Transformation of Philadelphia (Ph)–negative myeloproliferative neoplasms (MPNs) to myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) is associated with poor response to chemotherapy and short survival. Fifty-four patients with Ph-negative MPN (including 21 essential thrombocythemia [ET], 21 polycythemia vera [PV], 7 primary myelofibrosis, and 5 unclassified MPN) who had progressed to AML (n = 26) or MDS (n = 28) were treated with azacitidine in a patient-named program. Overall response rate was 52% (24% complete response [CR], 11% partial response [PR], 8% marrow CR or CR with incomplete recovery of cytopenias, 9% hematologic improvement) and median response duration was 9 months. Prognostic factors were for overall response the underlying MPN (71% vs 33% responses in ET and PV, respectively; P = .016); prognostic factors for CR achievement were the underlying MPN (14% CR for PV vs 43% for ET; P = .040) and World Health Organization classification at transformation (36% vs 12% CR in MDS and AML, respectively, P = .038). Recurrence of chronic phase features of the initial MPN was observed in 39% of the responders. Median overall survival was 11 months. Azacitidine gives encouraging results in Ph-negative MPN having progressed to AML or MDS, but response duration is short, and consolidation treatments have to be evaluated.

Philadelphia (Ph)–negative myeloproliferative neoplasm (MPN) progress to acute myeloid leukemia (AML), preceded or not by myelodysplastic syndromes (MDS) in 5%-10% of the cases after 10 years and probably at an even higher incidence with longer follow-up.1-4  Those transformations are associated with poor response to chemotherapy (including anthracycline/cytosine arabinoside–based intensive chemotherapy [IC] or lower dose regimens), to a high risk of relapse after allogeneic stem cell transplantation (SCT), resulting in poor survival in most cases.5,6 

Aberrant cytosine-phosphate-guanosine island methylation of tumor suppressor genes plays a key role in the progression of MDS.7  In MPN, the mechanisms underlying progression to MDS and AML are poorly known, although hypermethylation of p15INK4b and p16INK4a genes has been described during the leukemic evolution of myeloid metaplasia.8  The hypomethylating agent azacitidine has shown significant survival benefit in higher risk MDS.9,10  In MPN, azacitidine appears to have limited efficacy in primary myelofibrosis (PMF) and myelofibrosis occurring during the evolution of essential thrombocythemia (ET) or polycythemia vera (PV) despite the induction of global hypomethylation.11,12  However, to our knowledge, response to azacitidine has not been evaluated in series of MDS and AML occurring during the course of Ph-negative MPN. We report here our experience with azacitidine treatment in 54 patients with MDS or AML secondary to MPN.

Patients

After approval of azacitidine by the US Food and Drug Administration (FDA) for the treatment of MDS in 2004 and before approval by the European Agency for the Evaluation of Medicinal Products (EMEA) at the end of 2008, the French health agency (Agence Française de Sécurité Sanitaire des Produits de Santé [AFSSAPS]) opened a compassionate patient-named program (Autorisation Temporaire d'Utilization) of azacitidine in higher risk MDS and poor-risk AML, in cooperation with the Groupe Francophone des Myélodysplasies (GFM). All patients with international prognostic scoring system intermediate 2 or high-risk MDS (de novo or therapy related) could be included in this program. For untreated AML, patients could only enter the program if they were considered “unfit” for IC, based on age (generally > 65 years), comorbidities, or on characteristics associated with poor response to IC (AML after MDS or MPN, complex cytogenetic findings). All applications were reviewed by AFSSAPS (including review by one expert in the field) for approval, and patient informed consent was required before inclusion. A short case report form was sent to the treating physician after inclusion.

Between September 2004 and January 2009, 931 patients were included in this program; 735 of them, who had received ≥ 1cycle of azacitidine and were treated in centers that had accepted to send data for all their patients included in the program to GFM, were considered evaluable (centers that did not send information on all their patients were considered not evaluable, to avoid any bias). All participating centers approved this study. Of those 735 patients, 54 with MDS or AML after Ph-negative MPN form the basis of the present study. The study was conducted according to the Declaration of Helsinki.

Diagnosis and classification of MDS and AML was made according to the World Health Organization (WHO) classification.13  However, because azacitidine has also been approved by FDA and EMEA in refractory anemia with excess blasts in transformation (RAEB-t), considered as AML in the WHO classification but as MDS in the French-American-British (FAB) classification,14  analysis of results was also made with FAB criteria for AML and MDS. Although most initial diagnoses of MPN have been made before the Janus kinase-2 (JAK2) V617F era, using both the Polycythemia Vera Study Group15,16  and WHO 2001 criteria,17  JAK2 mutational status was available in 41 patients. Patients with unclassified MPN as the primary disorder, including atypical chronic myeloid leukemia (CML) according to WHO 2008 criteria,13  were eligible, but chronic myelomonocytic leukemia and refractory anemia with ringed sideroblasts with thrombocytosis as primary or secondary disorder were excluded.

Treatment

Azacitidine was to be administered subcutaneously at the approved FDA/EMEA schedule (75 mg/m2/d during 7 days every 28 days) for ≥ 4 cycles. Dose reductions were made in case of grade > 2 side effects according to National Cancer Institute toxicity criteria (Common Terminology Criteria for Adverse Events v3.0), except for neutropenia or thrombocytopenia or both. Delays in cycles were recommended in case of grade 4 cytopenias, until recovery above baseline values or on the physician's decision. All responders after 4-6 cycles of azacitidine were to continue treatment until progression. Transfusion thresholds, in agreement with AFSSAPS were as follows. Recommendations for red blood cell transfusions were hemoglobin (Hb) level of < 8 g/dL or higher (9-10 g/dL) in case of severe infection, underlying cardiac or pulmonary disease, or severe symptoms of anemia; recommendations for prophylactic platelet transfusions were platelet count < 20 × 109/L or less in case of fever, rapid platelet decrease, mucositis, or concomitant coagulopathy.

Assessment of response

Response was evaluated after 4-6 cycles by blood count, bone marrow aspirate, and cytogenetic studies. Response was defined for MDS as achievement of complete response (CR), partial response (PR), marrow CR (mCR) or hematologic improvement (HI) according to the International Working Group (IWG) 2006 criteria18 ; for AML as achievement of CR, PR, or CRi (ie, CR with incomplete recovery of cytopenias) according to IWG-AML 2003 criteria.19  Patients in CR/CRi were allowed to have persistent blood or marrow features or both of MPN, provided the blast percentage and peripheral blood cytopenias satisfied response criteria.20  Complete cytogenetic response was defined by the disappearance of all chromosomal abnormalities without appearance of new ones and partial cytogenetic response by at least a 50% reduction of the number of mitoses with any chromosomal abnormality, in agreement with IWG 2006 criteria for MDS.20  Recurrence of initial features of MPN was defined by a Hb level > 17 g/dL in men and > 15 g/dL in women if associated with an increase of ≥ 0.2 g/L from a person's baseline value, or a sustained platelet count > 450 × 109/L, in agreement with WHO 2008 criteria for MPN classification.13 

Data collection and statistical analysis

Because the case report form sent to treating centers on inclusion was relatively short, missing data were subsequently collected by clinical research associates of the GFM. Prognostic factors of response were analyzed in the whole population and separately in MDS and AML by univariate and multivariate analyses (the latter with the use of a logistic regression model). Characteristics were compared by chi-square or nonparametric tests (Fisher exact test) for qualitative variables, and quantitative variables were compared by Wilcoxon and Kruskal-Wallis tests. Censored end points were estimated by the nonparametric Kaplan-Meier method,21  then compared between groups by the log-rank test.22  Analyses were made at the reference date of December 1, 2009, with R software 2.8.1 (The R Project).

Pretreatment characteristics of the patient population

The study population consisted of 34 men and 20 women, with a median age of 69.5 years (range, 37-89 years) at inclusion in the azacitidine program. The initial MPN was PV in 21 patients (39%), ET in 21 patients (39%), PMF in 7 patients (13%), and unclassified MPN in 5 patients (9%). JAK2 V617F mutational status, available in 41 patients during MPN chronic phase, was positive in 24 patients (59%; Table 1). During MPN chronic phase, 50 of 54 patients had received cytoreductive therapy, whereas 4 patients had only received antiaggregating or anticoagulant therapy. Cytoreductive therapy had been hydroxyurea (HU) in 34 patients, pipobroman in 20 patients (11 of whom had also received HU before or after pipobroman; Table 2). Four patients with 4 PV had received radiophosphorus (P32), 2 patients with PV had received interferon alfa, and 3 patients with atypical CML had received imatinib mesylate.

Table 1

Patient characteristics and treatment results according to initial MPN

All patientsPVETPMFMPNuOverall PP (ET vs PV)
No. of patients 54 21 21   
Median age, y 68.5 66 71 73 71 NS NS 
JAK2 V617F positive in chronic phase, n/N (%) 24/41 (59) 12/14 (86) 9/16 (56) 2/7 (29) 1/4 (25) .023 NS 
Median time to transformation, mo 71 154 70 19 19 < .001 .004 
AML at transformation, n (%) 26 (48) 13 (62) 10 (48) 3 (43) NS NS 
MDS at transformation, n (%) 28 (52) 8 (38) 11 (52) 4 (57) 5 (100)   
Complex karyotype at transformation, n (%) 16 (30) 8 (44) 6 (33) 2 (33) NS NS 
Overall response to azacitidine, n (%) 28 (52) 7 (33) 15 (71) 4 (57) 2 (40) NS .016 
Restoration of chronic phase MPN features after azacitidine, n (%) 11 (20) 3 (14) 8 (38) NS NS 
Median survival from onset of azacitidine, mo 11 16 10 NS NS 
All patientsPVETPMFMPNuOverall PP (ET vs PV)
No. of patients 54 21 21   
Median age, y 68.5 66 71 73 71 NS NS 
JAK2 V617F positive in chronic phase, n/N (%) 24/41 (59) 12/14 (86) 9/16 (56) 2/7 (29) 1/4 (25) .023 NS 
Median time to transformation, mo 71 154 70 19 19 < .001 .004 
AML at transformation, n (%) 26 (48) 13 (62) 10 (48) 3 (43) NS NS 
MDS at transformation, n (%) 28 (52) 8 (38) 11 (52) 4 (57) 5 (100)   
Complex karyotype at transformation, n (%) 16 (30) 8 (44) 6 (33) 2 (33) NS NS 
Overall response to azacitidine, n (%) 28 (52) 7 (33) 15 (71) 4 (57) 2 (40) NS .016 
Restoration of chronic phase MPN features after azacitidine, n (%) 11 (20) 3 (14) 8 (38) NS NS 
Median survival from onset of azacitidine, mo 11 16 10 NS NS 

PV indicates polycythemia vera; ET, essential thrombocythemia; PMF, primary myelofibrosis; MPNu, myeloproliferative neoplasm unclassified; NS, not significant; JAK2, Janus kinase-2; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; and MPN, myeloproliferative neoplasm.

Table 2

Clinical characteristics and treatment of MPN before transformation

nThrombotic events, (%)*Antiaggregating therapy or anticoagulant therapy, n (%)*HU, n (%)*Median duration of HU, moPipobroman, n (%)*Median duration of pipobroman, moP32 or alkylating agent, n (%)Median time to progression, mo
ET 21 3 (14) 11 (52) 14 (66) 54 6 (29) 66  70 
PV 21 8 (38) 7 (33) 16 (76) 60 14 (66) 60 4 (19) 154 
PMF 1 (14) 1 (14) 1 (14)    19 
MPNu 1 (20) 2 (40) 3 (60) 12    19 
nThrombotic events, (%)*Antiaggregating therapy or anticoagulant therapy, n (%)*HU, n (%)*Median duration of HU, moPipobroman, n (%)*Median duration of pipobroman, moP32 or alkylating agent, n (%)Median time to progression, mo
ET 21 3 (14) 11 (52) 14 (66) 54 6 (29) 66  70 
PV 21 8 (38) 7 (33) 16 (76) 60 14 (66) 60 4 (19) 154 
PMF 1 (14) 1 (14) 1 (14)    19 
MPNu 1 (20) 2 (40) 3 (60) 12    19 

HU indicates hydroxyurea; ET, essential thrombocythemia; PV, polycythemia vera; PMF, primary myelofibrosis; and MPNu, myeloproliferative neoplasm unclassified;

*

During MPN phase.

Three of the patients with MPNu received imatinib.

Median time from diagnosis of MPN to MDS/AML was 71 months (range, 3-398 months) in the whole cohort, 89 months for patients with AML, and 58 months for patients with MDS (P = .6). Eight patients had already been treated after transformation by IC followed by allogeneic SCT in 3 patients, IC alone in 2 patients, and low-dose cytosine arabinoside in 3 patients. At the onset of azacytidine, 5 patients were refractory and 3 patients were in relapse after those treatments.

At inclusion in the azacitidine program, 26 patients (48%) had AML, including 9 with RAEB-t (17%), and 28 patients (52%) had MDS, including 9 (17%) with RAEB-1 and 19 (35%) with RAEB-2.

Eight patients (15%) had cytogenetic failure, 10 (19%) had normal karyotype, and 36 (67%) had 1 or several clonal abnormalities: 12 patients had 1 abnormality, including isolated −7/7q− in 5 patients and isolated +8 in 2 patients; 8 patients had 2 abnormalities; and 16 patients (30%) had a complex karyotype (≥ 3 abnormalities) that included −7/del 7q in 9 patients, and del 5q/−5 in 11 patients, with a median number of 5.5 abnormalities (range, 3-9 abnormalities). Chromosome 17 abnormalities were observed in 11 patients (del 17p in 8 patients and −17 in 3 patients). In MDS (n = 28), the International Prognostic Scoring System was int-1 in 2 patients (7%), int-2 in 12 patients (43%), and high in 10 patients (36%), whereas the remaining 4 patients with MDS had cytogenetic failure (Table 3). Four of the 5 unclassified MPN had normal karyotype (and 1 cytogenetic failure) during MPN phase and 3 had monosomy 7 at transformation. Finally, the 15 patients with JAK2-V617F tested at the time of transformation still had JAK2-V617F mutation, whereas the remaining 9 were not tested.

Table 3

Patient characteristics at transformation

AML (n = 26), n (%)MDS (n = 28), n (%)
Karyotype   
    Favorable*  
    Intermediate* 10 (38)  
    Unfavorable* 12 (46)  
    Failure 4 (15)  
    Good  5 (18) 
    Intermediate  4 (14) 
    Poor  15 (54) 
    Failure  4 (14) 
WHO classification   
    Marrow blasts > 30% 17 (65)  
    Marrow blasts 20%-30% (RAEB-t) 9 (35)  
    RAEB-1  9 (32) 
    RAEB-2  19 (68) 
IPSS   
    Low  
    Intermediate 1  2 (7) 
    Intermediate 2  12 (43) 
    High  10 (36) 
    NE  4 (14) 
AML (n = 26), n (%)MDS (n = 28), n (%)
Karyotype   
    Favorable*  
    Intermediate* 10 (38)  
    Unfavorable* 12 (46)  
    Failure 4 (15)  
    Good  5 (18) 
    Intermediate  4 (14) 
    Poor  15 (54) 
    Failure  4 (14) 
WHO classification   
    Marrow blasts > 30% 17 (65)  
    Marrow blasts 20%-30% (RAEB-t) 9 (35)  
    RAEB-1  9 (32) 
    RAEB-2  19 (68) 
IPSS   
    Low  
    Intermediate 1  2 (7) 
    Intermediate 2  12 (43) 
    High  10 (36) 
    NE  4 (14) 

AML indicates acute myeloid leukemia; MDS, myelodysplastic syndrome; WHO, World Health Organization; RAEB-t, refractory anemia with excess blasts in transformation; RAEB-1, refractory anemia with excess blasts 1; RAEB-2, refractory anemia with excess blasts 2; and NE, not evaluable.

*

Medical Research Council classification.

International Prognostic Scoring System classification.

Cytogenetic failure.

Response to azacitidine

The median number of cycles of azacitidine administered was 6 (range, 1-28 cycles). Thirty-five (65%) patients received azacitidine at the FDA/EMEA-approved schedule (75 mg/m2/d during 7 days every 28 days), whereas 19 patients (35%) received an attenuated dosing or schedule (100 mg/d during 7 days or 75 mg/m2/d during 5 days). The main reason to reduce the number of days to 5 per cycle was the difficulty for some centers to administer the drug during weekends, whereas the main reason to use a flat dose of 100 mg/d was patients' older age or frailty or both (including renal failure, observed in 5 cases). Thirteen patients (24%) received < 4 cycles of azacitidine because of early death in 9, restoration of MPN features in 2 patients, intercurrent illness in 1 patient, and 1 patient's decision 1; whereas 41 patients (76%) received ≥ 4 cycles.

As seen in Table 4, response was achieved in 28 of 54 patients (52%), including 13 CR (24%), 1 CRi (2%), 3 mCR (6%), 6 PR (11%), and 5 stable disease with hematologic improvement (9%; HI-E = 3 and HI-P = 2). Four of the 11 patients with baseline cytogenetic abnormality who achieved CR had cytogenetic evaluation at response, and 3 of those 4 patients achieved complete cytogenetic response.

Table 4

Response to azacitidine in MPN having progressed to MDS and AML

All (n = 54)WHO classification at progression
AML (n = 26)MDS (n = 28)
Responses    
    CR, n ( % ) 10 (19) 2 (8) 8 (29) 
    Cytogenetic CR, n (%) 3 (6) 1 (4) 2 (7) 
    CRi, n (%) 1 (2) 1 (4)  
    Marrow CR, n (%) 3 (6) * 3 (11) 
    PR, n (%) 6 (11) 6 (23)  
    Stable with HI, n (%) 5 (9) * 5 (21) 
    Stable without HI, n (%) 3 (6) * 3 (11) 
    Progression, n (%) 24 (44) 16 (62) 8 (29) 
Overall response rate, n (%) 28 (52)   
    CR + CRi + PR, n (%)  10 (38)  
    CR + marrow CR + PR + stable with HI, n (%)*   18 (64) 
All (n = 54)WHO classification at progression
AML (n = 26)MDS (n = 28)
Responses    
    CR, n ( % ) 10 (19) 2 (8) 8 (29) 
    Cytogenetic CR, n (%) 3 (6) 1 (4) 2 (7) 
    CRi, n (%) 1 (2) 1 (4)  
    Marrow CR, n (%) 3 (6) * 3 (11) 
    PR, n (%) 6 (11) 6 (23)  
    Stable with HI, n (%) 5 (9) * 5 (21) 
    Stable without HI, n (%) 3 (6) * 3 (11) 
    Progression, n (%) 24 (44) 16 (62) 8 (29) 
Overall response rate, n (%) 28 (52)   
    CR + CRi + PR, n (%)  10 (38)  
    CR + marrow CR + PR + stable with HI, n (%)*   18 (64) 

MPN indicates myeloproliferative neoplasm; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; WHO, World Health Organization; CR, complete response, CRi, complete response with incomplete recovery of cytopenias; PR, partial response; HI, hematologic improvement.

*

International Working Group 2006 response criteria for MDS.

International Working Group 2003 response criteria for AML.

With a median follow-up of 17 months after response evaluation, 23 of the 28 responders had relapsed after a median of 6 months (range, 1.5-22 months), and 6 patients were still responding after a median of 15 months (range, ≥10 to ≥ 34 months). Median response duration was 9 months. One of the complete responders received allogeneic SCT and was alive in CR after 14 months.

Prognostic factors of response

The underlying MPN was the only prognostic factor for overall response rate (ORR), with a higher ORR in patients with ET (71%) than for patients with PV (33%; P = .016; Table 1). In the 7 cases of PMF, 4 responses, including 1 CR, 1 PR, and 2 HI, were observed. Other pretreatment factors, including age, sex, white blood cell count, percentage of bone marrow blasts, JAK2 mutational status, and karyotype, were not significantly correlated with ORR. In AML, the ORR was 38%, including 12% CR, 4% CRi, and 23% PR. In MDS, the ORR was 64%, including 36% CR, 11% mCR, and 21% stable disease with HI. The comparison between ORR obtained in AML and MDS was difficult, because of somewhat different response criteria in those 2 categories (Table 4). When classifying patients with the FAB criteria, the ORR was 59% in MDS and 35% in AML (P = NS; details not shown).

Prognostic factors of CR achievement were the type of underlying MPN (14% CR for PV vs 43% for ET; P = .040) and WHO classification at transformation (12% CR in patients with AML vs 36% in MDS; P = .038; the comparison being possible because of similar CR criteria in AML and MDS). When adjusted on underlying MPN, WHO classification at transformation became even more significant (P = .007).

Prognostic factors of response duration were the presence of JAK2 V617F mutation (median of 11 months vs 5 months for JAK2 wild type; P = .005) and the recurrence of chronic phase features of MPN at response (median of 9 vs 5 months in the absence of such recurrence; P = .005). Other pretreatment characteristics were not prognostic of response duration. In particular, median response duration was 9 months in AML (range, 1.6 to ≥ 17 months) compared with 6 months in MDS (range, 1.2 to ≥ 34 months; P = NS).

Recurrence of chronic phase features of underlying MPN

A somewhat unexpected finding was that, in 11 patients (20%), corresponding to 39% of the responders, and including 8 of the 21 (38%) patients with initial ET and 3 of the 21 patients (14%) with initial PV (P = .080), response to azacitidine was associated with recurrence of chronic phase features of the initial MPN, ie, thrombocythemia in 8 and polycythemia in 3. This response was seen after 1-5 cycles (median, 3 cycles) and required restarting cytoreductive therapy with HU or interferon in 4 of them. No such evolution was observed in the 4 patients with PMF who responded. In 2 patients, recurrence of chronic phase MPN features lead to early discontinuation of azacitidine after 3 courses (Table 5). At the onset of azacitidine, 3 of the 11 patients with recurrence of chronic phase MPN features had AML and 8 had MDS. JAK2 V617F mutation was present during the chronic MPN phase in 5 of the 9 tested patients, during transformation in 3 of 5 tested patients, and still positive in both patients tested during the second chronic MPN phase.

Table 5

Baseline characteristics and their evolution in patients who restored chronic phase MPN features during azacytidine treatment

Patient no.Age, y*Initial MPNJAK2 statusTime to progression, yWHO*Marrow blasts, %*Hb level at inclusion, g/LPlt count at inclusion, × 109/LCytogenetics*ResponseHb level at evaluation, g/LPlt count at evaluation, × 109/LCytoreductive therapy
60 PV WT 7.8 AML 8§ 8.4 20 44,XY,add(1)(q2?), del(5) (q22q33), del(7) (q22q36), −16,−17,−20,+mar[14] CRi 17.1 300  
61 PV V617F 22.2 RAEB-2 10 8.0 70 46,XX, del (20)(q11) CR 12.1 900 IFN 
10 71 PV V617F 11 RAEB-2 17 8.8 147 46,XY[20] CR 17.0 325  
14 82 ET NA 8.9 AML 34 8.4 22 Failure HI 10.2 658  
16 75 ET NA 14.4 RAEB-2 12 10.1 478 47,XY, del(15)(q1?5q2?5), del(19)(p11), +21 CR 14.5 871 BU 
19 85 ET V617F 16 RAEB-2 11 9.5 258 47,XY,+8[1]/46,XY[6] nuc ish (D8Z1x3) [30%] CR 10.6 790 HU 
24 87 ET V617F 15.3 RAEB-1 9.6 48 Failure CR 15.9 485  
33 59 ET WT 2.3 RAEB-2 18 8.0 135 45,XY, del(20)(q12)[14] HI 14.4 641 HU 
44 61 ET NA 13.8 RAEB-1 9.1 82 47,XX,+8 CR 15.3 547  
48 81 ET V617F 2.4 RAEB-2 10 7.4 58 46,XX,del(5)(q13q33)[7]/46,sl,del(17)(p11)[9]/46,XX[3] CR 13.4 591 HU 
50 61 ET V617F 6.8 AML 26 10 128 46,XY,(1;6)(q31;q16)del(5) (q13q35),der(7;17) (p10;q10), +8,+8, der(12;18)(p11;q11), der(17)t(15;17)(q2?;q22)[20] CR 15.1 808  
Patient no.Age, y*Initial MPNJAK2 statusTime to progression, yWHO*Marrow blasts, %*Hb level at inclusion, g/LPlt count at inclusion, × 109/LCytogenetics*ResponseHb level at evaluation, g/LPlt count at evaluation, × 109/LCytoreductive therapy
60 PV WT 7.8 AML 8§ 8.4 20 44,XY,add(1)(q2?), del(5) (q22q33), del(7) (q22q36), −16,−17,−20,+mar[14] CRi 17.1 300  
61 PV V617F 22.2 RAEB-2 10 8.0 70 46,XX, del (20)(q11) CR 12.1 900 IFN 
10 71 PV V617F 11 RAEB-2 17 8.8 147 46,XY[20] CR 17.0 325  
14 82 ET NA 8.9 AML 34 8.4 22 Failure HI 10.2 658  
16 75 ET NA 14.4 RAEB-2 12 10.1 478 47,XY, del(15)(q1?5q2?5), del(19)(p11), +21 CR 14.5 871 BU 
19 85 ET V617F 16 RAEB-2 11 9.5 258 47,XY,+8[1]/46,XY[6] nuc ish (D8Z1x3) [30%] CR 10.6 790 HU 
24 87 ET V617F 15.3 RAEB-1 9.6 48 Failure CR 15.9 485  
33 59 ET WT 2.3 RAEB-2 18 8.0 135 45,XY, del(20)(q12)[14] HI 14.4 641 HU 
44 61 ET NA 13.8 RAEB-1 9.1 82 47,XX,+8 CR 15.3 547  
48 81 ET V617F 2.4 RAEB-2 10 7.4 58 46,XX,del(5)(q13q33)[7]/46,sl,del(17)(p11)[9]/46,XX[3] CR 13.4 591 HU 
50 61 ET V617F 6.8 AML 26 10 128 46,XY,(1;6)(q31;q16)del(5) (q13q35),der(7;17) (p10;q10), +8,+8, der(12;18)(p11;q11), der(17)t(15;17)(q2?;q22)[20] CR 15.1 808  

MPN indicates myeloproliferative neoplasm; JAK2, Janus kinase-2; WHO, World Health Organization; Hb, hemoglobin; Plt, platelet, PV, polycythemia vera; WT, wild type; AML, acute myeloid leukemia; CRi, complete response with incomplete recovery of cytopenias; RAEB-2, refractory anemia with excess blasts 2; CR, complete response; IFN, interferon; ET, essential thrombocythemia; NA, not assessed; AML, acute myeloid leukemia; HI, hematologic improvement; BU, oral busulfan; HU, hydroxyurea; and RAEB-1, refractory anemia with excess blasts 1.

*

At inclusion.

At response.

After recurrence of chronic phase MPN features.

§

This patient was classified as AML on the basis of 31% circulating blasts.

According to IWG response criteria, 9 of the 11 patients with recurrence of chronic phase MPN features had achieved CR (including 2 cytogenetic CR) and 2 HI (patients 14 and 33 in Table 3). By univariate analysis, no prognostic factor of recurrence of chronic phase MPN features (including age, sex, marrow blast percentage, white blood cell count, platelet count, JAK 2 mutational status, previous MPN type) was found in the whole patient population and in responders to azacytidine. Nine of the 11 patients relapsed to AML/MDS after a median time of 8.6 months (range, 1.6-22 months), whereas 2 remained responders: 1 after ≥ 33 months, treated by interferon and 1 who received an allograft (with reduced-intensity conditioning regimen) in CR after 4 cycles of azacitidine, still alive in CR 14 months after transplantation.

Overall survival

With a median follow-up of 20 months (range, 3 to ≥ 39 months) from onset of azacitidine, 44 patients (81%) had died, 30 (68%) from overt disease progression, 5 (11%) from bleeding (including 3 during azacitidine treatment), 8 (18%) from sepsis (including 1 during azacitidine treatment), 1 (4%) from thrombosis without recurrence of PV, and 1 (4%) from cardiac failure. Ten patients (19%) were still alive after ≥ 3 to ≥ 39 months, including 4 patients in CR and 2 in persisting HI.

Median OS was 11 months (Figure 1), 8 months in AML and 14 months in MDS (P = NS). According to the FAB classification, median OS was 8 months in AML and 13 months in MDS (P = NS).

Figure 1

Overall survival.

Figure 1

Overall survival.

Close modal

OS did not significantly differ between patients with ET and PV as initial MPN (median,16 vs 7 months, respectively; P = .150; Figure 2). When adjusted on initial MPN (ET vs PV), OS, however, became significantly better for patients with MDS than for patients with AML (P = .01). In patients with initial PMF, median survival was 8 months. Other pretreatment characteristics including age, sex, platelet count, circulating blast count, Hb level, percentage of bone marrow blasts karyotype, and JAK2 mutational status were not predictive of OS. In multivariate analysis, adverse cytogenetics (including complex karyotype, −7 or del 7q) and higher marrow blast percentage (as continuous variable) were associated with worse OS (P = .03 for both parameters).

Figure 2

Survival in ET versus PV. The tick marks represent censored patients.

Figure 2

Survival in ET versus PV. The tick marks represent censored patients.

Close modal

Side effects of treatment

Grade 3-4 hematologic toxicities were the main side effects of azacitidine, mainly during the first 4 cycles of azacitidine: 23 patients (43%) and 3 patients (6%) had infection and bleeding requiring hospitalization, with a fatal outcome in 4 and 2 cases, respectively. Nonhematologic side effects included grade 3 skin rash in 1 patient, leading to early azacitidine discontinuation. Grade 1-2 nonhematologic side effects mainly included diarrhea, constipation, and local side effects at injection sites, as previously described for azacitidine.11,12 

This is, to our knowledge, the first report of a relatively large cohort of patients with Ph-negative MPN treated by a hypomethylating agent after having progressed to MDS/AML. We observed a 64% response rate to azacitidine in patients who had progressed to MDS and 38% in patients who had progressed to AML. The response rate was significantly higher in patients with ET than for patients with PV as initial MPN and, in close to 40% of the responders, response was associated with recurrence of chronic phase MPN features (thrombocytosis in ET and erythrocytosis in PV; Table 5).

The response rate to azacitidine observed in this series of MDS secondary to MPN was similar to that observed in MDS as a whole,9,10  although the CR rate was somewhat higher in the present report, whereas response duration was somewhat shorter (median of 9 months compared with 13.6 months in AZA 001 study and 21 months in CALGB 9221 study, but the last study included a high proportion of lower risk MDS).

The only significant prognostic factor for ORR was the type of underlying MPN. Indeed, patients evolving from ET responded better than patients evolving from PV while in transformed PMF; the number of patients was small to conclude (4 of the 7 patients responded). Patients with MDS at transformation and patients with ET as primary MPN had significantly higher CR rates than patients with AML at transformation and PV as primary MPN, respectively. In agreement with previous studies of azacitidine in MDS,10,23  complex karyotype was not a poor predictive factor for response. This is particularly important to consider in transformations of MPN, which often carry complex chromosomal abnormalities.

The response rates and survival we observed appeared better than those generally reported with other treatments in MDS/AML occurring during the course of MPN. Indeed, ≥ 4 series, totaling ∼ 200 AML/MDS after MPN treated with low-dose chemotherapy, IC or best supportive care only have been reported,5,24-26  all of them with poor results (Table 6). In particular, IC gave a CR rate of 46%, but remissions were short, and median progression-free survival was only 5 months in the M.D. Anderson study.26  In the absence of subsequent allogeneic SCT, all previously reported remissions were short, whereas median response duration in our series was somewhat longer, 9 months, after a median follow-up of 20 months. Moreover, the median survival of 11 months observed in our study was somewhat higher than in those series, where it ranged from 2.6 to 5 months with chemotherapy or supportive care.5,25,26  Interestingly, decitabine, another demethylating agent, also provided better survival than high-dose chemotherapy in blastic phase of CML.27,28 

Table 6

Outcome of MPN after MDS/AML transformation in literature series

StudySample sizeTreatmentCR/CRi, %Median survival with specific treatment, moOverall median survival, mo
Cervantes et al, 199124  13 Not detailed   3.0 
Passamonti, 20055  23 Best supportive care (n = 7)  2.5 2.9 
Low dose Ara-C (n = 4)    
6-Thioguanine (n = 4)    
Intensive chemotherapy(n = 8) 13 5.6  
Mesa et al, 200525  91 Best supportive care (n = 48)  2.0 2.6 
Oral busulfan/etoposide (n = 4) 2.9  
Vindesine (n = 9)    
Other (n = 6)    
Intensive chemotherapy (n = 24) 3.9  
Tam et al, 200826  74 Best supportive care (n = 19)   5.0 
Gemtuzumab (n = 4) 6.0  
Azacytidine (n = 3)    
Dasatinib (n = 2)    
Other (n = 3)    
Intensive chemotherapy (n = 41) 46 7.0  
StudySample sizeTreatmentCR/CRi, %Median survival with specific treatment, moOverall median survival, mo
Cervantes et al, 199124  13 Not detailed   3.0 
Passamonti, 20055  23 Best supportive care (n = 7)  2.5 2.9 
Low dose Ara-C (n = 4)    
6-Thioguanine (n = 4)    
Intensive chemotherapy(n = 8) 13 5.6  
Mesa et al, 200525  91 Best supportive care (n = 48)  2.0 2.6 
Oral busulfan/etoposide (n = 4) 2.9  
Vindesine (n = 9)    
Other (n = 6)    
Intensive chemotherapy (n = 24) 3.9  
Tam et al, 200826  74 Best supportive care (n = 19)   5.0 
Gemtuzumab (n = 4) 6.0  
Azacytidine (n = 3)    
Dasatinib (n = 2)    
Other (n = 3)    
Intensive chemotherapy (n = 41) 46 7.0  

A total of 201 patients were evaluated.

MPN indicates myeloproliferative neoplasm; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; CR, complete response; CRi, complete response with incomplete recovery of cytopenias; and Ara-C, cytosine arabinoside.

In our study, 4 of the 7 cases of transformed PMF responded, and their median survival was 8 months. Those findings in patients with blastic rather than fibrotic progression of MPN do not contradict the disappointing results obtained with hypomethylating agents in primary or secondary myelofibrosis.11,12 

Only 1 patient received allogeneic SCT after azacitidine, a small number largely related to the median age of our cohort (69.5 years), making difficult to evaluate this procedure after azacitidine.

Importantly, we observed in 39% of the responders reappearance of features of chronic phase MPN, including thrombocytosis in patients with ET and erythrocytosis in patients with PV. Those responses tended to occur early after treatment onset. To our knowledge, this type of evolution had only been reported in blast crisis of CML treated with IC or decitabine.28  In Ph-negative MPN having progressed to MDS/AML and treated successfully with chemotherapy, marrow histologic features of chronic phase MPN or of a JAK2 V617F clone have been reported25  but not to our knowledge recurrence of thrombocytosis or erythrocytosis. In 24 cases of leukemic transformation of PMF who received AML-like IC at the Mayo Clinic, 10 had after treatment bone marrow biopsy findings of chronic phase disease without increase of blast percentage.25  As in our experience, patients who reverted to chronic phase disease finally relapsed in the absence of allogeneic SCT. Those findings suggest that azacitidine may in some patients with AML/MDS transformation of MPN reverse the disease course to a chronic phase rather than suppress the MPN clone. A similar mechanism of action of azacitidine has also been suggested in de novo MDS, and it could implicate gene hypomethylation.29,30  Blastic progression of Ph-negative MPN is multifactorial but may indeed involve aberrant methylation of tumor suppressor genes,31  including the negative regulators of JAK– signal transducer and activator of transcription (STAT) signaling, suppressor of cytokine signaling-1 (SOCS-1), and suppressor of cytokine signaling-3 (SOCS-3).8,32,33  Such epigenetic deregulation may occur early during MPN progression, as for example, methylation of the CXCR4 chemokine promoter recently involved in the constitutive mobilization of CD34+ in PMF and reversed by azacitidine in vitro.34  Likewise, JAK2 was recently reported to directly interfere with chromatin assembly in the nucleus.35 

Altogether, our results suggest that azacitidine can induce a substantial response rate with relatively low toxicity in Ph-negative MPN having progressed to AML/MDS.26  Most responses are however short, and consolidation treatments, particularly allogeneic SCT or combination with other agents, are required in those patients for whom prognosis remains very poor.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank investigators of GFM group (Groupe Francophone des Myelodysplasies) who enrolled patients in this study and the French health agency (AFSSAPS).

This work was supported in part by an unrestricted research grant from Celgene to the GFM group.

Contribution: S.T. and R.I. collected the data and wrote the manuscript; V.S. performed statistical analysis; L.A. and P.F. designed the study and wrote the manuscript; and E.R., B.Q., Y.C., L. Sorin, F.D., T.C., J.D., L. Sanhes, V.E., C.D., P.T., S.H., C.S., J.-J.K., P.F., and L.A. enrolled the patients and collected the data.

Conflict-of-interest disclosure: P.F. has received research funding from Celgene and Amgen. The remaining authors declare no competing financial interests.

Correspondence: Lionel Ades, AP-HP, Hôpital Avicenne, Service d'Hématologie Clinique, Paris 13 University, 125 rue de Stalingrad, 93009 Bobigny, France; e-mail: lionel.ades@avc.aphp.fr.

1
Berk
 
PD
Goldberg
 
JD
Silverstein
 
MN
, et al. 
Increased incidence of acute leukemia in polycythemia vera associated with chlorambucil therapy.
N Engl J Med
1981
, vol. 
304
 
8
(pg. 
441
-
447
)
2
Finazzi
 
G
Caruso
 
V
Marchioli
 
R
, et al. 
Acute leukemia in polycythemia vera: an analysis of 1638 patients enrolled in a prospective observational study.
Blood
2005
, vol. 
105
 
7
(pg. 
2664
-
2670
)
3
Kiladjian
 
JJ
Rain
 
JD
Bernard
 
JF
Briere
 
J
Chomienne
 
C
Fenaux
 
P
Long-term incidence of hematological evolution in three French prospective studies of hydroxyurea and pipobroman in polycythemia vera and essential thrombocythemia.
Semin Thromb Hemost
2006
, vol. 
32
 
4 Pt 2
(pg. 
417
-
421
)
4
Kiladjian
 
J-J
Chevret
 
S
Abgrall
 
J-F
Chait
 
Y
Briere
 
J
Risk stratification for survival and clonal progression in essential thrombocythemia (ET): result of a prospective study of 108 patients with very long term follow up [abstract].
Blood
2008
, vol. 
112
 
11
 
Abstract 1747
5
Passamonti
 
F
Rumi
 
E
Arcaini
 
L
, et al. 
Leukemic transformation of polycythemia vera: a single center study of 23 patients.
Cancer
2005
, vol. 
104
 
5
(pg. 
1032
-
1036
)
6
Tam
 
CS
Kantarjian
 
H
Cortes
 
J
, et al. 
Dynamic model for predicting death within 12 months in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis.
J Clin Oncol
2009
, vol. 
27
 
33
(pg. 
5587
-
5593
)
7
Jiang
 
Y
Dunbar
 
A
Gondek
 
LP
, et al. 
Aberrant DNA methylation is a dominant mechanism in MDS progression to AML.
Blood
2009
, vol. 
113
 
6
(pg. 
1315
-
1325
)
8
Wang
 
JC
Chen
 
W
Nallusamy
 
S
Chen
 
C
Novetsky
 
AD
Hypermethylation of the P15INK4b and P16INK4a in agnogenic myeloid metaplasia (AMM) and AMM in leukaemic transformation.
Br J Haematol
2002
, vol. 
116
 
3
(pg. 
582
-
586
)
9
Silverman
 
LR
Demakos
 
EP
Peterson
 
BL
, et al. 
Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B.
J Clin Oncol
2002
, vol. 
20
 
10
(pg. 
2429
-
2440
)
10
Fenaux
 
P
Mufti
 
GJ
Hellstrom-Lindberg
 
E
, et al. 
Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study.
Lancet Oncol
2009
, vol. 
10
 
3
(pg. 
223
-
232
)
11
Quintas-Cardama
 
A
Tong
 
W
Kantarjian
 
H
, et al. 
A phase II study of 5-azacitidine for patients with primary and post-essential thrombocythemia/polycythemia vera myelofibrosis.
Leukemia
2008
, vol. 
22
 
5
(pg. 
965
-
970
)
12
Mesa
 
RA
Verstovsek
 
S
Rivera
 
C
, et al. 
5-Azacitidine has limited therapeutic activity in myelofibrosis.
Leukemia
2009
, vol. 
23
 
1
(pg. 
180
-
182
)
13
Vardiman
 
JW
Thiele
 
J
Arber
 
DA
, et al. 
The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes.
Blood
2009
, vol. 
114
 
5
(pg. 
937
-
951
)
14
Bennett
 
JM
Catovsky
 
D
Daniel
 
MT
, et al. 
Proposals for the classification of the myelodysplastic syndromes.
Br J Haematol
1982
, vol. 
51
 
2
(pg. 
189
-
199
)
15
Laszlo
 
J
Myeloproliferative disorders (MPD): myelofibrosis, myelosclerosis, extramedullary hematopoiesis, undifferentiated MPD, and hemorrhagic thrombocythemia.
Semin Hematol
1975
, vol. 
12
 
4
(pg. 
409
-
432
)
16
Berlin
 
NI
Diagnosis and classification of the polycythemias.
Semin Hematol
1975
, vol. 
12
 
4
(pg. 
339
-
351
)
17
Vardiman
 
JW
Harris
 
NL
Brunning
 
RD
The World Health Organization (WHO) classification of the myeloid neoplasms.
Blood
2002
, vol. 
100
 
7
(pg. 
2292
-
2302
)
18
Cheson
 
BD
Greenberg
 
PL
Bennett
 
JM
, et al. 
Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia.
Blood
2006
, vol. 
108
 
2
(pg. 
419
-
425
)
19
Cheson
 
BD
Bennett
 
JM
Kopecky
 
KJ
, et al. 
Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia.
J Clin Oncol
2003
, vol. 
21
 
24
(pg. 
4642
-
4649
)
20
Tam
 
CS
Kantarjian
 
H
Cortes
 
J
, et al. 
Dynamic model for predicting death within 12 months in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis.
J Clin Oncol
2009
, vol. 
27
 
33
(pg. 
5587
-
5593
)
21
Kaplan
 
E
Meier
 
P
Non parametric estimation from incomplete observation.
J Am Stat Assos
1958
, vol. 
53
 pg. 
457
 
22
Cox
 
D
Regression models and life-tables (with discussion).
J R Stat Soc B
1972
, vol. 
34
 pg. 
487
 
23
Kantarjian
 
H
Issa
 
JP
Rosenfeld
 
CS
, et al. 
Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study.
Cancer
2006
, vol. 
106
 
8
(pg. 
1794
-
1803
)
24
Cervantes
 
F
Tassies
 
D
Salgado
 
C
Rovira
 
M
Pereira
 
A
Rozman
 
C
Acute transformation in nonleukemic chronic myeloproliferative disorders: actuarial probability and main characteristics in a series of 218 patients.
Acta Haematol
1991
, vol. 
85
 
3
(pg. 
124
-
127
)
25
Mesa
 
RA
Li
 
CY
Ketterling
 
RP
Schroeder
 
GS
Knudson
 
RA
Tefferi
 
A
Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases.
Blood
2005
, vol. 
105
 
3
(pg. 
973
-
977
)
26
Tam
 
CS
Nussenzveig
 
RM
Popat
 
U
, et al. 
The natural history and treatment outcome of blast phase BCR-ABL- myeloproliferative neoplasms.
Blood
2008
, vol. 
112
 
5
(pg. 
1628
-
1637
)
27
Kantarjian
 
HM
O'Brien
 
SM
Keating
 
M
, et al. 
Results of decitabine therapy in the accelerated and blastic phases of chronic myelogenous leukemia.
Leukemia
1997
, vol. 
11
 
10
(pg. 
1617
-
1620
)
28
Sacchi
 
S
Kantarjian
 
HM
O'Brien
 
S
, et al. 
Chronic myelogenous leukemia in nonlymphoid blastic phase: analysis of the results of first salvage therapy with three different treatment approaches for 162 patients.
Cancer
1999
, vol. 
86
 
12
(pg. 
2632
-
2641
)
29
Baylin
 
SB
Ohm
 
JE
Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction?
Nat Rev Cancer
2006
, vol. 
6
 
2
(pg. 
107
-
116
)
30
Yu
 
L
Liu
 
C
Vandeusen
 
J
, et al. 
Global assessment of promoter methylation in a mouse model of cancer identifies ID4 as a putative tumor-suppressor gene in human leukemia.
Nat Genet
2005
, vol. 
37
 
3
(pg. 
265
-
274
)
31
Capello
 
D
Deambrogi
 
C
Rossi
 
D
, et al. 
Epigenetic inactivation of suppressors of cytokine signalling in Philadelphia-negative chronic myeloproliferative disorders.
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
504
-
511
)
32
Jost
 
E
do
 
ON
Dahl
 
E
, et al. 
Epigenetic alterations complement mutation of JAK2 tyrosine kinase in patients with BCR/ABL-negative myeloproliferative disorders.
Leukemia
2007
, vol. 
21
 
3
(pg. 
505
-
510
)
33
Fourouclas
 
N
Li
 
J
Gilby
 
DC
, et al. 
Methylation of the suppressor of cytokine signaling 3 gene (SOCS3) in myeloproliferative disorders.
Haematologica
2008
, vol. 
93
 
11
(pg. 
1635
-
1644
)
34
Bogani
 
C
Ponziani
 
V
Guglielmelli
 
P
, et al. 
Hypermethylation of CXCR4 promoter in CD34+ cells from patients with primary myelofibrosis.
Stem Cells
2008
, vol. 
26
 
8
(pg. 
1920
-
1930
)
35
Dawson
 
MA
Bannister
 
AJ
Gottgens
 
B
, et al. 
JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin.
Nature
2009
, vol. 
461
 
7265
(pg. 
819
-
822
)

Author notes

*

S.T. and R.I. contributed equally to this study.

Sign in via your Institution