are model to pre­dict sur­vival in patients with post poly­cy­the­mia vera and post
accom­pa­nied by chro­mo­somal abnor­mal­i­ties that lead to dysregulation of essen­tial thrombocythemia mye­lo­fi­bro­sis. Leukemia. 2017;31(12):2726-2731.
TP53. Blood Adv. 2018;2(24):3581-3589. 36. National Comprehensive Cancer Network. NCCN Clinical Practice Guide­
12. Khoury JD, Solary E, Abla O, et al. The 5th edi­tion of the World Health Orga­ lines in Oncology. Myeloproliferative Neoplasms. Version 2.2022. Plymouth
nization Classification of Haematolymphoid Tumours: mye­loid and his­tio­ Meeting, PA: NCCN; 2022.
cytic/den­dritic neo­plasms. Leukemia. 2022;36(7):1703-1719. 37. Gagelmann N, Ditschkowski M, Bogdanov R, et al. Comprehensive clin­i­cal-
13. Arber DA, Orazi A, Hasserjian RP, et al. International Consensus Classifica­ molec­u­lar trans­plant scor­ing sys­tem for mye­lo­fi­bro­sis under­go­ing stem
tion of Myeloid Neoplasms and Acute Leukemia: inte­grat­ing mor­pho­log­ cell trans­plan­ta­tion. Blood. 2019;133(20):2233-2242.

Molecular prog­nos­ti­ca­tion in MPN | 233


38. Gisslinger H, Klade C, Georgiev P, et al; PROUD-PV Study Group. Ropegin­ 41. Vannucchi AM, Verstovsek S, Guglielmelli P, et al. Ruxolitinib reduces JAK2
terferon alfa-2b ver­sus stan­dard ther­apy for polycythaemia vera (PROUD- p.V617F allele bur­den in patients with poly­cy­the­mia vera enrolled in the
PV and CONTINUATION-PV): a randomised, non-infe­ri­or­ity, phase 3 trial RESPONSE study. Ann Hematol. 2017;96(7):1113-1120.
and its exten­sion study. Lancet Haematol. 2020;7(3):e196-e208. 42. Kiladjian J-J, Cassinat B, Chevret S, et al. Pegylated inter­ feron-alfa-2a
39. Barbui T, Vannucchi AM, De Stefano V, et al. Ropeginterferon alfa-2b induces com­plete hema­to­logic and molec­u­lar responses with low tox­ic­ity
ver­
sus phle­ bot­omy in low-risk patients with polycythaemia vera (Low- in poly­cy­the­mia vera. Blood. 2008;112(8):3065-3072.
PV study): a multicentre, randomised phase 2 trial. Lancet Haematol.
2021;8(3):e175-e184.
40. Coltro G, Rotunno G, Mannelli L, et al. RAS/CBL muta­tions pre­dict resis­
tance to JAK inhib­i­tors in mye­lo­fi­bro­sis and are asso­ci­ated with poor prog­ © 2022 by The Amer­i­can Society of Hematology
nos­tic fea­tures. Blood Adv. 2020;4(15):3677-3687. DOI 10.1182/hema­tol­ogy.2022000339

Downloaded from http://ashpublications.org/hematology/article-pdf/2022/1/225/2021786/225vannucchi.pdf by guest on 12 December 2022

234 | Hematology 2022 | ASH Education Program

You might also like