The article by Fehniger et al in this issue of Blood suggests that lenalidomide at 50 mg daily is a qualitatively different, and potentially useful, therapy for AML.1 

To date the drug, at 10 mg daily, has found its greatest use in reducing red cell transfusion requirements in patients with low-grade myelodysplasia (< 10% blasts) and a deletion of the long arm of chromosome 5 (del 5q).2  Treating 33 acute myeloid leukemia (AML) patients age 60 years or older, none of whom had del 5q, Fehniger et al noted 6 complete remissions (CRs) and 4 CRs with incomplete count recovery (CRi). In contrast to remissions seen after daunorubicin and cytarabine (“conventional treatment”), remissions after lenalidomide occurred without marrow hypoplasia (cellularity < 10%), and there was no apparent relation between achievement of remission and lenalidomide-induced neutropenia. Also, unlike conventional treatment, cytogenetics (ie, intermediate vs “unfavorable” using Southwest Oncology Group/Eastern Cooperative Oncology Group criteria) were not the principal predictor of response. Response was unrelated to age (60-64 years, 65-69 years, or older than 69 years). Rather, there was a striking inverse relation between response and extent of disease, quantified by marrow blast percentage or number of circulating blasts. Extent of disease is typically not a major predictor of response to 3 + 7 but was found by Ades et al to be such in a trial administering lenalidomide to patients with “high-risk” myelodysplastic syndrome (MDS), or AML with up to 30% marrow blasts.3  Specifically, Ades et al reported a CR in 6 of 29 in patients with less than 20% marrow blasts but only 1 of 18 in patients with 20%-29% blasts who achieved a CR. Because all of these patients had a del 5q and a similar median age to the Fehniger et al study, the higher response rate in the latter (5 of 8 in patients with 20%-30% marrow blasts) likely results from use of a 50-mg rather than the conventional 10-mg dose used by Ades et al.3  Because more myelosuppression did not seem to translate into a higher response rate, the reason for the dose-response relationship is not immediately obvious. However, the lenalidomide dose-response relation does appear steeper than that seen with conventional treatment; for example, 10 mg vs 50 mg of lenalidomide is comparable to the dose-response relationship of 100 vs 3000 mg/m2 cytarabine.

Perhaps the most striking difference with conventional treatment is the seeming lack of a relation (P = .37) between survival and response category (CR vs CRi). For many years response to induction therapy for AML was considered CR or no CR, based on 50-year-old data suggesting that only CR increased survival.4  Walter et al have reported that with conventional treatment, CR was associated with longer survival than CRp (CR with a platelet count < 100 000/μL).5  Although it seems reasonable to hypothesize that CRi will be associated with shorter survival than CRp given that it appears a lesser response requiring no recovery of neutrophils, large databases are not readily equipped to address the relative value of CRi. And certainly, what is found regarding survival in patients with CR versus CRi receiving conventional treatment may not apply with other therapies, as has been suggested with azacitidine.6  Fehniger et al are careful to note that their ability to detect longer survival with CR than with CRi was limited by patient numbers, and indeed the data in their Table 3 indicate that the median survival for CRi was 8.5 months versus at least 16 months for CR. Because patients are interested in “response” primarily as it affects survival, the survival value of responses less than CR will undoubtedly be further elucidated in the future.

Although lenalidomide is probably qualitatively distinct from conventional therapy, outcomes after its use—as noted by Fehniger et al—do not appear obviously better. While agreeing with the authors that lack of randomization hinders comparison, the CR rate was plausibly lower than might be expected had some of these patients (for example, those age 60-65 years with a normal karyotype) received conventional therapy7  (or, more recently, escalated doses of daunorubicin8 ). However, as widely recognized, it is important to move to a more “personalized” approach. In particular, it may be possible to identify patients (eg, those with low blast counts and other, to be discovered, markers) whose survival with 50 mg of lenalidomide is superior to that seen with more conventional therapy.

Lenalidomide's future in AML almost certainly lies in combination with other agents such as azacitidine9  or conventional treatment. Indeed, many new anti-AML drugs appear promising in single-arm phase 2 trials such as that reported by Fehniger et al. A very incomplete list includes plerixafor, sapacitabine, voreloxin, suberoylanilide hydroxamic acid(SAHA), AT-406, and bortezomib. It is not immediately apparent which of these is better than conventional treatment. This uncertainty together with the large number of drugs to test will likely sound the death knell for the conventional phase 3 trial, especially when utilizing conventional hazard ratios. Phase 3 trials are ill-suited for a disease as heterogeneous as AML. Both in Europe and recently in US cooperative groups, smaller comparative trials have been explored, under the assumption that the worst false-negative results when a drug is not studied at all.10  In this way, as in the introduction of qualitatively distinct drugs such as lenalidomide, clinical research in AML is truly in ferment.

Conflict-of-interest disclosure: The author declares no competing financial interests. ■

1
Fehniger
 
TA
Uy
 
GL
Trinkaus
 
K
, et al. 
A phase 2 study of high-dose lenalidomide as initial therapy for older patients with acute myeloid leukemia.
Blood
2010
, vol. 
117
 
6
(pg. 
1828
-
1833
)
2
List
 
A
Dewald
 
G
Bennett
 
J
, et al. 
Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion.
N Engl J Med
2006
, vol. 
355
 
14
(pg. 
1456
-
1465
)
3
Ades
 
L
Boehrer
 
S
Prebet
 
T
, et al. 
Efficacy and safety of lenalidomide in intermediate-2 or high-risk myelodysplastic syndromes with 5q deletion: results of a phase 2 study.
Blood
2009
, vol. 
113
 
17
(pg. 
3947
-
3952
)
4
Freireich
 
EJ
Gehan
 
EA
Sulman
 
D
Boggs
 
DR
Frei
 
E
The effect of chemotherapy on acute leukemia in the human.
J Chronic Dis
1961
, vol. 
14
 (pg. 
593
-
608
)
5
Walter
 
RB
Kantarjian
 
HM
Huang
 
X
, et al. 
Effect of complete remission and responses less than complete remission on survival in acute myeloid leukemia: a combined Eastern Cooperative Oncology Group, Southwest Oncology Group, and M. D. Anderson Cancer Center Study.
J Clin Oncol
2010
, vol. 
28
 
10
(pg. 
1766
-
1771
)
6
Fenaux
 
P
Mufti
 
GJ
Hellstrom-Lindberg
 
E
, et al. 
Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia.
J Clin Oncol
2009
, vol. 
28
 
4
(pg. 
562
-
569
)
7
Wheatley
 
K
Brookes
 
CL
Howman
 
AJ
, et al. 
Prognostic factor analysis of the survival of elderly patients with AML in the MRC AML11 and LRF AML14 trials.
Br J Haematol
2009
, vol. 
145
 
5
(pg. 
598
-
605
)
8
Lowenberg
 
B
Ossenkoppele
 
GJ
van Putten
 
W
, et al. 
High-dose daunorubicin in older patients with acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1235
-
48
)
9
Sekeres
 
MA
List
 
AF
Cuthbertson
 
D
, et al. 
Phase I combination of lenalidomide and azacitidine in patients with higher risk myelodysplastic syndromes.
J Clin Oncol
2010
, vol. 
28
 
13
(pg. 
2253
-
2258
)
10
Estey
 
E
Thall
 
PF
New designs for phase 2 clinical trials
Blood
2003
, vol. 
102
 
2
(pg. 
442
-
448
)
Sign in via your Institution