The use of all-trans retinoic acid (ATRA) in combination with chemotherapy has markedly improved the prognosis for patients with acute promyelocytic leukemia (APL); the higher complete remission (CR) and survival rates now reported in this disease almost approach those obtained for other highly curable hematologic malignancies. Of 77 patients with APL who were consecutively treated at a single institution and who achieved CR after induction and consolidation therapy, 5 (6.5%) acquired therapy-related myelodysplasia (tMDS), acute myelogenous leukemia (AML), or both (tMDS–AML). Of these, 3 of 46 (6.5%) patients received front-line chemotherapy with or without ATRA and acquired tMDS–AML while in first remission of APL. Two underwent repeated chemotherapy cycles with ATRA because of APL relapse and acquired tMDS–AML while in the second or third remission of APL. In 2 patients, clinical and biologic characteristics of tMDS–AML were as expected for postalkylating forms (long latency, MDS phase preceding AML, karyotypic aberrations involving chromosomes 5 or 7), even though one of them had not previously received alkylating drugs. Three of the 5 patients died shortly after tMDS–AML diagnosis, one is alive with tMDS, and one is alive and in CR after allogeneic bone marrow transplantation. The occurrence of tMDS–AML after successful therapy for APL is an emerging problem. The availability of prognostic score systems at initial diagnosis and monitoring of residual disease by polymerase chain reaction might allow better tailoring of treatment intensity in APL to spare unnecessary toxicity and to minimize the risk for tMDS–AML in patients who are presumably cured.

Acute promyelocytic leukemia (APL) is a subtype of acute myelogenous leukemia (AML) that is characterized by peculiar clinical and biologic features. These include severe hemorrhagic diathesis at presentation, specific chromosome translocation t(15;17) resulting in the fusion of promyelocytic (PML) and retinoic acid receptor α (RARα) genes, and unique in vitro and in vivo responses to the differentiating agent all-transretinoic acid (ATRA).1-3 Front-line use of ATRA combined with chemotherapy has recently contributed remarkable improvement in the prognostic outlook of APL, converting this once frequently fatal leukemia to a highly curable disease.4-12 

The development of therapy-related myelodysplasia or AML (tMDS–AML) after treatment for other tumors is one of the most serious complications occurring after chemotherapy for highly curable malignancies such as breast cancer, Hodgkin disease, non-Hodgkin lymphoma, and childhood acute lymphoblastic leukemia.13-15Among chemotherapy agents, alkylating drugs and topoisomerase II inhibitors such as anthracyclines and epipodophillotoxins have been frequently associated with the development of tMDS–AML. Regarding tMDS–AML occurring after treatment for APL, sporadic cases have been reported to date,16-24 but no studies have investigated this issue by analyzing large series of patients. We report here our experience with the development of tMDS–APL in a consecutive group of 77 patients with APL treated at a single institution.

Patients

Eighty-eight patients with APL consecutively diagnosed and treated at the Department of Cellular Biotechnology and Hematology of the University La Sapienza of Roma from January 1989 to September 1998 are included in this analysis. A minimum follow-up of 2 years after completion of induction therapy was considered for enrollment into the study. Diagnoses of APL were initially established by morphologic and cytochemical criteria following the French-American-British (FAB) guidelines25 and were confirmed in all patients by Southern blot analysis, reverse transcription–polymerase chain reaction (RT-PCR), or both using specific primers and probes as described.26,27 

Therapy for acute promyelocytic leukemia

Two consecutive protocols were used.

GIMEMA 0389.

Twenty-eight patients who received diagnoses from January 1989 to March 1993 were randomly assigned to undergo induction treatment with idarubicin (IDA) alone (10 mg/m2 for 4 days) versus IDA at the same dosage plus cytarabine (ARA-C) (200 mg/m2 continuous infusion [c.i.] for 7 days).28 Patients in complete remission (CR) were administered 3 polychemotherapy consolidation courses as reported.4,28 At the end of consolidation, patients in CR were randomly assigned to undergo maintenance therapy with methotrexate (MTX, 15 mg/m2 per week) and 6-mercaptopurine (6-MP, 90 mg/m2 per day) for 2 years versus no further therapy.

AIDA protocol.

Sixty patients given diagnoses from April 1993 to December 1998 underwent the AIDA regimen as reported.4 

Follow-up studies

Patients were monitored at regular time intervals after the end of consolidation therapy. Bone marrow samples were collected every 3 to 4 months and were analyzed by RT-PCR for PML–RARα amplification as reported elsewhere.4,26 The diagnosis of tMDS–AML was established according to the FAB criteria.29 

Cytogenetic analysis

Karyotypic analyses were carried out in marrow samples collected at the time of evolution in tMDS–tAML in all patients using direct technique and short-term culture (24 hours). The GTG banding method was used, and karyotypes were defined according to standard nomenclature.

Of 88 consecutive patients with newly diagnosed APL, 8 died during induction, 2 died during consolidation therapy, one did not achieve molecular remission at the end of consolidation, and the remaining 77 (87.5%) obtained hematologic and molecular remission after induction and consolidation therapy. Five patients (3 of 53 or 5.6% in the AIDA 0493 study and 2 of 24 or 8.3% in the GIMEMA 0389 study) acquired tMDS–AML during follow-up. Initial clinical features and therapies for APL in these 5 patients are reported in Table1. Patient 1 had been reported previously.30 Three patients (patients 1-3) were in first CR when tMDS–AML was diagnosed, whereas 2 patients (patients 4-5) received further treatment for APL relapse before tMDS–AML developed, including alkylating agents as part of the conditioning regimen before autologous stem cell transplantation (AuSCT). Of the 2 latter patients, patient 4 received a diagnosis of tMDS–AML when in second CR, and patient 5 acquired tMDS–AML while in third CR. In all 5 patients, RT-PCR monitoring indicated molecular remission (ie negativity of the PML–RARα test) at the time of tMDS–AML diagnosis.

Table 1.

Main characteristics of patients at APL diagnosis and treatments administered

Patient no.Sex/age, yWBC (× 109/L)Platelets (× 109/L)PML/RARα IsoformFront-line therapy*Maintenance treatmentAPL relapseSalvage therapy for APL relapse
F/16 1.4 185 ND GIMEMA 0389 MTX, 6-MP No No 
F/52 1.5 12 BCR1 AIDA ATRA plus MTX, 6-MP No No  
M/63 6.3 99 BCR1 AIDA ATRA plus MTX, 6-MP No No  
M/35 43.7 30 ND GIMEMA 0389 MTX, 6-MP First ATRA + CHT + AuSCT 
F/31 19.5 210 BCR3 AIDA MTX, 6-MP First ATRA + CHT + AuSCT 
       Second ATRA + CHT 
Patient no.Sex/age, yWBC (× 109/L)Platelets (× 109/L)PML/RARα IsoformFront-line therapy*Maintenance treatmentAPL relapseSalvage therapy for APL relapse
F/16 1.4 185 ND GIMEMA 0389 MTX, 6-MP No No 
F/52 1.5 12 BCR1 AIDA ATRA plus MTX, 6-MP No No  
M/63 6.3 99 BCR1 AIDA ATRA plus MTX, 6-MP No No  
M/35 43.7 30 ND GIMEMA 0389 MTX, 6-MP First ATRA + CHT + AuSCT 
F/31 19.5 210 BCR3 AIDA MTX, 6-MP First ATRA + CHT + AuSCT 
       Second ATRA + CHT 

ND indicates not determined.

*

As determined by RT-PCR. See “Patients, materials, and methods” for front-line treatment protocols GIMEMA 0389 and AIDA.

Chemotherapy for APL relapse included mitoxantrone and cytarabine.

Southern blot analysis was used in these 2 patients to identify RARα and PML gene rearrangement.

The main morphologic and karyotypic features of the tMDS–AML phase—time latency between APL and tMDS–AML diagnosis, treatment received for tMDS–AML, and patient outcomes—are reported in Table2. In all patients, progressive pancytopenia was detected before diagnosis of tMDS–AML. A 2-month phase of tMDS preceded tAML diagnosis in patients 3 and 4. In patient 1, trilineage myelodysplasia was diagnosed concomitantly with tAML (FAB M4). Evolution into tAML was not detected in patients 3 and 5 (the latter underwent allogeneic SCT shortly after MDS diagnosis). Cytogenetic characterization revealed numeric abnormalities involving chromosome 5 or 7 in 2 patients (patients 2, 4), balanced t(10;11)(p14;q21) in patient 1, and a normal karyotype in patient 5, whereas it failed because of lack of evaluable metaphases in patient 3. In patient 1, the involvement of the MLL gene was ruled out by Southern blot analysis, as reported elsewhere.30 

Table 2.

Clinical and biologic characteristics of the tMDS-AML phase and treatment outcome

Patient no.Latency, moPML/RARα status*tMDS phase (mo)KaryotypetMDS-AML treatmentOutcome
48 Negative No t(10;11)(p14;q21) MTZ/AraC/VP16 allogeneic SCT Died of GVHD (day + 50) 
43 Negative Yes (+ 18) Monosomy 7 Supportive care Alive in tMDS (at 18 mo)  
46 Negative Yes (2) Failure Supportive care Died of disease progression (1 mo) 
48 Negative Yes (2) Del(5q−) Supportive care Died of disease  
 (33 from second CR)      progression (5 mo) 
24 Negative Yes (5) 46xx Allogeneic SCT Alive in CR  
 (2 from second CR)      (+ 12 mo from SCT) 
Patient no.Latency, moPML/RARα status*tMDS phase (mo)KaryotypetMDS-AML treatmentOutcome
48 Negative No t(10;11)(p14;q21) MTZ/AraC/VP16 allogeneic SCT Died of GVHD (day + 50) 
43 Negative Yes (+ 18) Monosomy 7 Supportive care Alive in tMDS (at 18 mo)  
46 Negative Yes (2) Failure Supportive care Died of disease progression (1 mo) 
48 Negative Yes (2) Del(5q−) Supportive care Died of disease  
 (33 from second CR)      progression (5 mo) 
24 Negative Yes (5) 46xx Allogeneic SCT Alive in CR  
 (2 from second CR)      (+ 12 mo from SCT) 
*

By RT-PCR with 10−4 sensitivity.

In light of poor performance status, 3 patients (patients 2-4) received only supportive care as therapy for tMDS. Of these, one is alive with tMDS 18 months after diagnosis of tMDS, and 2 died of progressive disease shortly after tMDS development. Patient 1 underwent reinduction and consolidation therapy followed by allogeneic SCT and died on day +50 from hepatic GvHD. Finally, patient 5, who acquired MDS in third CR, received supportive care for 5 months and then underwent allogeneic SCT. She is alive and in CR from APL or MDS 12 months after SCT.

Since the advent of ATRA, APL is increasingly reported as curable.4-12 Thus, larger numbers of long-term survivors of this disease are expected in the near future, and, as a consequence, more patients will be at risk for late complications related to antileukemic treatment. In fact, with few exceptions,31conventional chemotherapy is still part of the protocol used in the front-line therapy for the disease. Furthermore, chemotherapy is considered essential to obtain sustained molecular remission, which in turn correlates with prolonged survival and potential cure.1-3 

Current APL chemotherapy protocols usually include high-dose anthracyclines, mitoxantrone, and epipodophillotoxins—in other words, topoisomerase II inhibitors whose leukemogenic potential is well established.32 All these agents were administered as part of the initial treatment in the 2 protocols reported in our study. In addition, 2 of the 5 patients described received further treatment with alkylating agents because of APL relapse. As reported in other tAML studies,13-15 evolution from tMDS to overt tAML was rapid in 2 patients. However, we also observed a patient (patient 2) with a prolonged and indolent tMDS phase that lasted more than 18 months despite the fact that the tMDS clone harbored a poor prognostic aberration (monosomy 7). Interestingly, such a lesion would have led to a diagnosis of alkylating agent–related tMDS according to the World Health Organization (WHO) classification,33 yet this patient was never administered alkylating drugs. The latter finding is in agreement with a recent report by Au et al,24 who described a patient with APL treated without alkylating agents who acquired tMDS with chromosome 5 and 7 abnormalities. Together, these observations suggest that the pathobiologic associations proposed by the WHO to distinguish epipodophillotoxin-related from alkylating-related tMDS may not always hold true. Other clinical and biologic features of patients in our study were similar to those usually reported for tMDS–AML, with a median time latency of 46 months and evidence of unexplained pancytopenia and macrocytosis preceding tMDS diagnosis.

Three patients in the current study acquired tMDS while in prolonged first hematologic and molecular remission; 2 of them died, one of disease progression and the other of transplant-related toxicity. It is conceivable that these patients were cured of APL. Because studies have also suggested that a relevant proportion of patients with newly diagnosed APL are overtreated,9,31,34 we emphasize the need for better tailoring of treatment intensity in this disease by identifying risk categories at diagnosis and by prospective minimal residual disease monitoring. Besides identifying patients at risk for relapse and in need of further treatment, the use of RT-PCR of PML–RARα during remission might spare the development of unnecessary toxicity in potentially cured patients.

Supported by RomAIL–ONLUS, Associazione Italiana Ricerca sul Cancro, and Ministero dell'Università e della Ricerca Scientifica e Tecnologica.

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 U.S.C. section 1734.

1
Fenaux
P
Chomienne
C
Degos
A
Acute promyelocytic leukemia: biology and treatment.
Semin Oncol.
24
1997
92
102
2
Frankel
SR
Powell
BL
Current approaches to acute promyelocytic leukemia.
Diagnostic and therapeutic advances in hematologic malignancies.
Tallman
MS
Gordon
LI
1999
125
153
Kluwer Academic Publishers
Boston, MA
3
Lo Coco
F
Nervi
C
Avvisati
G
Mandelli
F
Acute promyelocytic leukemia: a curable disease.
Leukemia.
12
1998
1866
1880
4
Mandelli
F
Diverio
D
Avvisati
G
et al
Molecular remission in PML/RAR alpha-positive acute promyelocytic leukemia by combined all-trans retinoic acid and idarubicin (AIDA) therapy: Gruppo Italiano-malattie Ematologiche Maligne dell'Adulto and Associazione Italiana di Ematologia ed Oncologia Pediatrica Cooperative Groups.
Blood.
90
1997
1014
1021
5
Estey
E
Thal
PG
Pierce
S
Kantarjian
H
Keating
M
Treatment of newly diagnosed acute promyelocytic leukemia without cytarabine.
J Clin Oncol.
15
1997
483
490
6
Tallman
MS
Andersen
JW
Schiffer
CA
et al
All-trans retinoic acid in acute promyelocytic leukemia.
N Engl J Med.
337
1997
1201
1208
7
Asou
N
Adachi
J
Tamura
J
et al
Analysis of prognostic factors in newly diagnosed acute promyelocytic leukemia treated with all-trans retinoic acid and chemotherapy.
J Clin Oncol.
16
1998
78
85
8
Burnett
AK
Grimwade
D
Solomon
E
Wheatley
K
Goldstone
AH
for the MRC Adult Leukemia Working Party
Presenting white blood cell count and kinetics of molecular remission predict prognosis in acute promyelocytic leukemia treated with all-trans retinoic acid: result of the randomized MRC trial.
Blood.
93
1999
4131
4143
9
Sanz
MA
Martin
G
Rayon
C
et al
A modified AIDA protocol with anthracycline-based consolidation results in high antileukemic efficacy and reduced toxicity in newly diagnosed PML/RARa-positive acute promyelocytic leukemia.
Blood.
94
1999
3015
3021
10
Wang
Z
Sun
G
Shen
Z
Chen
S
Chen
Z
Differentiation therapy for acute promyelocytic leukemia with all-trans retinoid acid: 10-year experience of its application.
Chin Med J.
112
1999
963
967
11
Fenaux
P
Chastang
C
Chevret
S
et al
A randomized comparison of ATRA followed by chemotherapy and ATRA plus chemotherapy, and the role of maintenance therapy in newly diagnosed acute promyelocytic leukemia.
Blood.
94
1999
1192
1200
12
Lengfelder
E
Reichert
A
Schoch
C
et al
Double induction strategy including high dose cytarabine in combination with all-trans retinoic acid: effects in patients with newly diagnosed acute promyelocytic leukemia: German AML Cooperative Group.
Leukemia.
14
2000
1362
1370
13
Carli
PM
Sgro
C
Parchin-Geneste
N
et al
Increase therapy-related leukemia secondary to breast cancer.
Leukemia.
14
2000
1014
1017
14
Micallef
JN
Lillington
DM
Apostolidis
J
et al
Therapy-related myelodysplasia and secondary acute myelogenous leukemia after high-dose therapy with autologous hematopoietic progenitor cell support for lymphoid malignancies.
J Clin Oncol.
18
2000
947
955
15
Leone
G
Mele
L
Pulsoni
A
Equitani
F
Pagano
L
The incidence of secondary leukemia.
Haematologica.
84
1999
937
945
16
Jubashi
T
Nagai
K
Myazaki
Y
et al
A unique case of t(15;17) acute promyelocytic leukaemia (M3) developing into acute myeloblastic leukemia (M1) with t(7;21) at relapse.
Br J Haematol.
83
1993
665
668
17
Myazaki
H
Ino
T
Sobue
R
et al
Translocation (3;21)(q26;q22) in treatment-related acute leukemia secondary to acute promyelocytic leukemia.
Cancer Genet Cytogenet.
74
1994
84
86
18
Hatzis
T
Standen
GR
Howell
RT
Savill
C
Wagstaff
M
Scott
GL
Acute promyelocytic leukemia (M3): relapse with acute myeloblastic leukemia (M2) and dic(5;17)(q11;p11).
Am J Hematol.
48
1995
40
44
19
Bseiso
AW
Kantarijan
H
Estey
E
Myelodysplastic syndrome following successful therapy of acute promyelocytic leukemia.
Leukemia.
11
1997
168
169
20
Felice
M
Rossi
I
Gallego
M
et al
Acute trilineage leukemia with monosomy of chromosome 7 following an acute promyelocytic leukemia.
Leuk Lymphoma.
34
1999
409
413
21
Sawada
H
Morimoto
H
Wake
A
Yamasaki
Y
Izumi
Y
Therapy-related acute myeloid leukemia with a t(10;11)(q23;p15) following successful chemotherapy for acute promyelocytic leukemia with t(15;17).
Int J Hematol.
69
1999
270
271
22
Zompi
S
Legrand
O
Bouscary
D
et al
Therapy-related acute myeloid leukemia after successful therapy for acute promyelocytic leukemia with t(15;17): a report of two cases and a review of the literature.
Br J Haematol.
110
2000
610
613
23
Pecci
A
Invernizzi
R
A therapy-related myelodysplastic syndrome with unusual features in a patient treated for acute promyelocytic leukemia.
Haematologica.
86
2001
102
103
24
Au
WY
Lam
C
Ma
E
Man
C
Wan
T
Kwong
YL
Therapy-related myelodysplastic syndrome after eradication of acute promyelocytic leukemia: cytogenetic and molecular features.
Hum Pathol.
32
2001
126
129
25
Bennett
JM
Catovsky
D
Daniel
MT
et al
Proposed revised criteria for the classification of acute myeloid leukemia: a report of the French-American-British cooperative group.
Ann Intern Med.
103
1985
620
625
26
Lo Coco
F
Diverio
D
Pandolfi
PP
et al
Molecular evaluation of residual disease as a predictor of relapse in acute promyelocytic leukemia.
Lancet.
340
1992
1437
1438
27
Diverio
D
Lo Coco
F
D'Adamo
F
et al
Identification of DNA rearrangements at the RARα locus in all patients with acute promyelocytic leukemia and mapping of APL breakpoints within the RARα second intron.
Blood.
79
1992
3331
3336
28
Avvisati
G
Event-free survival (EFS) duration in newly diagnosed acute promyelocytic leucemia (APL) is favorably influenced by induction treatment with idarubicin alone: final results of the GIMEMA randomized study LAP 0389 comparing IDA vs IDA + ARA-C in newly diagnosed APL [abstract].
Blood.
94
1999
505
29
Bennett
JM
Catovsky
D
Daniel
MT
et al
Proposals for the classification of the myelodysplastic syndromes.
Br J Haematol.
51
1982
189
199
30
Todisco
E
Testi
A
Avvisati
G
et al
Therapy-related acute myelomonocytic leukemia following successful treatment for acute promyelocytic leukemia.
Leukemia.
9
1995
1533
1535
31
Estey
EH
Giles
FJ
Kantarjian
H
et al
Molecular remissions induced by liposomal-encapsulated all-trans retinoic acid in newly diagnosed acute promyelocytic leukemia.
Blood.
94
1999
2230
2235
32
Karp
JE
Sarkodee-Adoo
CB
Therapy-related acute leukemia.
Clin Lab Med.
20
2000
71
81
33
Harris
NL
Jaffe
ES
Diebold
J
et al
World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting–Airlie House, Virginia, November 1997.
J Clin Oncol.
17
1999
3835
3849
34
Sanz
MA
Lo Coco
F
Martin
G
et al
Definition of relapse risk and role of non anthracycline drugs for consolidation in patients with acute promyelocytic leukemia: a joint study of the PETHEMA and GIMEMA cooperative groups.
Blood.
96
2000
1247
1253

Author notes

Roberto Latagliata, Cattedra di Ematologia, Via Benevento 6-00161, Rome, Italy; e-mail: rob.lati@libero.it.

Sign in via your Institution