Você está na página 1de 11

CME ARTICLE

A JH
CME Information: Primary myelofibrosis: 2017 update on
diagnosis, risk-stratification and management

CME Editor: Ayalew Tefferi, M.D.


Author: Ayalew Tefferi, M.D.

If you wish to receive credit for this activity, please refer to the website: www.wileyhealthlearning.com

䊏 Accreditation and Designation Statement:


Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical
education for physicians.
Blackwell Futura Media Services designates this journal-based CME for a maximum of 1 AMA PRA Category 1 CreditTM. Physicians should
only claim credit commensurate with the extent of their participation in the activity.

䊏 Educational Objectives
Upon completion of this educational activity, participants will be better able to:

 Highlight the 2016 revision on the classification and diagnosis of primary myelofibrosis, both overt and prefibrotic.
 Discuss contemporary risk stratification and the role of molecular prognostication.
 Provide an overview on risk-adapted treatment strategies.

䊏 Activity Disclosures
No commercial support has been accepted related to the development or publication of this activity.
Author and CME Editor: Ayalew Tefferi, MD has no relevant financial relationships to disclose.
This activity underwent peer review in line with the standards of editorial integrity and publication ethics maintained by American Journal of
Hematology. The peer reviewers have no conflicts of interest to disclose. The peer review process for American Journal of Hematology is single
blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review.
Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services’ Policy on Activity Disclosure and
Conflict of Interest. The primary resolution method used was peer review and review by a non-conflicted expert.

䊏 Instructions on Receiving Credit


This activity is intended for physicians. For information on applicability and acceptance of continuing medical education credit for this activity,
please consult your professional licensing board.
This activity is designed to be completed within one hour; physicians should claim only those credits that reflect the time actually spent in the
activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial
publication. Additionally, up to 3 attempts and a score of 70% or better is needed to pass the post test.
Follow these steps to earn credit:

 Log on to www.wileyhealthlearning.com
 Read the target audience, educational objectives, and activity disclosures.
 Read the activity contents in print or online format.
 Reflect on the activity contents.
 Access the CME Exam, and choose the best answer to each question.
 Complete the required evaluation component of the activity.
 Claim your Certificate.

This activity will be available for CME credit for twelve months following its launch date. At that time, it will be reviewed and potentially
updated and extended for an additional twelve months.

C 2016 Wiley Periodicals, Inc.


V

American Journal of Hematology, Vol. 91, No. 12, December 2016 1261
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES
AJH Educational Material A JH
Primary myelofibrosis: 2017 update on diagnosis,
risk-stratification, and management
Ayalew Tefferi*

Disease overview: Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by


stem cell-derived clonal myeloproliferation that is often but not always accompanied by JAK2, CALR or MPL
mutation, abnormal cytokine expression, bone marrow fibrosis, anemia, splenomegaly, extramedullary
hematopoiesis (EMH), constitutional symptoms, cachexia, leukemic progression and shortened survival.
Diagnosis: Diagnosis is based on bone marrow morphology. The presence of JAK2, CALR or MPL mutation
is supportive but not essential for diagnosis; approximately 90% of patients carry one of these mutations
and 10% are “triple-negative.” None of these mutations are specific to PMF and are also seen in essential
thrombocythemia (ET). According to the revised 2016 World Health Organization (WHO) classification and
diagnostic criteria, “prefibrotic” PMF (pre-PMF) is distinguished from “overtly fibrotic” PMF; the former might
mimic ET in its presentation and it is prognostically relevant to distinguish the two.
Risk stratification: The Dynamic International Prognostic Scoring System-plus (DIPSS-plus) uses eight
predictors of inferior survival: age >65 years, hemoglobin <10 g/dL, leukocytes >25 3 109/L, circulating
blasts 1%, constitutional symptoms, red cell transfusion dependency, platelet count <100 3 109/L and
unfavorable karyotype (i.e., complex karyotype or sole or two abnormalities that include 18, 27/7q-, i(17q),
inv(3), 5/5q-, 12p-, or 11q23 rearrangement). The presence of 0, 1, “2 or 3” and 4 adverse factors defines
low, intermediate-1, intermediate-2 and high-risk disease with median survivals of approximately 15.4, 6.5, 2.9
and 1.3 years, respectively. Most recently, DIPSS-plus-independent adverse prognostic relevance has been
demonstrated for certain mutations including ASXL1 and SRSF2 whereas patients with type 1/like CALR
mutations, compared to their counterparts with other driver mutations, displayed significantly better survival.
Risk-adapted therapy: Observation alone is a reasonable treatment strategy for asymptomatic low or
intermediate-1 DIPSS-plus risk disease, especially in the absence of high-risk mutations. All other patients
with high or intermediate-2 risk disease, or those harboring high-risk mutations such as ASXL1 or SRSF2,
should be considered for stem cell transplant, which is currently the only treatment modality with the
potential to favorably modify the natural history of the disease. Non-transplant candidates should be
encouraged to participate in clinical trials, since the value of conventional drug therapy, including the use of
JAK2 inhibitors, is limited to symptoms palliation and reduction in spleen size. Specifically, JAK2 inhibitors
have not been shown to induce complete clinical or cytogenetic remissions or significantly affect JAK2/
CALR/MPL mutant allele burden. Splenectomy is considered for drug-refractory splenomegaly. Involved field
radiotherapy is most useful for post-splenectomy hepatomegaly, non-hepatosplenic EMH, PMF-associated
pulmonary hypertension and extremity bone pain.
Am. J. Hematol. 91:1262–1271, 2016. VC 2016 Wiley Periodicals, Inc.

䊏 Disease Overview
The World Health Organization (WHO) classification system for hematopoietic tumors was recently revised and the 2016 document recognizes
several major categories of myeloid malignancies including acute myeloid leukemia (AML) and related neoplasms, myelodysplastic syndromes
(MDS), myeloproliferative neoplasms (MPN), MDS/MPN overlap, mastocytosis, eosinophilia-associated myeloid/lymphoid neoplasms with specific
mutations (e.g., PDGFR) and myeloid neoplasms with germline predisposition (Fig. 1).[1]
“BCR-ABL1-negative MPN” is an operational sub-category of MPN that includes polycythemia vera (PV), essential thrombocythemia (ET) and
primary myelofibrosis (PMF).[2] The 2016 WHO classification system distinguishes prefibrotic (prePMF) from overtly fibrotic PMF (Figs. 1 and
2).[1,2] PMF, PV and ET are all characterized by stem cell-derived clonal myeloproliferation and presence of somatic mutations that are opera-
tionally classified into “driver” and “other” mutations; the former include JAK2, CALR and MPL and the latter LNK, CBL, TET2, ASXL1, IDH,
IKZF1, EZH2, DNMT3A, TP53, SF3B1, SRSF2, U2AF1 or other mutations (Table I) [3–5]. It is generally believed that driver mutations are essen-
tial for the MPN phenotype whereas the “other” mutations might contribute to disease progression and leukemic transformation.

Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota


*Correspondence to: Ayalew Tefferi, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905. E-mail: tefferi.
ayalew@mayo.edu
Received for publication: 19 October 2016; Accepted: 20 October 2016
Am. J. Hematol. 91:1262–1271, 2016.
Published online: in Wiley Online Library (wileyonlinelibrary.com).
DOI: 10.1002/ajh.24592

C 2016 Wiley Periodicals, Inc.


V

1262 American Journal of Hematology, Vol. 91, No. 12, December 2016 doi:10.1002/ajh.24592
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES Primary myelofibrosis

Figure 1. 2016 World Health Organization (WHO) classification of myeloid malignancies. [Color figure can be viewed at wileyonlinelibrary.com]

In PMF, clonal myeloproliferation is associated with reactive bone The presence of these genetic markers, therefore, strongly supports a
marrow fibrosis, osteosclerosis, angiogenesis, extramedullary hemato- diagnosis of PMF, in the presence of a myeloid neoplasm associated
poiesis (EMH) and abnormal cytokine expression. Clinical manifesta- with bone marrow fibrosis.
tions in PMF include severe anemia, marked hepatosplenomegaly, PMF should be distinguished from other closely related myeloid
constitutional symptoms (e.g., fatigue, night sweats, fever), cachexia, neoplasms including chronic myeloid leukemia (CML), PV, ET,
bone pain, splenic infarct, pruritus, thrombosis and bleeding [6]. Inef- MDS, chronic myelomonocytic leukemia (CMML) and “acute
fective erythropoiesis and EMH are the main causes of anemia and myelofibrosis.” The presence of dwarf megakaryocytes raises the pos-
organomegaly, respectively. Other disease complications include sibility of CML and should be pursued with BCR-ABL1 cytogenetic
symptomatic portal hypertension that might lead to variceal bleeding or molecular testing. Patients who otherwise fulfill the diagnostic cri-
or ascites and non-hepatosplenic EMH that might lead to cord com- teria for PV should be labeled as “PV” even if they display substantial
pression, ascites, pleural effusion, pulmonary hypertension or diffuse bone marrow fibrosis [13]. Prefibrotic PMF can mimic ET in its pre-
extremity pain. It is currently assumed that aberrant cytokine produc- sentation and mutation profile (both can express JAK2, CALR or
tion by clonal cells and host immune reaction contribute to PMF- MPL mutations) [14] careful morphologic examination is necessary
associated bone marrow stromal changes, ineffective erythropoiesis, for distinguishing the two; megakaryocytes in ET are large and
EMH, cachexia and constitutional symptoms [7]. Causes of death mature-appearing whereas those in prefibrotic PMF display abnormal
include leukemic progression that occurs in approximately 20% of maturation with hyperchromatic and irregularly folded nuclei [10];
patients but many patients also die of comorbid conditions including the distinction between ET and pre-fibrotic PMF is prognostically rel-
cardiovascular events and consequences of cytopenias including infec- evant [15]. MDS should be suspected in the presence of dyserythro-
tion or bleeding [8]. poiesis or dysgranulopoiesis [16]. CMML is a possibility in the
presence of peripheral blood monocyte count of greater than 1 3
䊏 Diagnosis 109/L [17]. Patients with acute myelofibrosis (either acute panmyelo-
sis with myelofibrosis or acute megakaryoblastic leukemia) usually
Current diagnosis of PMF is based on the 2016 WHO-criteria and present with severe constitutional symptoms, pancytopenia, mild or
involves a composite assessment of clinical and laboratory features no splenomegaly, and increased circulating blasts [18].
(Fig. 2) [2]. The diagnosis of post-PV or post-ET MF should adhere
to criteria published by the International Working Group for MPN
Research and Treatment (IWG-MRT) (Table II) [9]. Peripheral blood 䊏 Risk Stratification
leukoerythroblastosis (i.e., presence of nucleated red cells, immature Robust prognostic modeling in PMF started with the development
granulocytes and dacryocytes) is a typical but not invariable feature of the International Prognostic Scoring System (IPSS) in 2009 [19].
of PMF; prefibrotic PMF might not display overt leukoerythroblasto- The IPSS for PMF is applicable to patients being evaluated at time of
sis [10]. Bone marrow fibrosis in PMF is usually associated with initial diagnosis and uses five independent predictors of inferior sur-
JAK2V617F or mutant CALR, or MPL, trisomy 9 or del(13q) [11,12]. vival: age >65 years, hemoglobin <10 g/dL, leukocyte count >25 3

doi:10.1002/ajh.24592 American Journal of Hematology, Vol. 91, No. 12, December 2016 1263
Tefferi ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES

Figure 2. 2016 Revised WHO Diagnostic Criteria for Myeloproliferative Neoplasms (From After et al Blood 2016, 127, 2391). [Color figure can be viewed at
wileyonlinelibrary.com]

109/L, circulating blasts 1% and presence of constitutional symp- a >80% two-year mortality in PMF was predicted by monosomal
toms [19]. The presence of 0, 1, 2 and 3 adverse factors defines karyotype, inv(3)/i(17q) abnormalities, or any two of circulating blasts
low, intermediate-1, intermediate-2 and high-risk disease. The corre- >9%, leukocytes 40 3 109/L or other unfavorable karyotype [27].
sponding median survivals were 11.3, 7.9, 4 and 2.3 years [19]. Similarly, inferior survival in PMF has been associated with nulli-
The IWG-MRT subsequently developed a dynamic prognostic zygosity for JAK2 46/1 haplotype [28], low JAK2V617F allele burden
model (DIPSS) that utilizes the same prognostic variables used in [29,30], or presence of IDH [31], EZH2 [32], SRSF2 [33], or ASXL1
IPSS but can be applied at any time during the disease course [20]. [34] mutations. Survival in PMF was also affected by increased serum
DIPSS assigns two, instead of one, adverse points for hemoglobin IL-8 and IL-2R levels as well as serum free light chain levels, both
<10 g/dL and risk categorization is accordingly modified: low (0 independent of DIPSS-plus [35,36].
adverse points), intermediate-1 (1 or 2 points), intermediate-2 (3 or 4 Most recently, the phenotypic and prognostic relevance of “driver”
points) and high (5 or 6 points). The corresponding median survivals and “other” mutations was carefully investigated in a series of collabo-
were not reached, 14.2, 4 and 1.5 years [20]. rative studies from the Mayo Clinic, USA and University of Florence,
IPSS- and DIPSS-independent risk factors for survival in PMF Italy. In one of these studies involving 254 patients with PMF, reported
were subsequently identified and included unfavorable karyotype (i.e., mutational frequencies were 58% for JAK2, 25% CALR, 8% MPL and
complex karyotype or sole or two abnormalities that include 18, 27/ 9% wild-type for all three mutations (i.e., triple-negative) [12]. CALR
7q-, i(17q), inv(3), 25/5q-, 12p- or 11q23 rearrangement) [21,22], mutational frequency in JAK2/MPL-unmutated cases was 74%. CALR
red cell transfusion need [23,24] and platelet count <100 3 109/L mutations were associated with younger age, higher platelet count and
[25]. Accordingly, DIPSS was modified into DIPSS-plus by incorpo- lower DIPSS-plus score. CALR-mutated patients were also less likely to
rating these three additional DIPSS-independent risk factors: platelet be anemic, require transfusions or display leukocytosis. Spliceosome
count <100 3 109/L, red cell transfusion need and unfavorable kar- mutations were infrequent in CALR-mutated patients.
yotype [26]. The four DIPSS-plus risk categories based on the afore- In terms of prognostic relevance, in a subsequent study of 570
mentioned eight risk factors are low (no risk factors), intermediate-1 patients [37], the authors reported the longest survival in CALR 1
(one risk factor), intermediate-2 (two or 3 risk factors) and high ASXL1- patients (median 10.4 years) and shortest in CALR–ASXL11
(four or more risk factors) with respective median survivals of 15.4, patients (median 2.3 years). CALR 1 ASXL11 and CALR–ASXL1-
6.5, 2.9 and 1.3 years (Fig. 3) [26]. patients had similar survival and were grouped together in an inter-
Since the publication of DIPSS-plus, several studies that suggest mediate risk category (median survival 5.8 years). Guglielmelli et al
additional prognostic information have been published. For example, subsequently demonstrated the additional value of the number of

1264 American Journal of Hematology, Vol. 91, No. 12, December 2016 doi:10.1002/ajh.24592
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES Primary myelofibrosis

TABLE I. Somatic Mutations in Primary Myelofibrosis (PMF) and the Closely Related BCR-ABL1-Negative Myeloproliferative Neoplasms (MPN) Including Polycy-
themia Vera (PV) and Essential Thrombocythemia (ET)

Chromosome Mutational
Mutations location frequency Pathogenetic relevance

JAK2 (Janus kinase 2) 9p24 PV  96% Contributes to abnormal myeloproliferation and pro-
JAK2V617F exon 14 mutation ET 55% genitor cell growth factor hypersensitivity
PMF  65%
JAK2 exon 12 mutation 9p24 PV  3% Contributes to primarily erythroid myeloproliferation
CALR (Calreticulin) 19p13.2 PMF  25% Wild-type CALR is a multi-functional Ca21 binding
Exon 9 deletions and insertions ET  20% protein chaperone mostly localized in the endo-
PV 0% plasmic reticulum
MPL (Myeloproliferative leukemia virus oncogene) 1p34 ET 3% Contributes to primarily megakaryocytic
MPN-associated MPL mutations involve exon 10 PMF  10% myeloproliferation
LNK (as in Links) a.k.a. SH2B3 12q24.12 PV  rare Wild-type LNK is a negative regulator of JAK2
(a membrane-bound adaptor protein) ET rare signaling
MPN-associated mutations were PMF  rare
monoallelic and involved exon 2 BP-MPN  10%
TET2 (TET oncogene family member 2) 4q24 PV 16% TET proteins catalyze conversion of 5-
ET5% methylcytosine (5mC) to 5-hydroxymethylcytosine
PMF 17% (5hmC), which favors demethylated DNA. Both
Mutations involve several exons BP-MPN 17% TET1 and TET2 display this catalytic activity. IDH
and TET2 mutations might share a common path-
ogenetic effect.
ASXL1 (Additional Sex Combs-Like 1) 20q11.1 ET 3% Wild-type ASXL1 is needed for normal hematopoie-
PMF 13% sis and might be involved in co-activation of tran-
Exon 12 mutations BP-MPN 18% scription factors and transcriptional repression.
IDH1/IDH2 (Isocitrate dehydrogenase) 2q33.3/ PV 2% IDH mutations induce loss of activity for the conver-
Exon 4 mutations 15q26.1 ET1% sion of isocitrate to 2-ketoglutarate (2-KG) and
PMF 4% gain of function in the conversion of 2-KG to 2-
BP-MPN 20% hydroxyglutarate (2-HG). 2-HG might be the medi-
ator of impaired TET2 function in cells with
mutant IDH expression.
EZH2 (enhancer of zeste homolog 2) 7q36.1 PV 3% Wild-type EZH2 is part of a histone methyltransfer-
Mutations involve several exons PMF 7% ase (polycomb repressive complex 2 associated
MDS 6% with H3 Lys-27 trimethylation). MPN-associated
EZH2 mutations might have a tumor suppressor
activity, which contrasts with the gain-of-function
activity for lymphoma-associated EZH2 mutations.
DNMT3A (DNA cytosine methyltransferase 3a) 2p23 PV 7% DNA methyl transferases are essential In establish-
PMF 7% ing and maintaining DNA methylation patterns in
Most frequent mutations affect amino acid R882 BP-MPN 14% mammals
CBL (Casitas B-lineage lymphoma proto-oncogene) 11q23.3 PV rare CBL is an E3 ubiquitin ligase that marks mutant kin-
Exon 8/9 mutations ETrare ases for degradation. Transforming activity
MF 6% requires loss of this function.
IKZF1 (IKAROS family zinc finger 1) 7p12 CP-MPN rare IKZF1 is a transcription regulator and putative tumor
Mostly deletions including intragenic BP-MPN 19% suppressor
TP53 (tumor protein p53) Exons 4 through 9 17p13.1 PMF  4% A tumor suppressor protein that targets genes that
BP-MPN  27% regulate cell cycle arrest, apoptosis and DNA
repair.
SF3B1 (splicing factor 3B subunit 1) 2q33.1 PMF  7% SF3B1 is a component of the RNA spliceosome.
Exons 14 and 15, mostly SF3B1 mutations are closely associated with ring
sideroblasts.
SRSF2 (serine/arginine-rich splicing factor 2) Exon 2 17q25.1 PMF  17% SRSF2 is a component of the RNA spliceosome,
whose dysfunction promotes defects in alterna-
tive splicing.
U2AF1(U2 Small Nuclear RNA Auxiliary Factor 1) 21q22.3 PMF  16% U2AF1 is subunit of the U2 small nuclear ribonucleo-
protein auxiliary factor involved in pre-mRNA
processing

Mutational frequencies in blast phase (BP) disease are also provided.


Key: MPN, myeloproliferative neoplasms; ET, essential thrombocythemia; PV, polycythemia vera; PMF, primary myelofibrosis; MF includes both PMF and post-
ET/PV myelofibrosis; BP-MPN, blast phase MPN; CP-MPN, chronic phase MPN;.
See text for references.

prognostically-detrimental mutations [38]. Most recently, the favor- Risk factors for leukemia-free survival in PMF include 3% circu-
able prognostic impact of CALR mutations in PMF was shown to be lating blasts, platelet count <100 3 109/L and presence of unfavor-
restricted to its type 1 or type 1-like variants whereas there was no able karyotype [42,43]. Although DIPSS has been shown to predict
prognostic difference between the other driver mutations [39,40]. Fur- leukemia-free survival [44], in the aforementioned DIPSS-plus study
thermore, targeted next generation sequencing (NGS) studies have of 793 patients with PMF [26], the only two risk factors for leukemic
identified ASXL1, SRSF2, CBL, KIT, RUNX1, SH2B3 and CEBPA transformation were unfavorable karyotype and platelet count <100
mutations as having an adverse effect on overall or leukemia-free sur- 3 109/L; 10-year risk of leukemic transformation were 12% in the
vival [41]. absence of these two risk factors and 31% in the presence of one or

doi:10.1002/ajh.24592 American Journal of Hematology, Vol. 91, No. 12, December 2016 1265
Tefferi ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES

TABLE II. International Working Group for Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) Recommended Criteria for Post-Polycythemia
Vera and Post-Essential Thrombocythemia Myelofibrosis [9]

Criteria for post-polycythemia vera myelofibrosis

Required criteria:
1 Documentation of a previous diagnosis of polycythemia vera as defined by the WHO criteria (see Table II)
2 Bone marrow fibrosis grade 2–3 (on 0–3 scale) or grade 3–4 (on 0–4 scale) (see footnote for details)
Additional criteria (two are required):
1 Anemia or sustained loss of requirement for phlebotomy in the absence of cytoreductive therapy
2 A leukoerythroblastic peripheral blood picture
3 Increasing splenomegaly defined as either an increase in palpable splenomegaly of  5 cm (distance of the tip of the spleen from the
left costal margin) or the appearance of a newly palpable splenomegaly
4 Development of  1 of three constitutional symptoms: >10% weight loss in 6 months, night sweats, unexplained fever (>37.58C)
Criteria for post-essential thrombocythemia myelofibrosis
Required criteria:
1 Documentation of a previous diagnosis of essential thrombocythemia as defined by the WHO criteria (see Table II)
2 Bone marrow fibrosis grade 2–3 (on 0–3 scale) or grade 3–4 (on 0–4 scale) (see footnote for details)
Additional criteria (two are required):
1 Anemia and a  2 g/dL decrease from baseline hemoglobin level
2 A leukoerythroblastic peripheral blood picture
3 Increasing splenomegaly defined as either an increase in palpable splenomegaly of  5 cm (distance of the tip of the spleen from the left
costal margin) or the appearance of a newly palpable splenomegaly
4 Increased lactate dehydrogenase
5 Development of  1 of three constitutional symptoms: >10% weight loss in 6 months, night sweats, unexplained fever (>37.58C)

Grade 2–3 according to the European classification: [94] diffuse, often coarse fiber network with no evidence of collagenization (negative trichrome stain) or
diffuse, coarse fiber network with areas of collagenization (positive trichrome stain). Grade 3–4 according to the standard classification: [95] diffuse and
dense increase in reticulin with extensive intersections, occasionally with only focal bundles of collagen and/or focal osteosclerosis or diffuse and dense
increase in reticulin with extensive intersections with coarse bundles of collagen, often associated with significant osteosclerosis.

Figure 3. Survival data of 793 patients with primary myelofibrosis evaluated at time of their first Mayo Clinic referral and stratified by their Dynamic Interna-
tional Prognostic Scoring System (DIPSS-plus) that employs eight variables: Age>65 yrs; Hgb <10 g/dL;R8C transfusion-dependent; platelets<1003 10(9)/L;
WBC > 25 3 10(9)/L;>1% circulating blasts; constitutional symptoms; karyotype. [Color figure can be viewed at wileyonlinelibrary.com]

both risk factors. As is becoming evident for overall survival, 䊏 Risk-Adapted Therapy
leukemia-free survival is also significantly compromised in patients
carrying certain mutations including IDH and SRSF2 [31,33]. CALR/ The only treatment modality that is currently capable of prolong-
ASXL1 mutational status and the number of prognostically- ing survival or potential cure in PMF is allogeneic stem cell trans-
detrimental mutations, as outlined above, were also predictive of leu- plant (ASCT). Unfortunately, ASCT in PMF is currently associated
kemic transformation [37,38]. In the most recent NGS study of PMF with at least 50% rate of transplant-related deaths or severe morbidi-
patients mentioned above, leukemia-free survival was adversely affect- ty, regardless of the intensity of conditioning regimens used [45].
ed by SRSF2, RUNX1, CEBPA and SH2B3 mutations [41]. Therefore, for the individual patient, the risk of ASCT must be justi-
fied, in the context of disease prognosis. On the other hand, current

1266 American Journal of Hematology, Vol. 91, No. 12, December 2016 doi:10.1002/ajh.24592
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES Primary myelofibrosis

Figure 4. Clinical and molecular risk stratification and risk-adapted therapy in primary myelofibrosis. [Color figure can be viewed at wileyonlinelibrary.com]

drug therapy for PMF is mostly palliative in scope and has not been that might alleviate anemia in a subset of JAK2-mutated patients
shown to favorably modify disease natural history or prolong survival; without marked splenomegaly or excess circulating blasts [59].
specifically, JAK inhibitor treatment in PMF has not been shown to Among currently investigational drugs (not FDA approved as yet),
reverse bone marrow fibrosis or induce complete or partial remis- those that have shown promise for the treatment of anemia include
sions; instead, its value is limited to symptoms relief and reduction in the JAK2 inhibitor momelotinib [60] and the telomerase inhibitor
spleen size [46–49]. imetelstat [61].
First-line therapy for MF-associated splenomegaly is hydroxyurea,
Management of DIPSS-plus low or intermediate-1 risk which is effective in reducing spleen size by half in approximately
patients 40% of patients [62]. Spleen response to hydroxyurea lasts for an
There is no evidence to support the value of specific therapy in average of one year and treatment side effects include myelosuppres-
asymptomatic patients with DIPSS-plus low or intermediate-1 risk sion and mucocutaneous ulcers. Both thalidomide and lenalidomide
disease. However, some of these patients might require palliative ther- might improve splenomegaly and thrombocytopenia in some patients
apy for anemia, splenomegaly, non-hepatosplenic EMH, bone pain, [52–54,56] The related agent pomalidomide might also alleviate
EMH-associated pulmonary hypertension or constitutional symptoms. thrombocytopenia in MF but is ineffective for the treatment of
In addition, cytoreductive therapy might be needed in the presence of splenomegaly [59]. Interferon (IFN)-a is of limited value in the treat-
extreme leukocytosis or thrombocytosis. ment of MF-associated splenomegaly [63]. The degree of splenomega-
Treatment of MF-associated anemia includes androgens (e.g., tes- ly in low or intermediate-1 risk patients is often not severe enough to
tosterone enanthate 400-600 mg IM weekly, oral fluoxymesterone require either splenectomy or radiotherapy. Similarly, I do not con-
10 mg TID) [50], prednisone (0.5 mg/kg/day) [50], danazol (600 mg/ sider the use of ruxolutinib (JAK1/2 inhibitor) in low or
day) [51], thalidomide (50 mg/day) 6 prednisone [52–54] or lenalido- intermediate-1 risk patients, considering the acute (e.g., anemia,
mide (10 mg/day) 6 prednisone [55,56] (10 mg/day). I do not use thrombocytopenia) and long-term toxicity, known (e.g., immunosup-
erythropoiesis stimulating agents (ESAs) because they are ineffective pression) and unknown, of the particular agent [64–67]. The same is
in transfusion-dependent patients and could exacerbate splenomegaly true for other investigational drugs that are currently not approved
[57]. Response rates to each one of the aforementioned drugs are in by the FDA [60,61].
the vicinity of 15 to 25% and response durations average about one Recommendations: Low or intermediate-1 risk asymptomatic
to two years. Lenalidomide works best in the presence of del(5q31) patients with PMF can be observed without any therapeutic interven-
[58]. Drug side effects include hepatotoxicity and virilizing effects for tion, unless they harbor high-risk mutations (e.g., ASXL1, SRSF2). Spe-
androgens, peripheral neuropathy for thalidomide and myelosuppres- cific therapy is considered only in the presence of symptoms. First-line
sion for lenalidomide. Pomalidomide is another thalidomide analog drugs of choice for symptomatic anemia include

doi:10.1002/ajh.24592 American Journal of Hematology, Vol. 91, No. 12, December 2016 1267
Tefferi ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES

thalidomide 1 prednisone, an androgen preparation or danazol. Pros- However, the benefit of the drug was antagonized by ruxolitinib-
tate cancer screening in men and monitoring of liver function tests are associated anemia (31% vs. 13.9%) and thrombocytopenia (34.2% vs.
necessary when considering treatment with androgen preparations. I 9.3%). In the COMFORT-2 trial that compared the drug with “best
use lenalidomide in the presence of del(5q) or in case of treatment fail- available therapy” (n 5 219) [71], the spleen response was 28.5% with
ure with thalidomide, danazol or androgens. First-line drug of choice ruxolitinib vs. 0% otherwise but the drug was detrimental in terms of
for symptomatic splenomegaly is hydroxyurea (starting dose 500 mg thrombocytopenia (44.5% vs. 9.6%), anemia (40.4% vs. 12.3%) and
TID). Of special challenge is the DIPSS-plus low or intermediate-1 risk diarrhea (24.0% vs. 11.0%). The long-term outcome of ruxolitinib
patient with a high-risk mutation; at present I prefer close monitoring therapy in MF was recently reported and disclosed a very high treat-
of such patients although I would not object to proceeding with ASCT ment discontinuation rate (92% after a median time of 9.2 months)
sooner than later. In general, I do not consider the use of JAK inhibi- and the occurrence of severe withdrawal symptoms during ruxolitinib
tors in low or intermediate-1 risk patients; I am concerned about more treatment discontinuation (“ruxolitinib withdrawal syndrome”) char-
harm than good because of immunosuppression associated with chronic acterized by acute relapse of disease symptoms, accelerated spleno-
use and exacerbation of anemia associated with some of these agents. megaly, worsening of cytopenias and occasional hemodynamic
decompensation, including a septic shock-like syndrome [48]. The 3-
Management of intermediate-2 or high-risk disease year follow-up information on COMFORT-2 suggested a 55% drug
PMF patients with DIPSS-plus high or intermediate-2 risk disease, discontinuation rate and a slight but significant improvement in sur-
or those with high-risk mutations such as ASXL1 and SRSF2, should vival, which is however confounded by the cross-over design of the
be considered for ASCT or investigational drug therapy, sooner than study and lack of substantial drug effect on JAK2V617F allele burden
later. In considering ASCT as a treatment modality, one should be or bone marrow fibrosis [46]. Furthermore, several reports have now
acutely aware of the risks involved. In one of the largest studies of associated the drug with serious opportunistic infections [72].
ASCT in PMF [45], 5-year disease-free survival (DFS) and treatment- Fedratinib, a selective JAK2 inhibitor, was initially evaluated in 59
related mortality (TRM) were 33% and 35% for matched related and patients with PMF or post-PV/ET MF, in a phase-1/2 study [73]. The
27% and 50% for unrelated transplants, respectively. Of note, out- dose limiting toxicity (DLT) was a reversible and asymptomatic
come did not appear to be favorably affected by reduced intensity increase in serum amylase/lipase and the maximum tolerated dose
conditioning (RIC). In another RIC transplant study, 5-year DFS was (MTD) was 680 mg/day. Grade 3 or 4 adverse events were all revers-
estimated at 51%; chronic graft-versus-host disease (cGVHD) ible and dose-dependent and included nausea (3%), vomiting (3%),
occurred in 49% of the patients and relapse (29%) was predicted by diarrhea (10%), asymptomatic mild increases in serum lipase (27%),
high-risk disease and prior splenectomy [68]. In the earlier study transaminases (27%) or creatinine (24%), thrombocytopenia (24%)
[45], the respective cGVHD and relapse rates for matched related and anemia (35%). By 6 or 12 months of treatment, 39% and 47% of
transplants were 40% and 32% and history of splenectomy did not patients, respectively, experienced a 50% decrease in palpable spleen
affect outcome. More recent outcome reports on ASCT in MF were size. In addition, the majority of patients with early satiety, fatigue,
more encouraging: 100-day mortality 13%, a relapse rate of 11%, and night sweats, cough or pruritus reported a durable resolution of their
a 7-year survival of 61% [69]. There is currently much interest in symptoms. Almost all patients with thrombocytosis and the majority
evaluating the use of JAK inhibitors before transplant with favorable with leukocytosis had normalization of their counts. Among 23
or unfavorable experiences reported by different investigators. I do patients with a baseline JAK2V617F allele burden of >20%, 9 (39%)
not currently advise the use of JAK inhibitors in the context of trans- had 50% decrease in allele burden. Effect on bone marrow patholo-
plant until more information on its value becomes available. gy was limited. In general, response was not affected by the presence
In my opinion, all non-transplant candidates with high or of JAK2V617F. Most recently, the results of a phase-3 study
intermediate-2 risk disease should first be offered participation in (n 5 289) comparing fedratinib at two different doses (500 or
investigational drug therapy before treatment with conventional drugs 400 mg/day) with placebo were disclosed and confirmed the efficacy
including JAK inhibitors. This is because the latter agents are unlikely of the drug in inducing spleen (49%, 47% and 1%, respectively) and
to significantly modify the natural history of the disease and would symptom response. However, reports of encephalopathy associated
not prevent progression into acute leukemia. JAK inhibitors are rea- with the use of the drug resulted in the withdrawal of the drug from
sonable palliative agents in patients who are both non-transplant can- further development. Other side effects included anemia (grade 3 or
didates and are unable to participate in clinical trials. JAK inhibitor 4 in 43% to 60%), thrombocytopenia (grade 3 or 4 in 17% to 27%)
ATP mimetics that have undergone clinical trials in MPN include and diarrhea (56% to 66%) [74].
ruxolitinib (INCB018424), fedratinib (SAR302503), momelotinib Momelotinib (MMB, GS-0387, CYT387) is a JAK1 and JAK2
(CYT387) and pacritinib (SB1518). Results of these studies so far sug- inhibitor. Among the first 60 patients patients treated in a phase-1/2
gest substantial differences among these drugs in their toxicity and study [75], MTD was 300 mg/day and DLT included grade 3 head-
efficacy profiles, some of which might be linked to their variable in ache and hyperlipasemia. Anemia and spleen responses were 59%
vitro activity against other JAK and non-JAK kinase targets. Among and 48%, respectively. Among 33 patients who were red cell-
the aforementioned agents, ruxolitinib is now FDA approved, fedrati- transfused in the month prior to study entry, 70% achieved a mini-
nib was withdrawn from the market because of side effects (encepha- mum 12-week period without transfusions (range 4.7->18.3 months).
lopathy), pacritinib is currently on FDA-imposed full clinical hold Most patients experienced constitutional symptoms improvement.
because of side effects (deaths from intracranial hemorrhage, cardiac Grade 3/4 adverse reactions included thrombocytopenia (32%),
arrest and heart failure) and momelotinib is undergoing phase-3 hyperlipasemia (5%), elevated liver transaminases (3%) and headache
study.///// (3%). New-onset treatment-related peripheral neuropathy was
Two randomized studies comparing ruxolitinib with either placebo observed in 22% of patients (sensory symptoms, grade 1) and the
or best supportive care have now been published [70,71]. In the particular side effect was further elaborated in a subsequent follow-up
COMFORT-1 trial that compared the drug with placebo (n 5 309) study [76]. The study was subsequently expanded to include 166
[70], the spleen response rate was approximately 42% for ruxolitinib patients treated at either 150 mg or 300 mg once-daily, or 150 mg
versus <1% for placebo. In addition, about 46% of patients experi- twice-daily for 9 months, with similar results [77]. The drug is cur-
enced substantial improvement in their constitutional symptoms. rently undergoing phase-3 study.

1268 American Journal of Hematology, Vol. 91, No. 12, December 2016 doi:10.1002/ajh.24592
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES Primary myelofibrosis

Pacritinib (SB1518) is a JAK2/FLT3 inhibitor. In phase-1 studies, experimental drug therapy. I have yet to be satisfied by the value of
myelosuppression was minimal and 400 mg/d was chosen as the rec- any currently available JAK inhibitor and strongly advise patients to
ommended dose for phase-2 study, which included 34 patients [78]. continue participating in clinical trials.
The most common treatment-related adverse events were gastrointes-
tinal, especially diarrhea. Spleen response rate was 44% (32% by Management of refractory disease and specific disease
MRI) and accompanied by symptoms response. Anemia response was complications
reported in 2 (6%) patients. The drug was undergoing phase-3 study, Hydroxyurea-refractory splenomegaly is often managed by splenec-
compared to best available therapy, when increased deaths from tomy [82]. Other indications for splenectomy include symptomatic
intracranial hemorrhage, cardiac arrest and heart failure were portal hypertension, thrombocytopenia and frequent red blood cell
observed and resulted in FDA-imposed full clinical hold. transfusions. In a recent report of 314 splenectomized patients with
The above observations demonstrate major differences in toxicity MF [83], more than 75% benefited from the procedure and the bene-
and activity profile among several JAK inhibitor small molecules and fit lasted for a median of one year; specific benefits included becom-
underscore the need to evaluate more such drugs before making any ing transfusion-independent and resolution of severe
conclusions regarding the value of anti-JAK2 therapy in MF or relat- thrombocytopenia. Perioperative complications occurred in 28% of
ed MPN. It is also becoming evident that some of the salutary effects the patients and included infections, abdominal vein thrombosis and
of these drugs might be the result of a potent anti-cytokine activity. bleeding. Overall perioperative mortality rate was 9%. Approximately
JAK-STAT activation leads to Akt/mTOR activation as well and it 10% of patients experienced progressive hepatomegaly and 29%
is therefore reasonable to evaluate the therapeutic activity of Akt and thrombocytosis after splenectomy. Median survival after splenectomy
mTOR inhibitors. In a phase 1/2 study involving the mTOR inhibitor was 19 months. Leukemic transformation was documented in 14% of
everolimus including 39 MF patients, the commonest toxicity was patients whose survival was not different than that of patients without
grade 1-2 stomatitis. A >50% reduction in splenomegaly occurred in “leukemic transformation,” although others had suggested otherwise
20% of the patients evaluated and the constitutional symptoms [84].
response was 69%; 80% experienced complete resolution of pruritus Splenic irradiation (100 cGy in 5–10 fractions) induces transient
[79]. Drug effect on cytosis or anemia was modest and on reduction in spleen size but can be associated with severe pancytope-
JAK2V617F burden negligible. Overall IWG-MRT response rate was nia [85]. Non-hepatosplenic EMH might involve the vertebral col-
23%. umn, lymph nodes, pleura and peritoneum (ascites) and is effectively
Currently, many other drugs are being tested for their therapeutic treated with low-dose radiotherapy (100–1000 cGy in 5 to 10 frac-
value in MF and the most intriguing results were recently reported tions) [86]. Diagnosis of MF-associated pulmonary hypertension is
with the use of imetelestat, which is a 13-mer lipid-conjugated oligo- confirmed by a technetium 99m sulphur colloid scintigraphy and
nucleotide that targets the RNA template of human telomerase treatment with single-fraction (100 cGy) whole-lung irradiation has
reverse transcriptase. In the particular study, imetelstat was adminis- been shown to be effective [87]. Single fraction of 100 to 400 cGy
tered as a 2-hour intravenous infusion (9.4 mg per kilogram of body involved field radiotherapy has also been shown to benefit patients
weight) every 1 to 3 weeks. A complete or partial remission occurred with MF-associated extremity pain [88].
in 7 of 33 patients (21%), with a median duration of response of 18 Transjugular intrahepatic portosystemic shunt might be considered
months for complete responses. Bone marrow fibrosis was reversed in to alleviate symptoms of portal hypertension. Recent technical advan-
all 4 patients who had a complete response, and a molecular response ces in the procedure and the introduction of specially coated stents
occurred in three of the four patients. Response rates were 32% have greatly improved shunt patency and clinical efficacy of TIPS in
among patients without an ASXL1 mutation versus 0% among those general. Current TIPS indications include recurrent variceal bleeding
with an ASXL1 mutation. The rate of complete response was 38% and refractory ascites, both of which could accompany advanced MF.
among patients with a mutation in SF3B1 or U2AF1 versus 4% The therapeutic value of TIPS has not been systematically studied in
among patients without a mutation in these genes. Treatment-related MF but relevant information is available from several case reports
adverse events included grade 4 thrombocytopenia (in 18% of that confirm feasibility and efficacy [89].
patients), grade 4 neutropenia (in 12%), grade 3 anemia (in 30%), Recommendations: At present, my first-line choice for the manage-
and grade 1 or 2 elevation in levels of total bilirubin (in 12%), alka- ment of drug-refractory splenomegaly is participation in experimental
line phosphatase (in 21%), and aspartate aminotransferase (in 27%); drug therapy. Both splenectomy and low-dose radiotherapy are reason-
these observations have led to additional clinical trials in other mye- able alternative treatment options. Prophylactic therapy with hydroxy-
loid malignancies [61,80,81]. urea is advised to prevent post-splenectomy thrombocytosis [[82]].
Recommendations: Considering the lack of effective drug therapy in Post-splenectomy thrombosis might be prevented by instituting short
PMF, the risk of transplant-related complications might be justified in term systemic anticoagulation. Laparoscopic splenectomy is not advised
those patients in whom median survival is expected to be <5 years in the setting of MF [[90]] and data on the value of splenic artery
and leukemic transformation risk >20%. These include DIPSS-plus embolization are limited [[91–93]]. I do not believe that splenectomy
high or intermediate-2 risk patients as well as those with high-risk increases the risk of leukemic transformation [[84]].
mutations. Non-transplant candidates are best managed with

䊏 References neoplasms: JAK2, MPL, TET2, ASXL1, CBL,


IDH and IKZF1. Leukemia 2010;24:1128–1138.
6. Tefferi A. Myelofibrosis with Myeloid Metapla-
sia. N Engl J Med 2000;342:1255–1265.
1. Arber DA, Orazi A, Hasserjian R, et al. The 4. Tefferi A, Thiele J, Vannucchi AM, et al. An 7. Tefferi A. Pathogenesis of myelofibrosis with
2016 revision to the World Health Organization overview on CALR and CSF3R mutations and a myeloid metaplasia. J Clin Oncol 2005;23:8520–
classification of myeloid neoplasms and acute proposal for revision of WHO diagnostic criteria 8530.
leukemia. Blood 2016;127:2391–2405. for myeloproliferative neoplasms. Leukemia 8. Mesa RA, Li CY, Ketterling RP, et al. Leukemic
2. Barbui T, Thiele J, Gisslinger H, et al. The 2016 2014;28:1407–1413. transformation in myelofibrosis with myeloid
revision of WHO classification of myeloprolifer- 5. Tefferi A, Finke CM, Lasho TL, et al. U2AF1 metaplasia: A single-institution experience with
ative neoplasms: Clinical and molecular advan- mutations in primary myelofibrosis are strongly 91 cases. Blood 2005;105:973–977.
ces. Blood Rev, in press. associated with anemia and thrombocytopenia 9. Barosi G, Mesa RA, Thiele J, et al. Proposed cri-
3. Tefferi A. Novel mutations and their functional despite clustering with JAK2V617F and normal teria for the diagnosis of post-polycythemia vera
and clinical relevance in myeloproliferative karyotype. Leukemia 2014;28:431–433. and post-essential thrombocythemia myelofibro-
sis: A consensus statement from the

doi:10.1002/ajh.24592 American Journal of Hematology, Vol. 91, No. 12, December 2016 1269
Tefferi ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES

International Working Group for Myelofibrosis myelofibrosis: A Mayo Clinic study of 884 kar- ruxolitinib with best available therapy for mye-
Research and Treatment. Leukemia 2008;22: yotypically annotated patients. Blood 2011;118: lofibrosis. Blood 2013;122:4047–4053.
437–438. 4595–4598. 47. Tefferi A. Challenges facing JAK inhibitor thera-
10. Kvasnicka HM, Thiele J. Prodromal myeloprolif- 28. Tefferi A, Lasho TL, Patnaik MM, et al. JAK2 py for myeloproliferative neoplasms. N Engl J
erative neoplasms: The 2008 WHO classifica- germline genetic variation affects disease suscep- Med 2012;366:844–846.
tion. Am J Hematol 2010;85:62–69. tibility in primary myelofibrosis regardless of 48. Tefferi A. JAK inhibitors for myeloproliferative
11. Hussein K, Van Dyke DL, Tefferi A. Conven- V617F mutational status: Nullizygosity for the neoplasms: Clarifying facts from myths. Blood
tional cytogenetics in myelofibrosis: Literature JAK2 46/1 haplotype is associated with inferior 2012;119:2721–2730.
review and discussion. Eur J Haematol 2009;82: survival. Leukemia 2010;24:105–109. 49. Tefferi A, Litzow MR, Pardanani A. Long-term
329–338. 29. Tefferi A, Lasho TL, Huang J, et al. Low outcome of treatment with ruxolitinib in myelo-
12. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2V617F allele burden in primary myelofi- fibrosis. N Engl J Med 2011;365:1455–1457.
JAK2 vs MPL-mutated or triple-negative myelo- brosis, compared to either a higher allele burden 50. Silverstein MN, Agnogenic Myeloid Metaplasia,
fibrosis: Clinical, cytogenetic and molecular or unmutated status, is associated with inferior Vol. 1. Acton Mass, Publishing Science Group;
comparisons. Leukemia 2014;28:1472–1477. overall and leukemia-free survival. Leukemia 1975.
13. Tefferi A, Thiele J, Orazi A, et al. Proposals and 2008;22:756–761. 51. Cervantes F, Mesa R, Barosi G. New and old
rationale for revision of the World Health Orga- 30. Guglielmelli P, Barosi G, Specchia G, et al. Iden- treatment modalities in primary myelofibrosis.
nization diagnostic criteria for polycythemia tification of patients with poorer survival in pri- Cancer J 2007;13:377–383.
vera, essential thrombocythemia, and primary mary myelofibrosis based on the burden of 52. Elliott MA, Mesa RA, Li CY, et al. Thalidomide
myelofibrosis: Recommendations from an ad JAK2V617F mutated allele. Blood 2009;114: treatment in myelofibrosis with myeloid meta-
hoc international expert panel. Blood 2007;110: 1477–1483. plasia. Br J Haematol 2002;117:288–296.
1092–1097. 31. Tefferi A, Jimma T, Sulai NH, et al. IDH muta- 53. Thomas DA, Giles FJ, Albitar M, et al. Thalido-
14. Tefferi A, Pardanani A. Genetics: CALR muta- tions in primary myelofibrosis predict leukemic mide therapy for myelofibrosis with myeloid
tions and a new diagnostic algorithm for MPN. transformation and shortened survival: Clinical metaplasia. Cancer 2006;106:1974–1984.
Nat Rev Clin Oncol 2014;11:125–126. evidence for leukemogenic collaboration with 54. Mesa RA, Steensma DP, Pardanani A, et al. A
15. Barbui T, Thiele J, Passamonti F, et al. Survival JAK2V617F. Leukemia 2012;26:475–480. phase 2 trial of combination low-dose thalido-
and disease progression in essential thrombocy- 32. Guglielmelli P, Biamonte F, Score J, et al. EZH2 mide and prednisone for the treatment of mye-
themia are significantly influenced by accurate mutational status predicts poor survival in mye- lofibrosis with myeloid metaplasia. Blood 2003;
morphologic diagnosis: An international study. lofibrosis. Blood 2011;118:5227–5234. 101:2534–2541.
J Clin Oncol 2011;29:3179–3184. 33. Lasho TL, Jimma T, Finke CM, et al. SRSF2 55. Tefferi A, Cortes J, Verstovsek S, et al. Lenalido-
16. Steensma DP, Hanson CA, Letendre L, et al. mutations in primary myelofibrosis: Significant mide therapy in myelofibrosis with myeloid
Myelodysplasia with fibrosis: A distinct entity? clustering with IDH mutations and independent metaplasia. Blood 2006;108:1158–1164.
Leuk Res 2001;25:829–838. association with inferior overall and leukemia- 56. Quintas-Cardama A, Kantarjian HM,
17. Patnaik MM, Parikh SA, Hanson CA, et al. free survival. Blood 2012;120:4168–4171. Manshouri T, et al. Lenalidomide plus predni-
Chronic myelomonocytic leukaemia: A concise 34. Vannucchi AM, Lasho TL, Guglielmelli P, et al. sone results in durable clinical, histopathologic,
clinical and pathophysiological review. Brit J Mutations and prognosis in primary myelofibro- and molecular responses in patients with myelo-
Haematol 2014;165:273–286. sis. Leukemia 2013;27:1861–1869. fibrosis. J Clin Oncol 2009;27:4760–4766.
18. Orazi A, O’Malley DP, Jiang J, et al. Acute pan- 35. Pardanani A, Lasho TL, Finke CM, et al. Poly- 57. Huang J, Tefferi A. Erythropoiesis stimulating
myelosis with myelofibrosis: An entity distinct clonal immunoglobulin free light chain levels agents have limited therapeutic activity in
from acute megakaryoblastic leukemia. Mod predict survival in myeloid neoplasms. J Clin transfusion-dependent patients with primary
Pathol 2005;18:603–614. Oncol 2012;30:1087–1094. myelofibrosis regardless of serum erythropoietin
19. Cervantes F, Dupriez B, Pereira A, et al. New 36. Tefferi A, Vaidya R, Caramazza D, et al. Circu- level. Eur J Haematol 2009;83:154–155.
prognostic scoring system for primary myelofi- lating interleukin (IL)28, IL-2R, IL-12, and IL- 58. Tefferi A, Lasho TL, Mesa RA, et al. Lenalido-
brosis based on a study of the International 15 levels are independently prognostic in prima- mide therapy in del(5)(q31)-associated myelofi-
Working Group for Myelofibrosis Research and ry myelofibrosis: A comprehensive cytokine pro- brosis: Cytogenetic and JAK2V617F molecular
Treatment. Blood 2009;113:2895–2901. filing study. J Clin Oncol 2011;29:1356–1363. remissions. Leukemia 2007;21:1827–1828.
20. Passamonti F, Cervantes F, Vannucchi AM, 37. Tefferi A, Guglielmelli P, Lasho TL, et al. CALR 59. Begna KH, Pardanani A, Mesa R, et al. Long-
et al. A dynamic prognostic model to predict and ASXL1 mutations-based molecular prognos- term outcome of pomalidomide therapy in mye-
survival in primary myelofibrosis: A study by tication in primary myelofibrosis: An interna- lofibrosis. Am J Hematol 2012;87:66–68.
the IWG-MRT (International Working Group tional study of 570 patients. Leukemia 2014;28: 60. Pardanani A, Abdelrahman RA, Finke C, et al.
for Myeloproliferative Neoplasms Research and 1494–1500 Genetic determinants of response and survival
Treatment). Blood 2010;115:1703–1708. 38. Guglielmelli P, Lasho TL, Rotunno G, et al. The in momelotinib-treated patients with myelofi-
21. Hussein K, Pardanani AD, Van Dyke DL, et al. number of prognostically detrimental mutations brosis. Leukemia 2015;29:741–744.
International Prognostic Scoring System- and prognosis in primary myelofibrosis: An 61. Tefferi A, Lasho TL, Begna KH, et al. A Pilot
independent cytogenetic risk categorization in international study of 797 patients. Leukemia Study of the Telomerase Inhibitor Imetelstat for
primary myelofibrosis. Blood 2010;115:496–499. 2014;28:1804–1810 Myelofibrosis. N Engl J Med 2015;373:908–919.
22. Caramazza D, Begna KH, Gangat N, et al. 39. Tefferi A, Lasho TL, Tischer A, et al. The prog- 62. Martinez-Trillos A, Gaya A, Maffioli M, et al.
Refined cytogenetic-risk categorization for over- nostic advantage of calreticulin mutations in Efficacy and tolerability of hydroxyurea in the
all and leukemia-free survival in primary myelo- myelofibrosis might be confined to type 1 or treatment of the hyperproliferative manifesta-
fibrosis: A single center study of 433 patients. type 1-like CALR variants. Blood 2014;124: tions of myelofibrosis: Results in 40 patients.
Leukemia 2011;25:82–88. 2465–2466. Ann Hematol 2010;89:1233–1237.
23. Tefferi A, Siragusa S, Hussein K, et al. Transfu- 40. Guglielmelli P, Rotunno G, Fanelli T, et al. Vali- 63. Tefferi A, Elliot MA, Yoon SY, et al. Clinical
sion-dependency at presentation and its acquisi- dation of the differential prognostic impact of and bone marrow effects of interferon alfa ther-
tion in the first year of diagnosis are both type 1/type 1-like versus type 2/type 2-like apy in myelofibrosis with myeloid metaplasia.
equally detrimental for survival in primary CALR mutations in myelofibrosis. Blood Cancer Blood 2001;97:1896.
myelofibrosis-prognostic relevance is indepen- J 2015;5:e360. 64. Marchetti M, Barosi G, Cervantes F, et al.
dent of IPSS or karyotype. Am J Hematol 2009; 41. Tefferi A, Lasho T, Finke C, et al. Targeted deep Which patients with myelofibrosis should
85:14–17. sequencing in primary myelofibrosis. Blood receive ruxolitinib therapy? ELN-SIE evidence-
24. Elena C, Passamonti F, Rumi E, et al. Red blood Adv, in press. based recommendations. Leukemia, in press.
cell transfusion-dependency implies a poor sur- 42. Tam CS, Kantarjian H, Cortes J, et al. Dynamic 65. Rudolph J, Heine A, Quast T, et al. The JAK
vival in primary myelofibrosis irrespective of model for predicting death within 12 months in inhibitor ruxolitinib impairs dendritic cell
International Prognostic Scoring System and patients with primary or post-polycythemia migration via off-target inhibition of ROCK.
Dynamic International Prognostic Scoring Sys- vera/essential thrombocythemia myelofibrosis. Leukemia 2016;30:2119–2123.
tem. Haematologica 2011;96:167–170. J Clin Oncol 2009;27:5587–5593. 66. Chen CC, Chen YY, Huang CE. Cryptococcal
25. Patnaik MM, Caramazza D, Gangat N, et al. 43. Huang J, Li CY, Mesa RA, et al. Risk factors for meningoencephalitis associated with the long-
Age and platelet count are IPSS-independent leukemic transformation in patients with prima- term use of ruxolitinib. Ann Hematol 2016;95:
prognostic factors in young patients with prima- ry myelofibrosis. Cancer 2008;112:2726–2732. 361–362.
ry myelofibrosis and complement IPSS in pre- 44. Passamonti F, Cervantes F, Vannucchi AM, 67. von Hofsten J, Johnsson Forsberg M, Zetterberg
dicting very long or very short survival. Eur J et al. Dynamic International Prognostic Scoring M. Cytomegalovirus retinitis in a patient who
Haematol 2010;84:105–108. System (DIPSS) predicts progression to acute received ruxolitinib. N Engl J Med 2016;374:
26. Gangat N, Caramazza D, Vaidya R, et al. DIPSS myeloid leukemia in primary myelofibrosis. 296–297.
plus: A refined Dynamic International Prognos- Blood 2010;116:2857–2858. 68. Kroger N, Holler E, Kobbe G, et al. Allogeneic
tic Scoring System for primary myelofibrosis 45. Ballen KK, Shrestha S, Sobocinski KA, et al. stem cell transplantation after reduced-intensity
that incorporates prognostic information from Outcome of transplantation for myelofibrosis. conditioning in patients with myelofibrosis: A
karyotype, platelet count, and transfusion status. Biol Blood Marrow Transplant 2010;16:358–367. prospective, multicenter study of the Chronic
J Clin Oncol 2011;29:392–397. 46. Cervantes F, Vannucchi AM, Kiladjian JJ, et al. Leukemia Working Party of the European
27. Tefferi A, Jimma T, Gangat N, et al. Predictors Three-year efficacy, safety, and survival findings Group for Blood and Marrow Transplantation.
of greater than 80% 2-year mortality in primary from COMFORT-II, a phase 3 study comparing Blood 2009;114:5264–5270.

1270 American Journal of Hematology, Vol. 91, No. 12, December 2016 doi:10.1002/ajh.24592
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES Primary myelofibrosis

69. Deeg HJ, Appelbaum FR. Indications for and patients with primary, post-polycythemia vera, myelofibrosis with myeloid metaplasia. Br J
current results with allogeneic hematopoietic and post-essential thrombocythemia myelofibro- Haematol 2002;118:813–816.
cell transplantation in patients with myelofibro- sis. ASH Annual Meeting Abstracts 2011;118: 88. Neben-Wittich MA, Brown PD, Tefferi A. Suc-
sis. Blood 2011;117:7185. 282. cessful treatment of severe extremity pain in
70. Verstovsek S, Mesa RA, Gotlib J, et al. A 79. Guglielmelli P, Barosi G, Rambaldi A, et al. myelofibrosis with low-dose single-fraction radi-
double-blind, placebo-controlled trial of ruxoliti- Safety and efficacy of everolimus, a mTOR ation therapy. Am J Hematol 2010;85:808–810.
nib for myelofibrosis. N Engl J Med 2012;366: inhibitor, as single agent in a phase 1/2 study in 89. Mishchenko E, Tefferi A. Treatment options for
799–807. patients with myelofibrosis. Blood 2011;118: hydroxyurea-refractory disease complications in
71. Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK 2069–2076. myeloproliferative neoplasms: JAK2 inhibitors,
inhibition with ruxolitinib versus best available 80. Tefferi A, Al-Kali A, Begna KH, et al. Imetelstat radiotherapy, splenectomy and transjugular
therapy for myelofibrosis. N Engl J Med 2012; therapy in refractory anemia with ring sidero- intrahepatic portosystemic shunt. Eur J Haema-
366:787–798. blasts with or without thrombocytosis. Blood tol 2010;85:192–199.
72. Heine A, Brossart P, Wolf D. Ruxolitinib is a Cancer J 2016;6:e405. 90. Feldman LS, Demyttenaere SV,
potent immunosuppressive compound: Is it time 81. Tefferi A. Telomerase inhibitor imetelstat in Polyhronopoulos GN, et al. Refining the selec-
for anti-infective prophylaxis? Blood 2013;122: essential thrombocythemia and myelofibrosis. N tion criteria for laparoscopic versus open sple-
3843–3844. Engl J Med 2015;373:2580–2581. nectomy for splenomegaly. J Laparoendosc Adv
73. Pardanani A, Gotlib JR, Jamieson C, et al. Safety 82. Tefferi A, Mesa RA, Nagorney DM, et al. Sple- Surg Tech 2008;18:13–19.
and efficacy of TG101348, a selective JAK2 nectomy in myelofibrosis with myeloid metapla- 91. Iwase K, Higaki J, Mikata S, et al. Laparoscopi-
inhibitor, in myelofibrosis. J Clin Oncol 2011;29: sia: A single-institution experience with 223 cally assisted splenectomy following preoperative
789–796. patients. Blood 2000;95:2226–2233. splenic artery embolization using contour embo-
74. Pardanani A, Harrison CN, Cortes JE, et al. 83. Mesa RA, Nagorney DS, Schwager S, et al. Palli- li for myelofibrosis with massive splenomegaly.
Results of a randomized, double-blind, placebo- ative goals, patient selection, and perioperative Surg Laparosc, Endosc Percutan Tech 1999;9:
controlled phase III study (JAKARTA) of the platelet management: Outcomes and lessons 197–202.
JAK2-selective inhibitor fedratinib (SAR302503) from 3 decades of splenectomy for myelofibrosis 92. Hiatt JR, Gomes AS, Machleder HI. Massive
in patients with myelofibrosis (MF). Blood 2013; with myeloid metaplasia at the Mayo Clinic. splenomegaly. Superior results with a combined
122:393. Cancer 2006;107:361–370. endovascular and operative approach. Arch Surg
75. Pardanani A, Laborde RR, Lasho TL, et al. Safe- 84. Barosi G, Ambrosetti A, Centra A, et al. Sple- 1990;125:1363–1367.
ty and efficacy of CYT387, a JAK1 and JAK2 nectomy and risk of blast transformation in 93. Hocking WG, Machleder HI, Golde DW. Splen-
inhibitor, in myelofibrosis. Leukemia 2013;27: myelofibrosis with myeloid metaplasia. Italian ic artery embolization prior to splenectomy in
1322–1327. Cooperative Study Group on Myeloid with Mye- end-stage polycythemia vera. Am J Hematol
76. Abdelrahman RA, Begna KH, Al-Kali A, et al. loid Metaplasia. Blood 1998;91:3630–3636. 1980;8:123–127.
Momelotinib treatment-emergent neuropathy: 85. Elliott MA, Chen MG, Silverstein MN, et al. 94. Thiele J, Kvasnicka HM, Facchetti F, et al. Euro-
Prevalence, risk factors and outcome in 100 Splenic irradiation for symptomatic splenomega- pean consensus on grading bone marrow fibro-
patients with myelofibrosis. Br J Haematol 2015; ly associated with myelofibrosis with myeloid sis and assessment of cellularity. Haematologica
169:77–80. metaplasia. Br J Haematol 1998;103:505–511. 2005;90:1128–1132.
77. Pardanani A, Gotlib J, Gupta V, et al. Update 86. Koch CA, Li CY, Mesa RA, et al. Nonhepatos- 95. Manoharan A, Horsley R, Pitney WR. The retic-
on the long-term efficacy and safety of momelo- plenic extramedullary hematopoiesis: Associated ulin content of bone marrow in acute leukaemia
tinib, a JAK1 and JAK2 inhibitor, for the treat- diseases, pathology, clinical course, and treat- in adults. Br J Haematol 1979;43:185–190.
ment of myelofibrosis. Blood 2013;122:108. ment. Mayo Clin Proc 2003;78:1223–1233.
78. Komrokji RS, Wadleigh M, Seymour JF, et al. 87. Steensma DP, Hook CC, Stafford SL, et al. Low-
Results of a phase 2 study of Pacritinib dose, single-fraction, whole-lung radiotherapy
(SB1518), a novel oral JAK2 inhibitor, in for pulmonary hypertension associated with

doi:10.1002/ajh.24592 American Journal of Hematology, Vol. 91, No. 12, December 2016 1271

Você também pode gostar