Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Received: 30 April 2023 | Accepted: 21 July 2023

DOI: 10.1111/bjh.19010

REVIEW

Updates on accelerated and blast phase myeloproliferative


neoplasms: Are we making progress?

Dina Mahdi1 | Jessica Spiers1 | Alexandros Rampotas1 | Nicola Polverelli2 |


Donal P. McLornan1

1
Department of Haematology, University
College Hospital, London, UK Summary
2
Unit of Blood Diseases and Stem Cell Management approaches for accelerated and blast phase myeloproliferative neo-
Transplantation, University of Brescia, plasms remain challenging for clinicians and patients alike. Despite many thera-
Brescia, Italy
peutic advances, outcomes for those patients who are not allogeneic haematopoietic
Correspondence cell transplant eligible remain, in general, very poor. Estimated survival rates for
Donal P. McLornan, Department of such blast phase patients is frequently reported as less than 6 months. No specific
Haematology and Stem Cell Transplantation,
University College London Hospitals, 3rd immunological, genomic or clinicopathological signature currently exists that ac-
Floor West, 250 Euston Road, London NW1 curately predicts the risk and timing of transformation, which frequently induces a
2PG, UK. high degree of anxiety among patients and clinicians alike. Within this review arti-
Email: donal.mclornan@nhs.net
cle, we provide an up-­to-­date summary of current understanding of the underlying
pathogenesis of accelerated and blast phase disease and discuss current therapeutic
approaches and realistic outcomes. Finally, we discuss how the horizon may look
with the introduction of more novel agents into the clinical arena.

K EY WOR DS
accelerated phase, AML, blast phase, essential thrombocythaemia, JAK2, MPN, polycythaemia vera

I N T RODUC T ION disease and discuss how novel agents may change the future
therapeutic landscape.
The last two decades have observed major advances in di-
agnostic and prognostic algorithms and therapeutic ap-
proaches across the range of ‘Philadelphia chromosome DE F I N I T IONS
negative’ chronic phase myeloproliferative neoplasms
(MPNs). However, despite these advances, the management MPN AP is classically defined as a persistence of 10%–­19%
of MPN in accelerated phase (AP) and blast phase (BP), par- blasts in the peripheral blood (PB) or bone marrow (BM)
ticularly for those patients who are not considered transplant and MPN BP by ≥20% blasts.2 For transformed cases, most
eligible or lack a suitable donor, remains very challenging. commonly the disease progresses from chronic phase to
There remains a lack of clear progress despite many novel MPN BP via AP but direct blastic transformation can also
agents entering the clinical arena. Overall survival (OS) rates occur. Hence, MPN AP is part of a dynamic progressive dis-
for those with BP disease remain sobering, with frequently ease spectrum leading towards BP disease at variable rates.
estimated survivals of less than 6 months for those treated An outstanding question has been how increasing blasts
with non-­curative intent.1 Within this practical review, we in the PB or BM, not yet reaching the assigned ‘cut off’ of
discuss the pathogenesis of disease progression to highlight ≥10% to define AP, can affect outcomes. Masarova and col-
potential therapeutic opportunities, provide an overview leagues evaluated if there was a differential effect on sur-
of contemporary management approaches for MPN AP/BP vival outcomes determined by blast % in a cohort of 1314

Dina Mahdi and Jessica Spiers are joint first authors.

© 2023 British Society for Haematology and John Wiley & Sons Ltd.

Br J Haematol. 2023;00:1–13.  wileyonlinelibrary.com/journal/bjh | 1


13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2 |    UPDATES ON ACCELERATED AND BLAST PHASE MYELOPROLIFERATIVE NEOPLASMS

myelofibrosis (MF) patients.3 In particular, to understand scoring system for transplant-­eligible patients (MIPSS70)
the impact of <10% PB blasts compared to MPN AP. Here, and MIPSS70-­version 2.0 scores were developed from a co-
patients with ≥5% PB or BM blasts actually had similar ad- hort of PMF patients, incorporating molecular information
verse features to those patients with MPN AP, displayed a to refine prognostication.15,16 For example, the category
twofold increase in leukaemic transformation compared identified as very high risk in MIPSS70-­plus has a 13.3-­fold
to those with lower blast %, and had comparable survival increased risk of BP, with an incidence of 23%, compared to
to MPN AP. This highlights that therapeutic intervention 11% in the low-­risk category.
should be considered early for such patients. These findings, The prognostic scores described above include the poorly
confirmed also by other groups, have led to suggestions that standardised variable of PB blast count assessment by mor-
the blast % for defining MPN AP could be lowered to 5% phology, which remains very operator dependent and indeed
although this has not yet been widely accepted.3-­5 the ‘cut off’ value as a significant variable in the model dif-
fers between the scoring systems. A recent study focussed
on the utility of multiparameter flow cytometric (MFC) as-
R ISK FAC TOR S A N D PAT HOGE N E SIS sessment of PB CD34+ count and degree of decreased side
OF AC CE L E R AT E D A N D BL A ST scatter of neutrophils in 363 MF patients. The authors cate-
PH A SE M PN gorised the group into three arbitrarily defined groups based
on these two findings and relative thresholds; MFC ‘low’,
The risks of transformation differ across the MPN spectrum. ‘intermediate’ or ‘high’, which independently correlated
The cumulative hazard of MPN BP for polycythaemia vera with survival. Next, they integrated these findings into the
(PV) is estimated at 2.3% and 5.5% and for essential throm- IPSS and MIPSS70+ models in place of morphological as-
bocythaemia (ET) at 0.7% and 2.1% at 10 and 15 years re- sessment of blast count. Patients were reclassified according
spectively.6,7 The risk of MPN BP is highest for primary MF to these MFC-­enhanced models, highlighting significantly
(PMF), in particular for those with so called ‘high molecular better risk stratification performance. Of note, this was not
risk’ (HMR) mutations, estimated between 10% and 20% at the case in the MYSEC-­PM model, perhaps in part due to
10 years post-­diagnosis.8 Risk factors for transformation, in smaller sample size.20
general, are based on clinicopathological features and, more A recent study evaluating risks of transformation in a co-
recently, molecular profiling. For PV, older age, leucocytosis, hort of 1306 patients with PMF, where 149 (11%) developed
an abnormal karyotype, increasing reticulin fibrosis, his- MPN ­BP, suggested a three-­tier risk model.21 Time-­to-­event
toric use of P32 and the presence of high-­risk mutations such Cox analysis demonstrated BP prediction risk factors within
as TP53, RUNX1, ASXL1, EZH2 and SRSF2, among others, 5 years of diagnosis for: age >70 years (hazard ratio [HR] 2.1),
are associated with a higher risk of transformation.1,7–­10 For the presence of moderate/severe anaemia (HR 1.9), circulat-
ET, again older age, leucocytosis, anaemia, extreme throm- ing blasts ≥3% (HR 3.3), and the presence of IDH1 mutations
bocytosis (>1000 × 109/L), increasing reticulin deposition (HR 4.3), SRSF2 mutations (HR 3.0) and ASXL1 mutations
and mutations in TP53, SRSF2, ASXL1, EZH2, U2AF1 and (HR 2.0). The high-­risk category had a BP incidence of 57%,
IDH1/2 have been detailed across studies as increasing trans- compared with only 8% for the low-­risk category.
formation risk to varying degrees.1,6,8,11–­13 The exact genetic, epigenetic and immunological events
For PMF, risk factors include age, increasing throm- driving disease acceleration and transformation still re-
bocytopenia, rising blasts in the PB, high-­risk cytogenetic quires further elucidation (Figure 1). Numerous studies have
anomalies such as monosomy 7, chromosome 17p deletions; highlighted the differences in the mutational landscape of
isochrome (17q), inv (3)/3q21, 12p−/12p11.2, 11q−/11q23, MPN BP compared to de novo acute myeloid leukaemia
+8 and complex or monosomal karyotypes. HMR muta- (AML). While the most common mutations seen in AML
tions such as ASXL1, EZH2, SRSF2, IDH1/2 and U2AF1, or (NPM1 and FLT3-­ITD) are rarely seen in MPN BP, there
being ‘triple-­negative’ for driver mutations, also increases is enrichment of mutations across a range of epigenetic
the risk in PMF.1,9,14-­17 Contemporary MF-­specific scores can modifiers (TET2, DNMT3A, IDH1/2), transcription factors
aid prediction of general risk. The Dynamic International (TP53, RUNX1, IKZF1) and splicing factors (SF3B1, U2AF1,
Prognostic Scoring System (DIPSS) was developed to predict SRSF2).11–­13,22–­27 LNK, PTP1N1 and MYC anomalies also ap-
survival in PMF using five clinicopathological factors (age pear to be overrepresented in MPN BP.25,28 The increased
≥65 years, haemoglobin <100 g/L, leucocytes ≥25 × 109/L, number of cumulative molecular mutations, as well as cyto-
circulating blasts ≥1% and constitutional symptoms), appli- genetic complexity, suggest that genomic instability plays an
cable at any stage in the disease course. Patients within the important role in transformation.29
intermediate-­2 and high-­risk categories have an estimated It is well documented that leukaemic clones with wild-­
7.8-­and 24.9 fold increased risk of MPN BP, respectively, type JAK2 can arise in patients whose chronic phase dis-
compared to those in the low-­risk category.18 This model ease harbours the JAK2 V617F mutation. 25,28 Two possible
was subsequently refined, as DIPSS-­plus, by addition of un- models of leukaemic transformation in these patients
favourable karyotype, thrombocytopenia and transfusion have been hypothesised. 28,30,31 In the first, an antecedent
dependency, all of which are independent risk factors for BP clone harbouring earlier genetic aberrations (e.g. in TET2
in MF.19 The mutation-­enhanced international prognostic and DNMT3) promoting a growth advantage, genomic
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAHDI et al.    | 3

F I G U R E 1 Models of myeloproliferative neoplasm (MPN) progression: MPN are frequently initiated by a driver mutation leading to the
development of the ‘MPN driver clone’. Additional phenomena driven by ‘chronic inflammation’ and the self-­reinforcing haematopoietic stem cell niche
paralleled with genomic instability can lead to the acquisition of further proleukaemic mutations. The increase in the mutational burden can lead to
the development of a ‘leukaemic stem cell’, which will drive the accelerated/blastic phase of the disease. This progression is depicted in a linear fashion,
but stepwise changes can accelerate the progression. Alternative pathways to MPN progression are also depicted. Pre-­existing clonal haematopoiesis
mutations can drive expansion of an aggressive clone if a secondary MPN mutation occurs, while ‘triple negative’ MPNs can also progress by acquisition
of further pro-­leukaemic mutations.
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4 |    UPDATES ON ACCELERATED AND BLAST PHASE MYELOPROLIFERATIVE NEOPLASMS

instability or both gives rise to two separate clones—­the Rodriguez-­Meira and colleagues have recently reported
leukaemic clone and the underlying MPN clone. Genetic on single-­cell multiomic analysis of haematopoietic stem/
mutations implicated in clonal haematopoiesis and fre- progenitor cells (HSPC) in TP53-­mutated AML arising
quently present in MPN can precede JAK2 V617F and from MPN.35 ‘TARGETseq’ analysis was performed on
associate with genomic instability, giving rise to a sub- Lin−CD34+ HSPCs from 14 TP53-­mutated AML patients
clone with multiple mutations which ultimately becomes and 9 age-­matched controls. In all cases, the dominant clone
the dominant leukaemic clone. The question of whether showed loss of wild-­type TP53 through varying patterns
a JAK2 V617F-­positive clone may give rise to wild-­type of clonal evolution. Here, chronic inflammation supressed
JAK2 AML, later losing this mutated allele or converting TP53 wild-­t ype HSPC while, in parallel, the survival advan-
to wild-­type JAK2 through mitotic recombination, gene tage of TP53 mutant cells was augmented. Of interest, three
conversion or deletion, has been excluded in a number distinct clusters of HPSC in TP53-­sAML were described, one
of clonality studies using paired chronic and BP sam- of which was characterised by overexpression of erythroid
ples. 28,30,31 In the second model, the leukaemia arises genes and another characterised by defective differentiation
from a phylogenetically unrelated haematopoietic stem and so-­called ‘quiescence signatures’.
cell from the MPN clone, perhaps reflecting DNA dam- The utilisation of patient-­derived xenograft (PDX) mu-
age from mutagenic treatments used in the chronic phase rine models that robustly engraft patient cells into immu-
of MPN, although this is unlikely to explain the majority nodeficient mice can increasingly inform on the complexity
of cases, given that contemporary care generally involves of MPN transformation events. For example, the human-
agents not known to associate with leukaemogeneis. 28 ised murine model described by Celik and colleagues
Leukaemic clones, which do harbour the driver mutation, reproduced patient genomic complexity and hierarchy,
are also seen, and likely arise due to the acquisition of ad- pathological features such as reticulin deposition, and can
ditional genomic aberrations. 30 aid prediction/characterisation of clonal evolution events.36
Luque Paz and colleagues described differential blastic Most importantly, these models are amenable to pharmaco-
transformation events at a molecular level for 49 patients logical and genomic manipulation to investigate therapeutic
with BP PV or ET utilising sequencing data.12 The most fre- vulnerability.
quent mutations were in TP53, TET2, RUNX1, ASXL1 and While there appears to be a significant association be-
EZH2, with no significant differences between PV and ET as tween inflammation and more advanced diseases phases,
regards incidence. Hierarchical classification was used to de- the exact interaction between the MPN clone, the adap-
lineate three groups of patients with different times to trans- tive and innate immune system and the bone marrow
formation. So-­called short-­term transformations (median niche requires further clarification. A number of streams
time to transformation 3-­years) occurred in patients with a of work have highlighted the link between disease-­
complex mutational landscape, with a median of seven mu- associated inflammation and risk of transformation. 37
tations, and were enriched for the presence of mutations in Increased levels of a range of cytokines including IL-­2 ,
RUNX1, IDH1/2, U2AF1 and TET2. Of note, there were no IL-­6 , sIL-­2RA and IL-­8 have been shown to significantly
TP53 mutations detected. In contrast, those demonstrat- associate with transformation to MPN BP. 37,38 Moreover,
ing a longer time to transformation (median 21 years) were generation of reactive oxygen species may contribute
enriched for TP53, NRAS and BCORL1 mutations in a less to disease progression by both augmenting JAK/STAT
complex mutational landscape. There was an equal distribu- signalling and inducing genomic instability, laying the
tion of ASXL1 mutations in both groups. Overall, the most foundation for acquisition of additional mutations. 24,37,39
frequently mutated gene at the time of blast transformation ‘Multiomic’ approaches are hence required to understand
was TP53 (45%). It is important to note that previous work the complex interaction between the bone marrow micro-
has suggested that TP53 mutations at a low allelic burden environment, ‘immunome’ and acquired genomic insta-
can be present for many years in chronic phase, while after bility/mutational landscape.
loss of the wild-­t ype TP53 allele, rapid clonal expansion and
leukaemic transformation frequently ensues.28,32 In this re-
gard, Li et al. recently evaluated a number of different TP53 OV E RV I E W OF T R E ATM E N T
allelic configurations in JAK2V617F/TP53 mutant MPN A PPROACH E S BA SE D ON F I T N E S S
murine models.33,34 Mice with biallelic inactivation of TP53 A N D T R A NSPL A N T E L IGI BI L I T Y
succumbed to rapid leukaemic transformation, of note de-
veloping a pure erythroid leukaemia which arose from the Current treatment options for MPN AP/BP still remain
megakaryocyte–­ erythroid progenitor (MEP) population. unsatisfactory overall and allo-­HCT represents the only
This work additionally highlighted the importance of bone curative strategy. Long-­term survival after allo-­HCT has
morphogenetic protein 2 (BMP2)/SMAD pathway activation been reported to range between 30% and 70% in retro-
in aberrant MEP self-­renewal and leukaemic initiation po- spective studies, dependent on patient-­ , disease-­and
tential, contributing to transformation, and also that target- transplant-­specific factors, and there is a trend towards
ing components of the DNA repair pathways (pCDC2 and improvements in allo-­ HCT outcomes in more recent
PARP) could be a rationale potential therapeutic strategy. years.40,41 However, considering the advanced age of
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAHDI et al.    | 5

many MPN AP/BP patients, allo-­HCT can be offered to Unfortunately, HMA therapy has not been prospec-
only a minority. At present, in some centres, the use of tively evaluated to examine survival benefit in MPN AP/
chronological age alone is largely outdated, and biologi- BP or in direct comparison with intensive chemotherapy;
cal fitness is of key importance. In addition, the role of current evidence base is drawn from small, frequently het-
comorbidities, as evaluated by the Haematopoietic Stem erogeneous cohorts. In 2010, the Groupe Francophone Des
Cell Transplantation Comorbidity Index (HCT-­CI), ap- Myelodysplasies reported on 26 MPN ­BP patients who had
pears to be of less importance among elderly subjects received azacytidine monotherapy.47 The overall response
compared to younger, particularly in transplant cohorts rate (ORR) was 38%, with a median OS of 8 months, and
from the most recent era.42 Novel multidimensional and combined CR/CR with incomplete haematological recovery
comprehensive geriatric assessment tools are becoming a (CR/CRi) was achieved in 12%. Other studies of either single
mainstay for the evaluation of eligibility for allo-­HCT in agent azacytidine or decitabine suggested OS estimates be-
the more elderly population.43,44 A modern approach to tween approximately 7 and 10 months, with CR/CRi rates of
patients with MPN AP/BP should hence include a com- less than 25%.48,49 In a recent meta-­analysis, monotherapy
prehensive evaluation of all patients up to 70–­75 years of with azacytidine or decitabine was found to have an ORR of
age; disease-­and patient-­specific characteristics should 38%, with significant heterogeneity amongst the studies.50
be considered along with patients' preferences and an Pooled ORR analysis did not show a statistically significant
overview of social and family support. Early referral is difference between azacytidine (42%) and decitabine (27%)
necessary with identification of donors. Achievement responses, suggesting no clear ‘winner’. Treating clinicians
of disease control, that is achieving complete remission should be aware that response are often short lived, ongoing
(CR) prior to transplant, and tailoring of the transplant transfusion dependency is frequent and HMA frequently do
platform can maximise allo-­HCT outcomes. Clinically not address MPN AP/BP-­related symptom burdens.
challenging cases remain where the decision-­ making
process is less than straightforward, including potential
allo-­HCT eligible patients where one or more comorbidi- Hypomethylating agents in combination with
ties exist that will predict higher non-­relapse mortality JAK inhibitors
(NRM), patients towards the upper end of ‘acceptable’
chronological age, the presence of a complex/monosomal Small case series initially reported the potential efficacy of
karyotype, particularly accompanied by the presence of ruxolitinib in combination with either HMA or low-­dose
a TP53 mutation, as the relapse risk may be unacceptable cytarabine.51 A phase 2 trial of ruxolitinib and decitabine
and the presence of difficult social or financial circum- enrolled 25 patients with MPN A ­ P/BP with an ORR of 44%;
stances. Clearly, MDT discussions are paramount in such median OS for all patients on study was 9.5 months and
cases to individualise such recommendations. hence not significantly longer than monotherapy studies.52
Another phase 1/2 study of the same combination enrolled
12 and 15 patients in phases 1 and 2, respectively. A total
T H E R A PY of 18 patients were treated at the recommended phase 2
dose (RP2D; 50 milligrams (mg) twice daily of ruxolitinib,
Hypomethylating agents alone along with 20 mg/m2/day of decitabine, given for 5 days per
cycle) with an ORR of 61% in these 18 patients, and a median
The hypomethylating agents (HMAs), azacytidine and survival of 8.4 months. Of note, four patients were bridged
decitabine, are frequently considered as monotherapy to allo-­HCT.53 Regarding azacytidine, the UK generated
for MPN AP/BP patients unsuitable for intensive chem- phase 1b PHAZAR study evaluated the combination of
otherapy and allo-­HCT. Utilisation in this setting has azacytidine and ruxolitinib in advanced MPN with an aim
originated from results generated in the myelodysplastic to establish the maximum tolerated dose, safety and clinical
neoplasm-­A ML arena. Practically, HMA can be favour- activity of this combination.54 A total of 34 patients were
able treatment options in MPN AP/BP due to good tol- recruited (15 MPN BP and 19 MPN AP) and a modified
erability, including for older patients or those with poor two-­stage continual reassessment model was implemented
performance status, paralleled with their potential abil- as regards dose escalation. Median number of azacytidine
ity to induce transient disease control in some. From a and ruxolitinib cycles received were 4 and 3 respectively;
biological perspective, hypermethylation of p15 and p16 overall, 20/34 patients were evaluable for disease response
genes has been shown in MPN BP, but not chronic phase, assessment. Toxicity was acceptable and as expected. A total
with resultant myeloid differentiation block, suggesting of 10/20 patients achieved either a PR or CR. Meaningful
that agents targeting dysregulated methylation status may red cell transfusion reduction was also seen in 3/14 patients.
be useful, in addition to demonstration of in vitro sensi- Median duration responses were relatively short: MPN AP
tivity of MPN-­derived cells with aberrant DNA methyla- 322 days and MPN BP 199 days. Longer term outcomes are
tion to HMAs.45,46 awaited but the approach is promising, with some long-­term
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6 |    UPDATES ON ACCELERATED AND BLAST PHASE MYELOPROLIFERATIVE NEOPLASMS

responses identified for MPN AP. The meta-­ analysis by Allo-­HCT for MPN AP/BP: Where are we now?
Chen et al., additionally compared single agent HMA versus
HMA with ruxolitinib.50 Here, responses with HMA and Multiple studies have evaluated the efficacy and safety of allo-­
ruxolitinib showed higher ORR than monotherapy (45% vs. SCT in the context of MPN AP/BP. The MD Anderson group
30%, p = 0.04) but no difference in rates of CR/CRi. reported, for the first time, the superiority of a transplant
approach over IC without allo-­SCT consolidation in MPN
BP.57 Long-­term disease control was only achievable with
Intensive chemotherapy allo-­SCT, with an impressive 73% OS rate after a median fol-
low-­up of 31 months. In contrast, patients who achieved CR
Historically, intensive chemotherapy (IC) regimens for after chemotherapy but did not undergo transplant had an
MPN ­BP generally mimicked those used for de novo AML. OS of 0%. These findings primarily supported the potency
Standard of care, in general, remains induction chemother- of the so-­called graft-­versus-­leukaemia (GVL) effect, in this
apy over HMA with a view to allo-­HCT in those deemed fit high-­risk setting. Baron et al. confirmed this observation
enough to tolerate potential treatment-­related toxicity. Few by leading an extensive analysis of the connection between
prospective studies have directly addressed comparison of graft-­versus-­host disease (GVHD) and GVL effects on a co-
regimens. While one small retrospective study evaluating hort of 2414 cases of secondary leukaemia, including cases
a heterogeneous range of induction and consolidation ap- arisen from antecedent MPNs, who underwent allo-­HCT in
proaches suggested a trend towards improved responses first or second CR.58 Limited and extensive chronic GVHD
following combinatorial therapy with mitoxantrone, etopo- (cGVHD) associated with a lower relapse incidence; how-
side and high-­dose cytarabine (NOVE-­HIDAC), this has ever, due to a higher risk of NRM among patients with ex-
not been supported in other studies.55 In this retrospective tensive cGHVD, only limited cGVHD translated into better
study, 38/75 patients underwent IC with a view to allo-­HCT. OS. The effect of GVL is also supported by studies highlight-
Of these, 76% of patients responded (18 with CR/CRi and ing objective response rates following donor lymphocyte in-
11 reverted to chronic phase); 17 patients subsequently un- fusions (DLIs) for relapse post allo-­HCT.59,60
derwent allo-­HCT. OS was significantly better in those re- One recent study focused on RIC allo-­HCT outcomes
sponding patients undergoing allo-­HCT compared to those for MPN AP, reporting on the outcomes of 35 patients com-
not-­transplanted (47% compared to 15% (p = 0.03). No sig- pared with those in chronic phase (n = 314). The estimated 5-­
nificant survival difference was evident between those re- year OS for the AP group was 65% versus 64% for the chronic
sponding to IC alone who did not undergo transplant and phase cohort; however, the AP cohort had a higher 5-­year
those treated non-­intensively (median survival of 9.4 and relapse incidence at 30%.
6.6 months, respectively; p = 0.18). Globally, multiple de novo Many centres and working groups have reported out-
AML style induction strategies are used for these patients comes of MPN BP allo-­HCT in Europe, Japan and North
despite lack of evidence or consensus recommendations. America (summarised in Table 1). With the limits of marked
Combinatorial approaches of intensive chemotherapy and heterogeneity in terms of induction treatments and trans-
venetoclax are also being evaluated. plant characteristics across studies, OS did not exceed
A large study of two separate cohorts (Mayo-­AGIMM) 20–­30% in the longer term. In particular, a retrospective
totalling 410 patients with MPN BP evaluating outcomes analysis by the Center for International Blood and Marrow
over time found no significant improvement had been made Transplant Research (CIBMTR) of 6030 AML transplanted
in the 15 years covered in this study period.1 In the first co- patients, including 177 with MPN BP, highlighted a worse
hort, 66/125 patients received IC, with 59% achieving CR OS and higher relapse risk for MPN BP compared to patients
or CRi compared to <5% with other agents such as HMAs. in remission from de novo AML or post-­myelodysplastic
In the second, 48/162 patients received IC with a combined neoplasm AML undergoing allo-­HCT.61 Longer duration
CRCRi rate of 35.4%. Of note, while the OS benefit of allo-­ of the underlying disease, older age and higher genomic
HCT over no transplant was confirmed, this study failed complexity leading to lower rates of CR may all contribute,
to demonstrate a significant difference in survival between highlighting the need for improved allo-­HCT strategies.
those achieving CR or CRi and those undergoing allo-­SCT Very recently, the Chronic Malignancies Working Party of
(irrespective of achievement of CR or CRi prior to trans- the EBMT published the largest transplant cohort of MPN
plant), with HR 0.6 (95% CI 0.3–­1.1) for allo-­HCT versus BP patients available to date.41 This study confirmed that
CR/CRi without allo-­ HCT. Other retrospective studies approximately 1/3 of patients could be cured with transplant
have confirmed no OS benefit between IC treated versus (32% OS at 5-­years) and revealed a gradual improvement in
non-­intensive treated cohorts without early allo-­HCT.56,57 outcomes over the years. Compared to previous experiences,
Optimal induction regimen and required depth of response the prognostic role of donor match was less evident, with no
require further evaluation but is limited by a lack of co-­ differences in OS according to donor type, although NRM
operative group trials. It is clear that novel approaches are was increased in mismatched related and unrelated donor
needed to get this challenging group of patients in remis- allo-­HCT. Patients with a good performance status who
sion safely followed by allo-­HCT, as this currently offers the achieved pretransplant CR had an expected survival of 60%
best chance of long-­term cure. at 3-­years, highlighting the potential benefits of utilising
TA BL E 1 List of the available studies on the role of allo-­SCT in blast phase MPN.

Transplant Median age


Author Year Pts period at allo-­SCT Pretreatment Conditioning Donor CIR NRM OS Favourable factors
MAHDI et al.

41
Orti 2023 663 2005–­2019 60 Chemo (49%) MAC (35%) MRD (28%) 51% at 5 years 24% at 5 years 32% at 5 years • KPS ≥90
HMA (6%) UD (63%) • CR at allo
Ruxolitinib (3%) MMRD (9%) • Earlier transplant period
Other (33%)
Shah87 2021 43 2001–­2016 59 Chemo (83%) MAC (67%) MSD (40%) 43% at 4 years 24% at 4 years 38% at 4 years • Earlier transplant period
HMA (23%) MUD (60%)
JAKi (21%)
None (16%)
Gupta61 2020 177 2001–­2015 59 Chemo (66%) MAC (53%) MRD (31%) 63% at 5 years (CR) 21% at 5 years (CR) 22% at 5 years (CR) • Younger age
HMA (4%) UD (62%) 71% (no CR) 23% (no CR) 11% (no CR) • Female gender
MMRD (7%) • KPS ≥90
• MRD
• Earlier transplant period
• Absence of TP53 mutation
• Absence of adverse
cytogenetics
Kröger88 2019 422 2000–­2014 59 NA MAC (34%) MRD (36%) 50% at 3 years 25% at 3 years 33% at 3 years • MRD
UD (64%) • BM as a source
• CR at allo
• Younger age
• KPS ≥90
• CMV seronegative
• Normal karyotype
• Absence of ex vivo TCD
Takagi89 2016 39 2000–­2013 57 Chemo (69%) MAC (38%) MRD (21%) 34.4% at 2 years 34.2% at 2 years 29.2% at 2 years • None identified
Untreated (31%) UD (38%)
UCB (41%)
Cahu90 2014 60a 2000–­2010 57 NA MAC (23%) MRD (38%) 68% at 3 years 22% at 3 yeares 18% at 3 years • CR at allo
UD (62%)
Alchalby40 2014 46 NA 55 Chemo (91%) MAC 43% MRD (37%) 47% at 3 years 28% at 1 year 33% at 3 years • CR at allo (on LFS)
UD (61%)
MMRD (2%)
Kennedy55 2013 17 1998–­2011 NA Chemo (100%) MAC (47%) MRD (70%) 24% 47% 47% at 2 years NA
UD (30%)
Ciurea91 2010 14 1994–­2008 59 Chemo (93%) MAC (21%) MRD (57%) 23% at 2 years 29% at 2 years 49% at 2 years None identified
MUD (29%)
MMRD (14%)
Tam57 2008 8 1994–­2007 57 Chemo (75%) NA MRD (63%) 13% 25% 73% at 5 years NA
UD (37%)

Abbreviations: BM, bone marrow; chemo, chemotherapy; CIR, cumulative incidence of relapse; CR, complete remission; HMA, hypomethylating agents; JAKi, JAK-­i nhibitors; KPS, Karnofsky Performance Score; LFS, leukaemia-­f ree
survival; MAC, myeloablative conditioning regimen; MMRD, mismatched related donor; MPN, myeloproliferative neoplasm; MRD, matched related donor; NA, not available; NRM, non-­relapse mortality; OS, overall survival; Pts,
  

patients; RIC, reduced intensity conditioning; TCD, T-­cell depletion; UCB, unit of cord blood; UD, unrelated donor.
|

a
7

13 Patients with MDS/MPN were included.

13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
8 |    UPDATES ON ACCELERATED AND BLAST PHASE MYELOPROLIFERATIVE NEOPLASMS

effective pretransplantation therapies that preserve recipient phase I trial gaining a response following therapy with this
fitness. In this regard, there is a general agreement among combination alongside chemotherapy, with some patients
the studies regarding the importance of pretransplant CR. being successfully bridged to allo-­HCT or chimeric antigen
However, MPN BP patients demonstrate lower CR rates receptor T-­cell (CAR-­T) therapy.65 Given such potential syn-
compared to de novo AML, in part related to the presence of ergy, data is urgently required in the MPN AP/BP setting. In
high-­risk cytogenetic or molecular features (e.g. TP53 mu- this regard, a phase I study (NCT05455294) has been opened
tations), the prevalence of which are more frequent in this investigating the triplet combination of navitoclax, veneto-
patient group as described above.25,62 The presence of such clax and decitabine across high-­risk myeloid neoplasms, in-
alterations predicts a lower propensity to reach deep and du- cluding MPN AP/BP.
rable responses, even in the context of novel approaches.56,63
To date, several questions remain unresolved. Which
pretransplant therapy is preferable for MPN BP remains an Liposomal daunorubicin and cytarabine
ongoing matter of debate, as no prospective and randomised
clinical trials have been designed with an intention to get to Liposomal cytarabine–­ daunorubicin is a combination of
allo-­HCT. Also, the extent and depth of response required daunorubicin and cytarabine in a 1:5 molar ratio. Ilyas and
before transplant remains unknown. From the transplant colleagues evaluated efficacy of this agent in a small cohort
perspective, the role of conditioning intensity and stem cell of MPN BP patients (n = 12), with a median age of 63 years.66
source, the use of alternative donors, post-­transplant strate- Response rates were disappointing overall, with only 3/12
gies for preventing relapse (adoptive immunotherapy with (25%) achieving a CR and 1/12 (8%) a partial response (PR),
pre-­emptive DLI, role of maintenance, etc.) are issues that in comparison to the previously demonstrated efficacy
both the MPN and transplant community needs to address in AML with myelodysplasia-­related changes (MRC) and
through co-­operative trials. therapy-­related AML where CR rates approached 48%.67 A
total of seven patients proceeded with second-­line therapy
following treatment failure; three out of five patients treated
NOV E L T H E R A PI E S : W H AT DO W E with HMA–­venetoclax or cytarabine–­venetoclax achieved a
H AV E NOW A N D W H AT M AY BE ON CR and one proceeded to allo-­HCT as did another patient
T H E HOR I Z ON ? post-­FLAG-­Ida. Further work is needed to understand if this
agent—­either alone or in combination with agents such as
Below, we discuss some of the currently available agents and venetoclax—­has a role in the management of MPN AP/BP,
also discuss potential future strategies. How best to navi- as current data remains disappointing.
gate the correct sequencing and combination approaches
with these agents remains an unknown; however, we have
attempted to highlight those of particular interest and where IDH1/IDH2 inhibitors
combination approaches may be more rational.
Compared to chronic phase MPN, the incidence of IDH1/
IDH2 mutations is much higher in MPN AP/BP; one study
BCL-­2 inhibition in MPN AP and BP highlighted that the incidence is up to 22% in those with
BP MPN.68 Ivosidenib is a potent and selective inhibitor of
Venetoclax, a selective BCL-­2 inhibitor, has been trialled mutated IDH1 whereas enasidinib is a selective inhibitor of
in combination with both HMA and IC regimens. The IDH2; both have shown activity in relapsed/refractory AML
combination of venetoclax and HMA was retrospectively but initial trials contained only a few MPN BP patients.
evaluated in 35 patients from a single centre with high-­risk Chifotides and colleagues published a study on the efficacy
myeloid disease: either MDS/MPN in accelerated or blastic of ivosidenib and enasidenib in 12 MPN BP patients either
phase, MPN AP/BP or AML with extramedullary disease. as monotherapy or in combination with HMA, venetoclax,
For the entire cohort, the composite CR rate was 42.9% with ruxolitinib or IC.69 This was a heterogeneous cohort with a
a median OS of 9.7 months. There was a suggestion that median age of 67 where seven patients harboured an IDH1
those with advanced MDS/MPN had higher CR rates and mutation (R132C [n = 6] and R132H [n = 1]) and five patients
an improved OS compared to those with advanced MPN but the IDH2 R140Q mutation; a majority (10/12) had JAK2
the numbers were too small for robust conclusions.64 In the V617F. Three patients achieved CR (enasidinib/ruxolitinib/
meta-­analysis by Chen and colleagues, HMA and venetoclax azacytidine [n = 1]; ivosidenib + venetoclax as a bridge to
yielded higher CR rates than HMA +/− ruxolitinib with a allo-­HCT [n = 1] and a transient CR on ivosideninib and IC
trend to improved survival. Further data on this combina- for 7 months [n = 1]) with a median OS of 19 months. Several
tion are required to guide future combination approaches. other patients achieved significant reductions in blasts al-
Of interest, venetoclax has been trialled in combination with beit did not achieve a CR. In addition, Patel et al. reported
low-­dose navitoclax, a BCL-­X L and BCL-­2 inhibitor. Most on eight patients with IDH2-­mutated MPN BP treated with
data have emerged in the heavily treated, refractory acute enasidenib—­six were initial treatments and for two it was
lymphoblastic leukaemia setting, with 60% of patients in one for management in the relapsed/refractory setting.70 ORR
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAHDI et al.    | 9

was 75% when MPN B ­ P criteria were used for response from the phase II MANIFEST study, evaluating the efficacy
and median OS was not reached with a median follow-­up of the BETi pelabresib for chronic phase MF patients. The
of 272 days. A phase II trial of 50–­100 mg of enasidenib and combination of ruxolitinib and pelabresib was well tolerated
ruxolitinib is currently open recruiting IDH2-­mutated MPN in JAK inhibitor naive patients and demonstrated improve-
AP/BP patients (NCT04281498). Further data on the opti- ments in symptom and spleen burden, alongside potential
mal sequencing and combination approaches with IDH1/ anaemia benefits.78 No currently recruiting trials are using
IDH2-­targeted therapies are needed. pelabresib in combination with either JAKi or HMA or
other agents in MPN AP/BP and, in our opinion, such com-
bination trials would be of high interest and should be pri-
Murine double minute 2 (MDM2) inhibitors oritised. Of interest, Saenz and colleagues showed that the
BETi JQ1 inhibited growth and induced apoptosis in MPN
Wang and colleagues initially demonstrated that MPN BP derived blast cells. Moreover, synergistic activity was shown
CD34+ cells contain higher levels of MDM2 than their nor- with both ruxolitinib and heat shock protein 90 inhibitors,
mal counterparts; MDM2 is a negative regulator of TP53 and highlighting potential future strategies for MPN AP/BP.79
hence MDM2 inhibitors, such as navtemadlin (KRT-­232),
can restore normal p53 functionality and induce apoptosis in
TP53 wild-­t ype (TP53WT) cells.71 The efficacy of navtemad- I M M U NOT H E R A PY A N D CE L LU L A R
lin has been shown in PDX MPN ­BP murine models.71 Here, T H E R A PY A PPROACH E S
monotherapy with navtemadlin depleted MPN BP blast cells
and additionally prolonged survival in these PDX mice. Vaccination strategies in chronic phase MPN and MPN AP/
Next, MPN ­BP PDX mice were treated with multiple cycles of BP remain in their relative infancy but with growing inter-
either low-­dose or high-­dose navtemadlin, at 3-­weekly inter- est among the community. Initial strategies in the chronic
vals. MPN BP stem cells and leukaemic blasts were reduced phase setting included a phase I clinical trial of the safety
in the spleen and PB but not the marrow, and there was an as- and efficacy of a ‘CALRLong36 peptide vaccine’, which in-
sociated survival improvement. Additional evaluation in this cluded 10 CALR mutated (CALR MUT) chronic phase MPN
PDX model suggested better efficacy of navtemadlin than patients, receiving a total of 15 vaccines over a 12-­month
decitabine. A phase 1b/2 trial (NCT04113616) is currently period.80 The majority demonstrated evidence of a vaccine
evaluating the safety and efficacy of navtemadlin in TP53WT induced response, which increased in ET patients over the
relapsed/ refractory MPN BP.72 study period but, in contrast, not after the third vaccine dose
in MF. However, despite such responses, no molecular or
clinical responses were identified demonstrating the signifi-
Telomerase inhibitors cant challenges of such approaches.
Gigoux and colleagues next evaluated the potential of
Imetelstat is a 13-­mer oligonucleotide, which displays in- heteroclitic peptide-­based vaccines for CALR MUT MPN.81
hibitory activity against telomerase. Clinically beneficial T cells recognise non-­self-­antigens via interaction between
responses have been highlighted to date in lower risk MDS the T-­cell receptor (TCR) and the peptide–­MHC-­I com-
and predominantly chronic phase MF.73,74 Interestingly, Ma plex (pMHC). Heteroclitic peptide modifications aim to
et al. have suggested that targeting telomerase activity with increase immunogenicity. This group had recognised that
imetelstat can in fact prevent survival and self-­renewal of in CALR MUT MPN there was an underrepresentation of
both preleukaemic stem cell (LSC) and LSC in MPN models, MHC-­I alleles that present CALR MUT neoepitopes with high
where both adenosine deaminase acting on RNA (ADAR1) affinity and hence appraised that heteroclitic CALR MUT
and hTERT activation appear pivotal.75 A novel Phase II trial peptides specifically designed for the MHC-­ I alleles of
(NCT05583552) investigating the efficacy and safety of im- CALR-­mutated MPN patients could elicit a CALR MUT cross-­
etelstat in patients with high risk MDS or AML who have reactive CD8+ T-­cell response. Their initial data emphasised
failed HMA therapy is ongoing. Given the findings to date, the potential therapeutic benefit of this approach but more
future trials focused on activity of this agent in MPN-­BP work is required to see if it is applicable to the MPN AP/BP
would be of interest. setting for transformed CALR MPN.
Arginase-­1 (ARG1)-­and programmed cell death ligand 1
(PD-­L1)-­derived peptide vaccines have demonstrated safety
BET inhibitors in a first in man phase I–­II study performed in nine JAK2-­
mutated chronic phase MPN patients but were insufficient
The BRD subfamily of bromodomain and extra-­terminal by themselves to generate clinical response.82 Both ARG1
(BET) proteins bind to acetylated chromatin and are in- and PD-­L1 can inhibit T-­cell activation and proliferation in
volved in gene transcription regulation.76 BET inhibitors MPN. Handlos Grausland and colleagues hypothesized that
(BETi) have entered the clinical arena across a number of by augmenting ARG1-­and PD-­L1-­specific T-­cell responses
haematological malignancies including AML, MPN and via peptide vaccines this in turn would lead to a reduction
lymphoma.77 In MF, strong clinical responses have emerged in immunosuppressive cells in MPN and hence result in
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10 |    UPDATES ON ACCELERATED AND BLAST PHASE MYELOPROLIFERATIVE NEOPLASMS

more MPN directed T-­cell responses. Future strategies may to help us personalise and optimise sequencing and com-
include combination of such peptide vaccines with the PD-­1 binatorial therapy. We additionally predict that, given the
checkpoint inhibitor, pembrolizumab. Although a previous aggressiveness of these disease states, MPN AP and BP re-
study with this agent alone in chronic phase MPN failed to main a prime group to explore novel immunotherapeutic
generate clinical benefit, improved T-­cell responses directed approaches including peptide vaccination and ultimately
at the MPN clone were observed.83 Novel studies are open- BiTEs and CART cells. Within the transplant-­eligible popu-
ing with vaccines targeting both JAK2 and CALR and results lation, we still need to optimise conditioning approaches and
are eagerly awaited alongside translational studies to under- prospectively evaluate optimisation of pretransplant ther-
stand how clinical efficacy may be enhanced. As these de- apy, depth of response and maintenance approaches in the
velop in the chronic phase MPN setting, we aspire for rapid post allo-­HCT setting to decrease the risk of relapse. Lastly,
transition to the MPN AP/BP cohorts. there remains an urgent unmet need of dynamic predictive
Effective and non-­ toxic chimeric antigen receptor T tools for transformation events, particularly in ET and PV.
(CART) or bispecific T-­ cell engagers (BiTE) remain the We look forward to further international collaboration to
holy grail in the management of MPN AP/BP but develop- answer such questions and ultimately improve patient prog-
ment is challenged by the lack of specific, truly universally nostication and outcomes.
expressed MPN blast cell antigens. CALR MUT, as discussed
above, is an appealing immunotherapeutic target, resulting AU T HOR C ON T R I BU T ION S
from a frameshift mutation that leads to a novel C Terminus All authors contributed equally to this work and reviewed
which is absent in other cells. Additionally, CALR MUT is ex- the final version.
pressed on the cell surface by binding to the thrombopoietin
(TPO) receptor.84 Antibody strategies have been developed AC K N O​W L E​D G E​M E N T S
and showed very promising results in preclinical studies.84 All authors contributed equally to this paper, critically re-
A ‘silent-­Fc’ antibody to block the CALR-­MPL interaction viewed and revised the paper and approved the final paper
has also been developed and will enter a phase I clinical trial, prior to submission.
initially in chronic phase disease. CART cell therapy or BiTE
approaches are also feasible and under evaluation in the pre- F U N DI N G I N F OR M AT ION
clinical setting. There is no funding associated with this work.
Targeting of CD33 or CD34 alone would likely lead to
multiple ‘off target’ effects on normal haematopoietic pro- C ON F L IC T OF I N T E R E S T S TAT E M E N T
genitors. Many approaches are being taken in the AML field None of the authors have a conflict of interest to declare.
to address these challenges and a host of early phase CART
trials are open globally, predominantly focused on differing ORC I D
targets in relapsed/refractory AML. These include CD33, Nicola Polverelli https://orcid.org/0000-0001-6297-9697
CD123, CLL-­ 1, CD70, TIM-­ 3 and CD93.85 Interestingly, Donal P. McLornan https://orcid.
Ramos et al. have recently reported the use of mass spec- org/0000-0003-1224-091X
trometry to evaluate surface protein expression—­t he ‘surfa-
ceome’—­in MPN BP cell lines compared to other AML cell TWITTER
lines.86 They reported over 70 membrane proteins enriched Donal P. McLornan @DrLornan
in MPN BP-­derived cell lines and identified specific poten-
tial targets such as the G protein receptor C3AR1, which is R EFER ENCES
enriched in MPN BP and not expressed in the endothelium, 1. Tefferi A, Mudireddy M, Mannelli F, Begna KH, Patnaik MM,
granulocytic or monocytic cells or donor-­derived normal Hanson CA, et al. Blast phase myeloproliferative neoplasm: Mayo-­
AGIMM study of 410 patients from two separate cohorts. Leukemia.
CD34+ cells, and CALR. Preclinical studies using immuno-
2018;32(5):1200–­10.
therapeutic strategies to target these antigens, and others, 2. Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, et al.
are ongoing and ideally will lead to rapid adoption in the The 5th edition of the World Health Organization Classification of
clinical trial arena. Haematolymphoid Tumours: myeloid and histiocytic/dendritic neo-
plasms. Leukemia. 2022;36(7):1703–­19.
3. Masarova L, Bose P, Pemmaraju N, Daver NG, Zhou L, Pierce S, et al.
Prognostic value of blasts in peripheral blood in myelofibrosis in the
C ONC LUSIONS ruxolitinib era. Cancer. 2020;126(19):4322–­31.
4. Masarova L, Bose P, Zahr AA, Cortes J, Kantarjian H, Verstovsek
MPN AP and BP still remain challenging diseases, particu- S. Do we need to re-­ define accelerated phase of myelofibrosis?
larly when allo-­HCT is not an option. HMA and venetoclax Correlation between blast percentage in myelofibrosis and outcomes.
Clin Lymphoma Myeloma Leuk. 2017;17:S352.
+/− JAK inhibitors can be effective for a period of time yet
5. Shahin OA, Chifotides HT, Bose P, Masarova L, Verstovsek S.
require more clarity on dosing and optimal scheduling. Accelerated phase of myeloproliferative neoplasms. Acta Haematol.
Prospective platform style trials based on comprehensive 2021;144(5):484–­99.
drug synergy studies are required to evaluate approaches in 6. Barbui T, Thiele J, Passamonti F, Rumi E, Boveri E, Ruggeri M, et al.
MPN AP/BP paralleled with dynamic translational studies Survival and disease progression in essential thrombocythemia are
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAHDI et al.    | 11

significantly influenced by accurate morphologic diagnosis: an inter- 25. Lasho TL, Mudireddy M, Finke CM, Hanson CA, Ketterling RP,
national study. J Clin Oncol. 2011;29(23):3179–­84. Szuber N, et al. Targeted next-­generation sequencing in blast phase
7. Tefferi A, Rumi E, Finazzi G, Gisslinger H, Vannucchi AM, myeloproliferative neoplasms. Blood Adv. 2018;2(4):370–­80.
Rodeghiero F, et al. Survival and prognosis among 1545 patients with 26. Lasho TL, Finke CM, Hanson CA, Jimma T, Knudson RA, Ketterling
contemporary polycythemia vera: an international study. Leukemia. RP, et al. SF3B1 mutations in primary myelofibrosis: clinical, his-
2013;27(9):1874–­81. topathology and genetic correlates among 155 patients. Leukemia.
8. Tefferi A, Guglielmelli P, Larson DR, Finke C, Wassie EA, Pieri L, 2012;26(5):1135–­7.
et al. Long-­term survival and blast transformation in molecularly an- 27. Klampfl T, Harutyunyan A, Berg T, Gisslinger B, Schalling M,
notated essential thrombocythemia, polycythemia vera, and myelofi- Bagienski K, et al. Genome integrity of myeloproliferative neo-
brosis. Blood. 2014;124(16):2507–­13. plasms in chronic phase and during disease progression. Blood.
9. Iurlo A, Cattaneo D, Gianelli U. Blast transformation in myelopro- 2011;118(1):167–­76.
liferative neoplasms: risk factors, biological findings, and targeted 28. Rampal R, Ahn J, Abdel-­Wahab O, Nahas M, Wang K, Lipson D,
therapeutic options. Int J Mol Sci. 2019;20(8):E1839. et al. Genomic and functional analysis of leukemic transformation of
10. Yogarajah M, Tefferi A. Leukemic transformation in myeloprolifer- myeloproliferative neoplasms. Proc Natl Acad Sci. 2014;111(50):E540
ative neoplasms: a literature review on risk, characteristics, and out- 1–­10. https://doi.org/10.1073/pnas.14077​92111
come. Mayo Clin Proc. 2017;92(7):1118–­28. 29. Courtier F, Carbuccia N, Garnier S, Guille A, Adélaïde J, Cervera N,
11. Gangat N, Wolanskyj AP, McClure RF, Li CY, Schwager S, Wu W, et al. Genomic analysis of myeloproliferative neoplasms in chronic
et al. Risk stratification for survival and leukemic transformation in and acute phases. Haematologica. 2017;102(1):e11–­4.
essential thrombocythemia: a single institutional study of 605 pa- 30. Beer PA, Delhommeau F, LeCouédic JP, Dawson MA, Chen E,
tients. Leukemia. 2007;21(2):270–­6. Bareford D, et al. Two routes to leukemic transformation after
12. Luque Paz D, Jouanneau-­Courville R, Riou J, Ianotto JC, Boyer F, a JAK2 mutation–­ positive myeloproliferative neoplasm. Blood.
Chauveau A, et al. Leukemic evolution of polycythemia vera and es- 2010;115(14):2891–­900.
sential thrombocythemia: genomic profiles predict time to transfor- 31. Theocharides A, Boissinot M, Girodon F, Garand R, Teo SS, Lippert
mation. Blood Adv. 2020;4(19):4887–­97. E, et al. Leukemic blasts in transformed JAK2-­V617F–­positive my-
13. Tefferi A, Guglielmelli P, Lasho TL, Coltro G, Finke CM, Loscocco eloproliferative disorders are frequently negative for the JAK2-­V617F
GG, et al. Mutation-­enhanced international prognostic systems for mutation. Blood. 2007;110(1):375–­9.
essential thrombocythaemia and polycythaemia vera. Br J Haematol. 32. Lundberg P, Karow A, Nienhold R, Looser R, Hao-­Shen H, Nissen I,
2020;189:291–­302. et al. Clonal evolution and clinical correlates of somatic mutations in
14. Tefferi A, Guglielmelli P, Nicolosi M, Mannelli F, Mudireddy M, myeloproliferative neoplasms. Blood. 2014;123(14):2220–­8.
Bartalucci N, et al. GIPSS: genetically inspired prognostic scoring 33. Li B, An W, Wang H, Baslan T, Mowla S, Krishnan A, et al. BMP2/
system for primary myelofibrosis. Leukemia. 2018;32(7):1631–­42. SMAD pathway activation in JAK2/p53-­ mutant megakaryocyte/
15. Guglielmelli P, Lasho TL, Rotunno G, Mudireddy M, Mannarelli C, erythroid progenitors promotes leukemic transformation. Blood.
Nicolosi M, et al. MIPSS70: mutation-­enhanced international prog- 2022;139(25):3630–­46.
nostic score system for transplantation-­age patients with primary 34. Brierley C, Psaila B. A tale of two alleles: TP53 and transformation in
myelofibrosis. J Clin Oncol. 2018;36(4):310–­8. MPNs. Blood. 2022;139(25):3567–­8.
16. Tefferi A, Guglielmelli P, Lasho TL, Gangat N, Ketterling RP, 35. Rodriguez-­Meira A, Norfo R, Wen WX, Chédeville AL, Rahman
Pardanani A, et al. MIPSS70+ version 2.0: mutation and karyotype-­ H, O'Sullivan J, et al. Deciphering TP53 mutant cancer evolution
enhanced international prognostic scoring system for primary my- with single-­cell multi-­omics. Cancer Biol Ther. 2022. https://doi.
elofibrosis. J Clin Oncol. 2018;36(17):1769–­70. org/10.1101/2022.03.28.485984
17. Grinfeld J, Nangalia J, Baxter EJ, Wedge DC, Angelopoulos N, Cantrill 36. Celik H, Krug E, Zhang CR, Han W, Issa N, Koh WK, et al. A hu-
R, et al. Classification and personalized prognosis in myeloprolifera- manized animal model predicts clonal evolution and therapeutic
tive neoplasms. N Engl J Med. 2018;379(15):1416–­30. vulnerabilities in myeloproliferative neoplasms. Cancer Discov.
18. Passamonti F, Cervantes F, Vannucchi AM, Morra E, Rumi E, Pereira 2021;11(12):3126–­41.
A, et al. A dynamic prognostic model to predict survival in primary 37. Fisher DAC, Fowles JS, Zhou A, Oh ST. Inflammatory pathophysiol-
myelofibrosis: a study by the IWG-­ MRT (International Working ogy as a contributor to myeloproliferative neoplasms. Front Immunol.
Group for Myeloproliferative Neoplasms Research and Treatment). 2021;12:683401.
Blood. 2010;115(9):1703–­8. 38. Panteli KE, Hatzimichael EC, Bouranta PK, Katsaraki A, Seferiadis
19. Gangat N, Caramazza D, Vaidya R, George G, Begna K, Schwager S, et al. K, Stebbing J, et al. Serum interleukin (IL)-­1, IL-­2 , sIL-­2Ra, IL-­6 and
DIPSS plus: a refined Dynamic International Prognostic Scoring System for thrombopoietin levels in patients with chronic myeloproliferative
primary myelofibrosis that incorporates prognostic information from karyo- diseases. Br J Haematol. 2005;130(5):709–­15.
type, platelet count, and transfusion status. J Clin Oncol. 2011;29(4):392–­7. 39. Hasselbalch HC, Bjørn ME. MPNs as inflammatory diseases: the evi-
20. Mannelli F, Bencini S, Coltro G, Loscocco GG, Peruzzi B, Rotunno G, dence, consequences, and perspectives. Mediat Inflamm. 2015;2015:1–­16.
et al. Integration of multiparameter flow cytometry score improves 40. Alchalby H, Zabelina T, Stübig T, van Biezen A, Bornhäuser M,
prognostic stratification provided by standard models in primary Di Bartolomeo P, et al. Allogeneic stem cell transplantation for
myelofibrosis. Am J Hematol. 2022;97(7):846–­55. myelofibrosis with leukemic transformation: a study from the
21. Vallapureddy RR, Mudireddy M, Penna D, Lasho TL, Finke CM, Myeloproliferative Neoplasm Subcommittee of the CMWP of the
Hanson CA, et al. Leukemic transformation among 1306 patients European Group for Blood and Marrow Transplantation. Biol Blood
with primary myelofibrosis: risk factors and development of a predic- Marrow Transplant. 2014;20(2):279–­81.
tive model. Blood Cancer J. 2019;9(2):12. 41. Ortí G, Gras L, Zinger N, Finazzi MC, Sockel K, Robin M, et al.
22. Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A. Outcomes after allogeneic hematopoietic cell transplant in patients
IDH1 and IDH2 mutation analysis in chronic-­and blast-­phase my- diagnosed with blast phase of myeloproliferative neoplasms: a ret-
eloproliferative neoplasms. Leukemia. 2010;24(6):1146–­51. rospective study from the Chronic Malignancies Working Party of
23. Jäger R, Gisslinger H, Berg T, Passamonti F, Cazzola M, Rumi E, et al. the European Society for Blood and Marrow Transplantation. Am J
Deletions of the transcription factor Ikaros in myeloproliferative neoplasms Hematol. 2023;98:628–­38.
at transformation to acute myeloid leukemia. Blood. 2009;114(22):435. 42. Malagola M, Polverelli N, Rubini V, Martino M, Patriarca F, Bruno B,
24. Baumeister J, Chatain N, Sofias AM, Lammers T, Koschmieder S. et al. GITMO registry study on allogeneic transplantation in patients
Progression of myeloproliferative neoplasms (MPN): diagnostic and aged ≥60 years from 2000 to 2017: improvements and criticisms.
therapeutic perspectives. Cell. 2021;10(12):3551. Transplant Cell Ther. 2022;28(2):96.e1–­96.e11.
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
12 |    UPDATES ON ACCELERATED AND BLAST PHASE MYELOPROLIFERATIVE NEOPLASMS

43. Lin RJ, Artz AS. Allogeneic hematopoietic cell transplantation for 59. Byrne JL, Beshti H, Clark D, Ellis I, Haynes AP, Das-­Gupta E, et al.
older patients. Hematology. 2021;2021(1):254–­63. Induction of remission after donor leucocyte infusion for the treat-
44. Polverelli N, Tura P, Battipaglia G, Malagola M, Bernardi S, Gandolfi ment of relapsed chronic idiopathic myelofibrosis following alloge-
L, et al. Multidimensional geriatric assessment for elderly hematolog- neic transplantation: evidence for a ‘graft vs. myelofibrosis’ effect. Br
ical patients (≥60 years) submitted to allogeneic stem cell transplan- J Haematol. 2000;108(2):430–­3.
tation. A French–­Italian 10-­year experience on 228 patients. Bone 60. McLornan DP, Hernandez-­Boluda JC, Czerw T, Cross N, Joachim
Marrow Transplant. 2020;55(12):2224–­33. Deeg H, Ditschkowski M, et al. Allogeneic haematopoietic cell trans-
45. Issa JPJ, Kantarjian HM. Targeting DNA methylation. Clin Cancer plantation for myelofibrosis: proposed definitions and management
Res. 2009;15(12):3938–­46. strategies for graft failure, poor graft function and relapse: best prac-
46. Wang JC, Chen W, Nallusamy S, Chen C, Novetsky AD. tice recommendations of the EBMT Chronic Malignancies Working
Hypermethylation of the P15INK4b and P16INK4a in agnogenic myeloid Party. Leukemia. 2021;35(9):2445–­59.
metaplasia (AMM) and AMM in leukaemic transformation: hyper- 61. Gupta V, Kim S, Hu ZH, Liu Y, Aljurf M, Bacher U, et al. Comparison
methylation of p15, p16 in Leukaemic transformation of MF. Br J of outcomes of HCT in blast phase of BCR-­ABL1−MPN with de novo
Haematol. 2002;116(3):582– ­6. AML and with AML following MDS. Blood Adv. 2020;4(19):4748–­57.
47. Thepot S, Itzykson R, Seegers V, Raffoux E, Quesnel B, Chait Y, 62. Versluis J, Lindsley RC. Transplant for TP53-­ mutated MDS
et al. Treatment of progression of Philadelphia-­ negative myelop- and AML: because we can or because we should? Hematology.
roliferative neoplasms to myelodysplastic syndrome or acute my- 2022;2022(1):522–­7.
eloid leukemia by azacitidine: a report on 54 cases on the behalf 63. Kim K, Maiti A, Loghavi S, Pourebrahim R, Kadia TM, Rausch CR,
of the Groupe Francophone des Myelodysplasies (GFM). Blood. et al. Outcomes of TP53 -­mutant acute myeloid leukemia with decit-
2010;116(19):3735–­42. abine and venetoclax. Cancer. 2021;127(20):3772–­81.
48. Badar T, Kantarjian HM, Ravandi F, Jabbour E, Borthakur G, Cortes 64. Sanber K, Ye K, Tsai HL, Newman M, Ambinder A, DeZern A,
JE, et al. Therapeutic benefit of decitabine, a hypomethylating agent, et al. P572: venetoclax and hypomethylating agent combinations
in patients with high-­risk primary myelofibrosis and myeloprolifera- for the treatment of advanced myeloproliferative neoplasms and
tive neoplasm in accelerated or blastic/acute myeloid leukemia phase. acute myeloid leukemia with extramedullary disease. HemaSphere.
Leuk Res. 2015;39(9):950–­6. 2022;6:471–­2 .
49. Andriani A, Montanaro M, Voso MT, Villivà N, Ciccone F, Andrizzi 65. Pullarkat VA, Lacayo NJ, Jabbour E, Rubnitz JE, Bajel A, Laetsch TW,
C, et al. Azacytidine for the treatment of retrospective analysis from et al. Venetoclax and navitoclax in combination with chemotherapy
the Gruppo Laziale for the study of Ph-­negative MPN. Leuk Res. in patients with relapsed or refractory acute lymphoblastic leukemia
2015;39(8):801–­4. and lymphoblastic lymphoma. Cancer Discov. 2021;11(6):1440–­53.
50. Chen J, Wang K, Xiao Z, Xu Z. Efficacy and safety of combination 66. Ilyas R, McCullough K, Badar T, Patnaik MMM, Alkhateeb HB,
therapies vs monotherapy of hypomethylating agents in acceler- Mangaonkar AA, et al. CPX-­351 for blast-­phase myeloproliferative
ated or blast phase of Philadelphia negative myeloproliferative neoplasm (MPN-­BP): Mayo Clinic experience in 10 consecutive pa-
neoplasms: a systematic review and meta-­ a nalysis. Ann Med. tients. Blood. 2022;140(Suppl 1):9704–­5.
2023;55(1):348– ­6 0. 67. Lancet JE, Uy GL, Cortes JE, Newell LF, Lin TL, Ritchie EK, et al.
51. Mwirigi A, Galli S, Keohane C, Raj K, Radia DH, Harrison CN, et al. CPX-­351 (cytarabine and daunorubicin) liposome for injection ver-
Combination therapy with ruxolitinib plus 5-­azacytidine or continu- sus conventional cytarabine plus daunorubicin in older patients with
ous infusion of low dose cytarabine is feasible in patients with blast-­ newly diagnosed secondary acute myeloid leukemia. J Clin Oncol.
phase myeloproliferative neoplasms. Br J Haematol. 2014;167(5):714–­6. 2018;36(26):2684–­92.
52. Mascarenhas JO, Rampal RK, Kosiorek HE, Bhave R, Hexner E, Wang 68. Tefferi A, Lasho TL, Abdel-­ Wahab O, Guglielmelli P, Patel J,
ES, et al. Phase 2 study of ruxolitinib and decitabine in patients with Caramazza D, et al. IDH1 and IDH2 mutation studies in 1473 patients
myeloproliferative neoplasm in accelerated and blast phase. Blood with chronic-­, fibrotic-­or blast-­phase essential thrombocythemia,
Adv. 2020;4(20):5246–­56. polycythemia vera or myelofibrosis. Leukemia. 2010;24(7):1302–­9.
53. Bose P, Verstovsek S, Cortes JE, Tse S, Gasior Y, Jain N, et al. A 69. Chifotides HT, Masarova L, Alfayez M, Daver N, Alvarado Y,
phase 1/2 study of ruxolitinib and decitabine in patients with post-­ Jabbour E, et al. Outcome of patients with IDH1/2-­mutated post–­
myeloproliferative neoplasm acute myeloid leukemia. Leukemia. myeloproliferative neoplasm AML in the era of IDH inhibitors. Blood
2020;34(9):2489–­92. Adv. 2020;4(21):5336–­42.
54. Drummond MW, Gaskell C, Harrison C, Mead AJ, Yap C, Jackson 70. Patel AA, Cahill K, Charnot-­ Katsikas A, Liu H, Gurbuxani S,
AE, et al. Phazar: a phase Ib study to assess the safety and tolerabil- Thirman M, et al. Clinical outcomes of IDH2 -­mutated advanced-­
ity of ruxolitinib in combination with azacitidine in advanced phase phase Ph-­negative myeloproliferative neoplasms treated with enas-
myeloproliferative neoplasms (MPN), including myelodysplastic syn- idenib. Br J Haematol. 2020;190(1):e48–­e51. https://doi.org/10.1111/
dromes (MDS) or acute myeloid leukaemia (AML) arising from MPN bjh.16709
[ISRCTN16783472]. Blood. 2020;136(Suppl 1):2–­3. 71. Wang X, Hu CS, Gillespie V, Krejsa CM, Hoffman R. Navtemadlin
55. Kennedy JA, Atenafu EG, Messner HA, Craddock KJ, Brandwein JM, (KRT-­232), a small molecule MDM2 inhibitor, is more effective than
Lipton JH, et al. Treatment outcomes following leukemic transforma- decitabine against myeloproliferative neoplasm-­ blast phase in a
tion in Philadelphia-­negative myeloproliferative neoplasms. Blood. patient-­derived Xenograft model. Blood. 2021;138(Suppl 1):3591.
2013;121(14):2725–­33. 72. Rampal R, Ramanathan S, Papayannidis C, von Bubnoff N, del Mar
56. Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA, Tefferi A. Tormo Díaz M, Vidriales MB, et al. An open-­label, multicenter, phase
Leukemic transformation in myelofibrosis with myeloid metaplasia: a 1b/2 study of navtemadlin (KRT-­232) in patients with relapsed/re-
single-­institution experience with 91 cases. Blood. 2005;105(3):973–­7. fractory acute myeloid leukemia secondary to myeloproliferative
57. Tam CS, Nussenzveig RM, Popat U, Bueso-­ Ramos CE, Thomas neoplasms. J Clin Oncol. 2022;40(16_suppl):TPS7063.
DA, Cortes JA, et al. The natural history and treatment outcome 73. Platzbecker U, Komrokji RS, Fenaux P, Zeidan AM, Sekeres MA,
of blast phase BCR-­ A BL− myeloproliferative neoplasms. Blood. Savona MR, et al. Imetelstat achieved prolonged, continuous trans-
2008;112(5):1628–­37. fusion independence (TI) in patients with heavily transfused non-­
58. Baron F, Labopin M, Savani BN, Beohou E, Niederwieser D, Eder M, Del(5q) lower-­risk myelodysplastic syndrome (LR-­MDS) relapsed/
et al. Graft-­versus-­host disease and graft-­versus-­leukaemia effects in refractory (R/R) to erythropoiesis stimulating agents (ESAs) within
secondary acute myeloid leukaemia: a retrospective, multicentre reg- the IMerge phase 2 study. Blood. 2022;140(Suppl 1):1106–­8.
istry analysis from the Acute Leukaemia Working Party of the EBMT. 74. Kuykendall A, Wan Y, Mascarenhas J, Kiladjian JJ, Vannucchi A,
Br J Haematol. 2020;188(3):428–­37. Wang J, et al. Favorable overall survival of imetelstat-­t reated relapsed/
13652141, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.19010 by Christian Medical College, Wiley Online Library on [07/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAHDI et al.    | 13

refractory myelofibrosis patients compared with closely matched real 85. Vishwasrao P, Li G, Boucher JC, Smith DL, Hui SK. Emerging CAR T
world data. Ann Hematol. 2022;101:139–­46. cell strategies for the treatment of AML. Cancer. 2022;14(5):1241.
75. Ma W, Balaian L, Mondala P, He Y, Mason C, Pham J, et al. Imetelstat 86. Ramos E, Wiita A. Profiling the cell surface of MPN blast phase sug-
inhibits telomerase and prevents propagation of ADAR1-­activated gests unique biology and therapeutic targets. Blood. 2022;140(Suppl 1):​
myeloproliferative neoplasm and leukemia stem cells. Blood. 6735–­6.
2020;136(Suppl 1):18. 87. Shah MV, Saliba RM, Varma A, Ciurea SO, Oran B, Olson A, et al.
76. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov Allogeneic stem cell transplant for patients with myeloproliferative
O, et al. Selective inhibition of BET bromodomains. Nature. neoplasms in blast phase: improving outcomes in the recent era. Br J
2010;468(7327):1067–­73. Haematol. 2021;193(5):1004–­8.
77. Abedin SM, Boddy CS, Munshi HG. BET inhibitors in the treatment 88. Kröger N, Eikema D, Köster L, Beelen D, de Wreede LC, Finke J,
of hematologic malignancies: current insights and future prospects. et al. Impact of primary disease on outcome after allogeneic stem
Onco Targets Ther. 2016;9:5943–­53. cell transplantation for transformed secondary acute leukaemia. Br J
78. Mascarenhas J, Kremyanskaya M, Patriarca A, Palandri F, Devos T, Haematol. 2019;185(4):725–­32.
Passamonti F, et al. MANIFEST: pelabresib in combination with rux- 89. Takagi S, Masuoka K, Uchida N, Kurokawa M, Nakamae H, Imada
olitinib for Janus Kinase Inhibitor Treatment-­Naïve Myelofibrosis. J K, et al. Allogeneic hematopoietic cell transplantation for leukemic
Clin Oncol. 2023; JCO2201972. https://doi.org/10.1200/JCO.22.01972 transformation preceded by Philadelphia chromosome–­ negative
79. Saenz DT, Fiskus W, Manshouri T, Rajapakshe K, Krieger S, Sun B, myeloproliferative neoplasms: a nationwide survey by the Adult
et al. BET protein bromodomain inhibitor-­based combinations are Acute Myeloid Leukemia Working Group of the Japan Society for
highly active against post-­ myeloproliferative neoplasm secondary Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant.
AML cells. Leukemia. 2017;31(3):678–­87. 2016;22(12):2208–­13.
80. Handlos Grauslund J, Holmström MO, Jørgensen NG, Klausen U, 90. Cahu X, Chevallier P, Clavert A, Suarez F, Michallet M, Vincent L,
Weis-­Banke SE, El Fassi D, et al. Therapeutic cancer vaccination with et al. Allo-­SCT for Philadelphia-­ negative myeloproliferative neo-
a peptide derived from the calreticulin exon 9 mutations induces plasms in blast phase: a study from the Societe Française de Greffe
strong cellular immune responses in patients with CALR-­mutant de Moelle et de Therapie Cellulaire (SFGM-­ TC). Bone Marrow
chronic myeloproliferative neoplasms. Front Oncol. 2021;11:637420. Transplant. 2014;49(6):756–­60.
81. Gigoux M, Holmström MO, Zappasodi R, Park JJ, Pourpe S, Bozkus 91. Ciurea SO, de Lima M, Giralt S, Saliba R, Bueso-­Ramos C, Andersson
CC, et al. Calreticulin mutant myeloproliferative neoplasms induce BS, et al. Allogeneic stem cell transplantation for myelofibrosis
MHC-­I skewing, which can be overcome by an optimized peptide with leukemic transformation. Biol Blood Marrow Transplant.
cancer vaccine. Sci Transl Med. 2022;14(649):eaba4380. 2010;16(4):555–­9.
82. Grauslund JH, Holmström MO, Martinenaite E, Lisle TL, Glöckner
HJ, El Fassi D, et al. An arginase1-­and PD-­L1-­derived peptide-­based
vaccine for myeloproliferative neoplasms: a first-­in-­man clinical trial.
Front Immunol. 2023;14:1117466.
How to cite this article: Mahdi D, Spiers J, Rampotas
83. Hobbs G, Cimen Bozkus C, Moshier E, Dougherty M, Bar-­Natan M,
Sandy L, et al. PD-­1 inhibition in advanced myeloproliferative neo- A, Polverelli N, McLornan DP. Updates on accelerated
plasms. Blood Adv. 2021;5(23):5086–­97. and blast phase myeloproliferative neoplasms: Are we
84. Reis E, Buonpane R, Celik H, Marty C, Lei A, Jobe F, et al. Discovery making progress? Br J Haematol. 2023;00:1–13.
of INCA033989, a monoclonal antibody that selectively antagonizes https://doi.org/10.1111/bjh.19010
mutant calreticulin oncogenic function in myeloproliferative neo-
plasms (MPNs). Blood. 2022;140(Suppl 1):14–­5.

You might also like