Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 1.
Published in final edited form as: Am J Hematol. 2015 Feb 27;90(6):483–486. doi: 10.1002/ajh.23981

Idarubicin, Cytarabine and Pravastatin as Induction Therapy for Untreated Acute Myeloid Leukemia and High-Risk Myelodysplastic Syndrome

Mazyar Shadman 1,2,*, Raya Mawad 3,*, Carol Dean 1, Tara Chen 4, Kathleen Shannon-Dorcy 1, Vicky Sandhu 1, Paul C Hendrie 5, Bart L Scott 1,2, Roland B Walter 1,5, Pamela S Becker 1,5, John Pagel 3, Elihu H Estey 1,5
PMCID: PMC4803479  NIHMSID: NIHMS671315  PMID: 25689471

Abstract

Background

Previous studies suggest that idarubicin/cytarabine(ara-C)/pravastatin (IAP) is an active salvage regimen for patients with AML. We therefore investigated this regimen in patients with newly-diagnosed AML or MDS (≥10% blasts).

Methods

Patients were eligible if the anticipated treatment-related mortality (TRM) was <10%. Patients received pravastatin (1,280 mg/day po; days 1-8), cytarabine (1.5 g/m2/day; days 4-7) and idarubicin (12 mg/m2/day, days 4-6). Up to 3 cycles of consolidation with a shortened course was permitted. The primary endpoints were “good CR” rate (CR on day 35 without minimal residual disease) and TRM in the first 28 days. The study was to stop if after each cohort of 5 patients (a) the Bayesian posterior probability was < 5% that the true “good CR rate” was ≥ 70% or (b) the posterior probability was >25% that the TRM rate was ≥5%.

Results

Twenty-four patients were included. Conventional CR was achieved in 15 (63%) patients but only 12 (50%) achieved “good CR”. 4 of 12 (33%) patients with “good CR” relapsed at median of 16 weeks (10.5-19). Five (21%) patients had refractory disease. Survival probability at 1 year was 72% (48.7-64). Two (8.3%) patients died within 28 days from multi organ failure. The most common grade 3-4 adverse effects were febrile neutropenia (75%) and diarrhea (25%). Based on the stopping rules accrual ceased after entry of these 24 patients.

Conclusion

IAP did not meet the pre-defined efficacy criteria for success. Therefore, we would not recommend this regimen for phase 3 testing in this patient subset.

Background

Although achieving a complete remission (CR) appears necessary for long-term survival in AML at least after receipt of 3+7 or congeners (1), CR per se is not generally sufficient to prolong survival. (1-3) One plausible explanation is that conventionally defined CRs vary widely in their quality, with only high quality CRs translating into a survival advantage. In particular, presence of minimal residual disease (MRD) at the time of CR or slow count recovery are independent risk factors for relapse and thus indicate a poor quality CR. (4, 5) Therefore, implementation of more stringent criteria for CR that account for MRD and time to count recovery seems logical.

In-vitro studies have suggested that high intracellular cholesterol levels in AML cells may be part of an adaptive chemoresistance mechanism and blockade of cholesterol synthesis with HMG-CoA reductase inhibitors (statins) restored chemosensitivity. (6, 7) Addition of statins to standard chemotherapy for AML has been explored in an attempt to improve outcomes. In a phase-I study, pravastatin was added to the standard induction regimen of idarubicin and high-dose cytarabine (Ara-C) and was associated with acceptable toxicity and resulted in clinical responses (CR and CRp) in both the frontline (11 of 15) and salvage (9 of 22) settings. (8, 9) In their 0919 trial, the SWOG noted that IAP produced a CR rate of 55% in 36 patients relapsing after a minimum CR/CRi duration of 3 months following last chemotherapy, thus meeting the pre-defined efficacy criterion. (10) The SWOG results prompted us to examine whether IAP would increase the CR rates without MRD and with rapid count recovery (“good CRs”) without increasing treatment related mortality (TRM) in adults with newly-diagnosed AML.

Patients and Methods

Eligibility

Patients age 18 to 74 with diagnoses of AML (except acute promyelocytic leukemia), refractory anemia with excess blast type 2 (RAEB-II) or myeloproliferative neoplasm (MPN) with ≥ 10% blasts in the bone marrow or peripheral blood according to the 2008 WHO classification were eligible. (11) Eligibility required an expected risk of TRM (death in the first 4 weeks after beginning induction therapy) less than 10%, as determined by an algorithm based on historical data from SWOG and MD Anderson. (12) Conventional eligibility criteria (performance status, creatinine, age) are subsumed in the TRM score. Bilirubin is not included in the TRM score but needed to be < 2.0 mg/ml due to hepatic excretion and metabolism of idarubicin. The study was approved by our institutional review board and all patients provided written informed consent.

Primary and secondary endpoints

The study had 2 primary endpoints, defined as those adaptively monitored during the study. The first was rate of “good CR”. Good CR includes conventional criteria for CR (absolute neutrophil count > 1,000/μl, platelet count > 100,000/μl, marrow with <5% morphologic blasts) and additionally the requirements that marrow MRD (detected by 10-color flow cytometry or conventional cytogenetic evaluation) be absent and that the above blood counts be attained. The details of the ten-color multiparametric flow cytometry are previously described by our group.(13) The second primary endpoint was rate of death within 28 days of starting induction therapy (TRM). Secondary endpoints were morphologic CR rate, relapse-free survival (RFS) and the overall survival (OS).

Treatment

Pravastatin 1,280 mg was administered orally daily from day 1 through day 8 of induction. This was given in 4 divided doses in the first 4 patients but, for ease of administration, was subsequently given once daily in line with previous trials. Idarubicin 12mg/m2 was administered intravenously daily on days 4 through 6 while ara-C 1.5g/m2/day was administered continuous IV infusion on days 4 through 7. Patients were taken off study if bone marrow examination between days 17-20 showed more than 60% morphologic blasts or if good CR was not observed, with bone marrow examination examined around day 35 to assess for flow cytometric or cytogenetic evidence of MRD.

Patient who achieved a “good CR” were allowed to receive up to three cycles of post-remission therapy with the same agents given as a shortened course: Pravastatin 1,280 mg orally daily from day 1 through 6. Idarubicin 12mg/m2 intravenously daily on days 4 through 5 and cytarabine 1.5 g/m2/day intravenously by continuous infusion on days 4 through 5.

To prevent/ameliorate fever and rash due to ara-C, prednisone 100mg daily could be administered on days 4 through 8 of induction and 4 through 7 of post CR remission therapy at the discretion of the treating physician. Prednisolone 1% eye drops were used to prevent ara-C-induced conjunctivitis and all patients received quinolones for antibacterial prophylaxis in addition to antiviral and antifungal prophylaxis.

Study design and statistical considerations

We used a Bayesian design to simultaneously monitor efficacy (good CR) and toxicity (TRM).(14, 15) Our institutional historical data indicated a “good CR” rate of 50% and a TRM rate of ≤5% and these numbers determined the study's prior probabilities. We aimed for 20% improvement in the good CR rate (50% to 70%) and a TRM ≤ 5. Outcomes were evaluated after each cohort of 5 patients, with the minimum sample size set at 20 and the maximum at 50.The study was to stop early if after evaluation of each cohort of 5 patients from numbers 21 through 45 either (a) the posterior probability was < 5% that the true “good CR rate” was ≥ 70% or (b) the posterior probability was >25% that the TRM rate was ≥ 5%. The operating characteristics of the design thus parameterized were such that if the true good CR and TRM rates were the same as historical the probability was 89% that accrual would stop before entry of 50 patients; this is analogous a false positive rate of 11%. If, on the other hand, the true good CR rate was > 70% and the true TRM rate < 5% the probability that 50 patients would be accrued was 68%, analogous to a power of 68%. As discussed below, the study stopped after accrual of 24 patients because the efficacy criteria were not met. The likelihood that stopping would have occurred at this time if the efficacy and TRM goals were likely to be met is discussed below.

Results

Patient characteristics (table 1)

Table 1. Patients Characteristics.

Total (n=24)
Median Age (range); years 57; (35-72)
Female Gender - n ;(%) 15; (62%)
Median WBC (range) at diagnosis × 103/μL 6.7 (0.4-111)
Diagnosis:
 AML, n (%) 21 (87.5%)
  De novo  18 (86%)
  Secondary AML - Prior therapy  1 (4.5%)
  Antecedent Hematologic Disorder  2 (9.5%)
 MDS, n (%) 2 (8.5%)
 CMML, n (%) 1 (4%)
ELN CG risk group, n (%) in AML patients (n=21)
Favorable 4 (19%)
Intermediate-I 4 (19%)
Intermediate-II 6 (28.5%)
Adverse 7 (33.5%)
Molecular markers in AML patients (n=21)
 FLT3-ITD positive 2 (8%)
 NPM1 positive 3 (14%)
 CEBPA 1 (4%)
IPSS-R score, n(%) in MDS patients (n=2)
Intermediate 1 (50%)
High 1 (50%)

TRM: treatment-related mortality, ELN: European leukemia network, IPSS-R: Revised International Prognostic Scoring System

Between May 2013 and April 2014, 24 patients with median age 57 (35-72) were treated. 21 had AML, two RAEB-2, and one CMML with > 10 blasts. 86% of the AML patients had de novo disease. Only 4 patients (19%) had favorable cytogenetics per European leukemia network (ELN) criteria.(16) The two MDS patients had IPSS-R scores of 6 and 3.5 (17).

Good CR and CR (table 2)

Table II. Outcomes.

Diagnosis Good CR* CR CRi Refractory TRM* Relapse HCT
All patients (24) 12 (50%) 15 (63%) 2 (8%) 5 (21%) 2 (8%) 5 (21%) 11 (46%)
AML (21); n (%) 10 (48%) 13 (62%) 1 (5%) 5 (24%) 2 (9%) 4 (19%) 9 (43%)
Risk groups
 Favorable (4) 3 0 0 0 1 0 0
 Intermediate-I (4) 3 1 0 0 0 1 3
 Intermediate-II (6) 2 1 1 1 1 2 2
 Adverse (7) 2 1 0 4 0 3 4
MDS (2) 2 (100%) 2(100%) 0 0 1 (50%) 1 (50%)
CMML (1) 0 0 1 (100%) 0 0 0 1 (100%)
*

Primary study endpoints ;

CR without evidence of MRD ;

per ELN criteria ; MRD: minimal residual disease, TRM: treatment-related mortality, HCT: hematopoietic stem cell transplantation

Twelve patients (50%; 95% confidence interval 29-71%) achieved “good CR”. 3 others had a CR by conventional criteria but were not considered a good CR because of MRD; thus the conventional CR rate was 15/24 (63%; 95% CI 31-78%). 2 patients met criteria for CRi, and 5 had persistent AML.

TRM and other induction toxicity (tables 2-3)

Table III. Grade 3 and 4 adverse events.

Adverse Event Number of patient (%)

Grade 3 Grade 4
ID
 Febrile Neutropenia 17 (71%) 1 (4%)
 Rash 3 (12.5%) 0
 Hand-foot syndrome 2 (8.5%) 0
 Septic Shock 0 1(4%)
GI
 Diarrhea 6 (25%) 0
 Nausea/Vomiting 4 (17%) 0
 Mucositis 3 (12.5%) 0
 Transaminitis 2 (8.5%) 0
 Colon pneumatosis 1 (4%) 0
 GI bleeding 1 (4%) 0
Renal
 Tumor lysis syndrome 1 (4%) 0

TRM occurred in 2 patients (8%; 95% CI 1-27%). Both patients died of multiple organ failure secondary to sepsis on days 13 and 19 with neutropenic enterocolitis predisposing in 1 of the 2. The incidence of grade 3-4 toxicity was similar to that observed with IA without pravastatin. (18)

Subsequent Outcomes

9 of the 12 patients (75%) who achieved good CR received further IAP; 4 (33%) received 1, 3 (25%) received 2, and 2 (16%) had 3 further courses. The principal reason for no post-remission IAP was suspected toxicity: GI side effects (nausea, vomiting and diarrhea) in 1 and rhabdomyolysis though related to pravastatin in 1. Five of the 12 good CR patients (42%) and 2 of the 3 non-good CR patients (67%) received HCT.

Four of the 12 good CR patients (33%) have relapsed at a median of 16 weeks (range : 10.5 – 19) from initial CR; median follow-up time in the remaining 41 weeks (range: 17-67) Comparable figures for the 3 patients with conventional CR but with evidence of MRD are relapse in 1 patient (33%) after 24 weeks while the median follow-up for other 2 patients is 28 weeks. 6 patients have died (median time to death = 26 weeks (range: 1- 42 weeks)) and 18 are alive at a median follow-up of 48 weeks (range 13-64). The Kaplan-Meier survival probability at 1 year was 72% (95% CI: 48.7-86.9%).

There was no difference in the level of serum cholesterol reduction between patients who achieved CR vs. ones who did not (7.8 vs. 0.5 mg/dL; p=0.4).

Discussion

IAP did not meet our criterion for efficacy (good CR rate of 70%), leading to suspension of the trial after accrual of 24 patients. Several questions arise: (1) what was the probability of a false negative result, (2) is “good CR” a valid endpoint, (3) was the sought after improvement unrealistic, and (4) why were the SWOG results in relapsed AML encouraging and our results in newly diagnosed patients not?

Beginning with the first question, the probability that early stopping would occur if the true good CR rate and TRM rates were the specified 70% and 5% respectively was 17% after entry of 25 patients. Hence it seems unlikely that our results are falsely negative at least with regard to these specifications. Of course this ignores the possibility that IAP might only be useful in certain subgroups. However, sample size calculations in AML are usually based on a similar view of the disease as a single entity. This is acknowledged to be unrealistic, begging the question how to design trials for small subgroups while retaining conventional power of 80-90% and p-value of 0.05 without requiring extraordinarily long times to complete accrual.

The most widely accepted endpoint in AML is survival. However occurrence of a sufficient number of deaths to comment realistically on survival can require 1-2 years. Hence using endpoints that can serve as surrogates for survival are desirable. Recent observations that improvements in CR rate do not necessarily translate into improvements in survival argue against use of CR as such a surrogate.(2, 3) Here we proceeded on the assumption that “good CR” might be a better surrogate than CR; while deriving from data showing that longer time to count recovery and presence of MRD at CR diminish the survival value of CR, this assumption remains to be tested. One attractive feature of using good CR as endpoint is that because good CR rates are lower than CR rates improvements in the former may be more plausible.

The degree of improvement that should be sought is also of interest. Certainly if we had specified a desired absolute improvement of 10% in good CR rate rather than 20% the study would not have stopped after accruing 24 patients. However, based on the results of our study, there is only an 18% probability that IAP is 10% better than historical regimens and an even lower chance (8%) that it is 15% better. Some would contend that improvements in AML occur in small increments and basing stopping treatment on larger increments is unrealistic and counterproductive. Others would counter that the number of patients and amount time of time needed to detect small improvements is far from desirable. This issue is likely to demand further discussion as the recognition that AML is several diseases each to be treated differently inevitably will mean fewer patients available for the various trials. It would seem that at least logistically the specified degree of improvement will have to be higher than currently, perhaps justified by the increased specificity of new therapies.

These issues aside it seems quite likely that our results in newly-diagnosed patients are not comparably satisfactory as those of Advani et al. in relapsed patients in the SWOG 0919 study. These authors reported a CR rate of 55% (20/36) vs. a CR rate of 63% (15/24) in our newly-diagnosed patients (95% CI for the difference in rates [-0.2 - 0.32], with “expected historical' rates of 30% and 70 -80% in the 2 groups respectively. A possible biologic explanation is that high intracellular cholesterol may figure more prominently as an adaptive mechanism leading to chemoresistance in relapsed AML and therefore the value of statin addition may be less in newly-diagnosed patients. Obviously, it is also possible that patient and disease characteristics may affect the outcome in AML more than treatment, here IAP. Although age and cytogenetics were similar in our patients and Advani et al.'s, attempts to develop prognostics models to predict treatment failure based on individual patient characteristics have not been successful. For example, in a recent analysis of more than 4500 patients using data from MRC/NCRI, HOVON/SAKK, SWOG and MD Anderson Cancer Center, failure to achieve a CR despite surviving the first 28 days of therapy (“resistance”) was as expected independently associated with age, performance status, white blood cell count, secondary disease, cytogenetic risk and FLT3-ITD/NPM1 mutation status at p < 0.001 for each. However, a model incorporating all these covariates had an area under the receiver operating curve (AUC) of only 0.79 essentially intermediate between certainties (AUC 1.0) and a coin flip (AUC 0.5).(19) Given this uncertainty it is possible that Advani et al's patients were “more favorable” than our in ways that remain difficult to define. As a corollary it is possible that our 24 patients were “more unfavorable” than our historical population, stressing the need for development of observed/expected ratios applicable to individual patients in a trial, although this would still suffer from the above-discussed limitations in our ability to derive expectations applicable to single arm trials.

With these constraints in mind our results suggest that despite its seeming success in relapsed patients IAP is not likely to produce a substantial increase in high quality remissions (good CRs) in newly diagnosed AML. Given various alternatives (for example volasertib)(20) we would not recommend the regimen for inclusion in a phase 3 trial in this population.

Acknowledgments

Supported by: NIH-5T32HL007093-38 (MS and RM)

Footnotes

Authors' contribution: MS,RM,JMP,EHE,PCH,BLS,RBW and PSB contributed to the conception, design, analysis and interpretation of the research; MS,RM and EHE wrote the manuscript; MS,RM,EHE,CD,KS,VS,TC contributed in data management; MS, RM,JMP,EHE, PCH,BLS,RBW,TC and PSB contributed vital data. All authors approved the final version of the manuscript.

Conflict of interest disclosures: The authors declare no competing financial interests related to this work.

References

  • 1.Walter RB, Kantarjian HM, Huang X, Pierce SA, Sun Z, Gundacker HM, Ravandi F, Faderl SH, Tallman MS, Appelbaum FR, Estey EH. 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. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010 Apr 1;28(10):1766–71. doi: 10.1200/JCO.2009.25.1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Burnett AK, Hills RK, Hunter AE, Milligan D, Kell WJ, Wheatley K, Yin J, McMullin MF, Dignum H, Bowen D, Russell NH, Group UKNCRIAW The addition of gemtuzumab ozogamicin to low-dose Ara-C improves remission rate but does not significantly prolong survival in older patients with acute myeloid leukaemia: results from the LRF AML14 and NCRI AML16 pick-a-winner comparison. Leukemia. 2013 Jan;27(1):75–81. doi: 10.1038/leu.2012.229. [DOI] [PubMed] [Google Scholar]
  • 3.Burnett AK, Russell NH, Hunter AE, Milligan D, Knapper S, Wheatley K, Yin J, McMullin MF, Ali S, Bowen D, Hills RK, Group UKNCRIAW Clofarabine doubles the response rate in older patients with acute myeloid leukemia but does not improve survival. Blood. 2013 Aug 22;122(8):1384–94. doi: 10.1182/blood-2013-04-496596. [DOI] [PubMed] [Google Scholar]
  • 4.Estey EH, Shen Y, Thall PF. Effect of time to complete remission on subsequent survival and disease-free survival time in AML, RAEB-t, and RAEB. Blood. 2000 Jan 1;95(1):72–7. [PubMed] [Google Scholar]
  • 5.Chen XXH, Bohm C, Wood BL, Pagel JM, Becker PS, Walter RB, Sandhu RK, Abkowitz JL, Appelbaum FR, Estey EH, editors. American Society of hematology (ASH) Atlanta, GA: 2012. Dec 8-11, The Relation of Clinical Response and Minimal Residual Disease and Their Prognostic Impact On Outcome in Acute Myeloid Leukemia. [DOI] [PubMed] [Google Scholar]
  • 6.Banker DE, Mayer SJ, Li HY, Willman CL, Appelbaum FR, Zager RA. Cholesterol synthesis and import contribute to protective cholesterol increments in acute myeloid leukemia cells. Blood. 2004 Sep 15;104(6):1816–24. doi: 10.1182/blood-2004-01-0395. [DOI] [PubMed] [Google Scholar]
  • 7.Li HY, Appelbaum FR, Willman CL, Zager RA, Banker DE. Cholesterol-modulating agents kill acute myeloid leukemia cells and sensitize them to therapeutics by blocking adaptive cholesterol responses. Blood. 2003 May 1;101(9):3628–34. doi: 10.1182/blood-2002-07-2283. [DOI] [PubMed] [Google Scholar]
  • 8.Kornblau SM, Banker DE, Stirewalt D, Shen D, Lemker E, Verstovsek S, Estrov Z, Faderl S, Cortes J, Beran M, Jackson CE, Chen W, Estey E, Appelbaum FR. Blockade of adaptive defensive changes in cholesterol uptake and synthesis in AML by the addition of pravastatin to idarubicin + high-dose Ara-C: a phase 1 study. Blood. 2007 Apr 1;109(7):2999–3006. doi: 10.1182/blood-2006-08-044446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Estey EH, Thall PF, Cortes JE, Giles FJ, O'Brien S, Pierce SA, Wang X, Kantarjian HM, Beran M. Comparison of idarubicin + ara-C-, fludarabine + ara-C-, and topotecan + ara-C-based regimens in treatment of newly diagnosed acute myeloid leukemia, refractory anemia with excess blasts in transformation, or refractory anemia with excess blasts. Blood. 2001 Dec 15;98(13):3575–83. doi: 10.1182/blood.v98.13.3575. [DOI] [PubMed] [Google Scholar]
  • 10.Advani AS, McDonough S, Copelan E, Willman C, Mulford DA, List AF, Sekeres MA, Othus M, Appelbaum FR. SWOG0919: a Phase 2 study of idarubicin and cytarabine in combination with pravastatin for relapsed acute myeloid leukaemia. British journal of haematology. 2014 Jul 18; doi: 10.1111/bjh.13035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, Harris NL, Le Beau MM, Hellstrom-Lindberg E, Tefferi A, Bloomfield CD. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009 Jul 30;114(5):937–51. doi: 10.1182/blood-2009-03-209262. [DOI] [PubMed] [Google Scholar]
  • 12.Walter RB, Othus M, Borthakur G, Ravandi F, Cortes JE, Pierce SA, Appelbaum FR, Kantarjian HA, Estey EH. Prediction of early death after induction therapy for newly diagnosed acute myeloid leukemia with pretreatment risk scores: a novel paradigm for treatment assignment. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2011 Nov 20;29(33):4417–23. doi: 10.1200/JCO.2011.35.7525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Walter RB, Buckley SA, Pagel JM, Wood BL, Storer BE, Sandmaier BM, Fang M, Gyurkocza B, Delaney C, Radich JP, Estey EH, Appelbaum FR. Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood. 2013 Sep 5;122(10):1813–21. doi: 10.1182/blood-2013-06-506725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Thall PF, Simon RM, Estey EH. Bayesian sequential monitoring designs for single-arm clinical trials with multiple outcomes. Statistics in medicine. 1995 Feb 28;14(4):357–79. doi: 10.1002/sim.4780140404. [DOI] [PubMed] [Google Scholar]
  • 15.Thall PF, Simon RM, Estey EH. New statistical strategy for monitoring safety and efficacy in single-arm clinical trials. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 1996 Jan;14(1):296–303. doi: 10.1200/JCO.1996.14.1.296. [DOI] [PubMed] [Google Scholar]
  • 16.Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, Dombret H, Fenaux P, Grimwade D, Larson RA, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz MA, Sierra J, Tallman MS, Lowenberg B, Bloomfield CD, European L. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010 Jan 21;115(3):453–74. doi: 10.1182/blood-2009-07-235358. [DOI] [PubMed] [Google Scholar]
  • 17.Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Sole F, Bennett JM, Bowen D, Fenaux P, Dreyfus F, Kantarjian H, Kuendgen A, Levis A, Malcovati L, Cazzola M, Cermak J, Fonatsch C, Le Beau MM, Slovak ML, Krieger O, Luebbert M, Maciejewski J, Magalhaes SM, Miyazaki Y, Pfeilstocker M, Sekeres M, Sperr WR, Stauder R, Tauro S, Valent P, Vallespi T, van de Loosdrecht AA, Germing U, Haase D. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012 Sep 20;120(12):2454–65. doi: 10.1182/blood-2012-03-420489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Atallah E, Cortes J, O'Brien S, Pierce S, Rios MB, Estey E, Markman M, Keating M, Freireich EJ, Kantarjian H. Establishment of baseline toxicity expectations with standard frontline chemotherapy in acute myelogenous leukemia. Blood. 2007 Nov 15;110(10):3547–51. doi: 10.1182/blood-2007-06-095844. [DOI] [PubMed] [Google Scholar]
  • 19.Walter RB, Othus M, Burnett AK, Lowenberg B, Kantarjian HM, Ossenkoppele GJ, Hills RK, Ravandi F, Pabst T, Evans A, Pierce SR, Vekemans MC, Appelbaum FR, Estey EH. Resistance prediction in AML: analysis of 4601 patients from MRC/NCRI, HOVON/SAKK, SWOG and MD Anderson Cancer Center. Leukemia. 2014 Aug 12; doi: 10.1038/leu.2014.242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Montalban-Bravo G, Garcia-Manero G. Novel drugs for older patients with acute myeloid leukemia. Leukemia. 2014 Aug 21; doi: 10.1038/leu.2014.244. [DOI] [PubMed] [Google Scholar]

RESOURCES