Between 10% and 40% of newly diagnosed patients with acute myeloid leukemia (AML) do not achieve complete remission with intensive induction therapy and are therefore categorized as primary refractory or resistant. Few of these patients can be cured with conventional salvage therapy. They need to be evaluated regarding eligibility for allogeneic hematopoietic stem cell transplantation (HSCT) as this is currently the treatment with the highest probability of cure. To reduce the leukemia burden prior to transplantation, salvage chemotherapy regimens need to be employed. Whenever possible, refractory/relapsed patients should be enrolled in clinical trials as we do not have highly effective and standardized treatments for this situation. Novel therapies include tyrosine kinase inhibitors, small-molecule inhibitors (eg, for Polo-like kinase 1 and aminopeptidase), inhibitors of mutated isocitrate dehydrogenase (IDH) 1 and IDH2, antibody-based therapies, and cell-based therapies. Although the majority of these therapies are still under evaluation, they are likely to enter clinical practice rapidly as a bridge to transplant and/or in older, unfit patients who are not candidates for allogeneic HSCT. In this review, we describe our approach to refractory/early relapsed AML, and we discuss treatment options for patients with regard to different clinical conditions and molecular profiles.

Primary refractory acute myeloid leukemia (AML) and early relapse remain among the most challenging scenarios in the management of AML. Primary refractory or resistant disease as defined by not achieving complete remission (CR) (ie, a remaining blast count of 5% or more after 1 to 2 cycles of intense induction therapy1 ) occurs in 10% to 40% of the patient population. Early relapse (mostly referred to by relapse within 6 months after CR1)2  must be differentiated from late relapse (>6 months) as response to salvage therapy, and overall survival (OS) is significantly different.3,4  Treatment decisions must be carefully taken and alternatives weighed against each other by looking at the complex picture of the patient and his or her age, performance status, comorbidities, cytogenetic findings, molecular profile, and patient preference.

The first question that we need to ask is whether there are means of reducing the risk for refractory disease and early relapse by the selection of the initial therapy; that is, is there a superior induction therapy compared with standard “7+3” (7 days of cytarabine and 3 days of anthracycline)? Although daunorubicin at 90 mg/m2 certainly is superior to 45 mg/m2 both in younger and the fit older patients5,6  including patients with FLT3-ITD,7  the direct comparison between daunorubicin 60 mg/m2 with 90 mg/m2 or 80 mg/m2 did not demonstrate any difference as to CR rate and OS8-10  including patients with FLT3-ITD,8  although higher-dose daunorubicin might improve outcome in core-binding factor AML.11  Thus, using 60 to 90 mg/m2 daunorubicin or 12 mg/m2 of idarubicin for 3 days is considered standard dosing. Whenever possible, we enroll newly diagnosed as well as relapsed patients into clinical trials. Outside trials, we use ICE (idarabucin 12 mg/m2 days 1, 3, and 5; cytarabine 100 mg/m2 continuously days 1-7; etoposide 100 mg/m2 days 1-3) as our standard induction therapy for fit patients tolerating intense treatment.12  In patients not fit for intensive chemotherapy, the induction therapy frequently has to be individualized taking patient and disease-specific characteristics into account.13  Outside trials, decitabine is our first choice for elderly patients unsuitable for intensive therapy. In countries where decitabine is not approved yet, low-dose cytarabine may be used instead. By introducing you to 2 patients from our practice, we would like to illustrate how we make treatment decisions for this patient group.

A 37-year-old woman was diagnosed with AML with 95% bone marrow blasts and a white count of 110 000/µL. Genetic analyses revealed a normal karyotype, an fms-like tyrosine kinase 3–internal tandem duplication (FLT3-ITD) with a length of 78 nucleotides and an allelic ratio of 0.67, and wild-type nucleophosmin 1 and CCAAT/enhancer-binding protein alpha (CEPBA). Induction chemotherapy with ICE was started, but the patient still showed 60% of blast in the bone marrow on day 15 indicating poor response. Therapy with FLA-IDA (fludarabine, cytarabine, idarubicine)14  was immediately started as second induction cycle and in addition as a bridge to allogeneic hematopoietic stem cell transplantation (HSCT). After this regimen, the patient achieved a partial remission with 8% bone marrow blasts. After 2 weeks without further chemotherapy, conditioning with fludarabine, amsacrine, cytarabine, and 4 Gy total body irradiation15  was initiated followed by allogeneic HSCT from her HLA-identical sister. Unfortunately, she relapsed 132 days after allogeneic HSCT. In the relapse sample, FLT3-ITD could be detected with an increase in the allelic ratio from 0.67 at time of diagnosis to 3.4 at time of relapse. After reduction of immunosuppressive therapy, the patient was started on a regimen consisting of sorafenib and 5-azacytidine (AZA) based on phase 2 data.16  With this regimen and 2 additional donor lymphocyte infusions, the patient’s AML was moderately controlled for 2 months. The aim was a second allogeneic HSCT with an alternative donor, but unfortunately she died because of septicemia related to her underlying AML.

Discussion for patient 1

Before diagnosing primary refractory disease, one has to define the time point when this has been determined. Primary refractory disease is usually diagnosed in patients who have not achieved CR after 2 cycles after induction chemotherapy.1  Beyond this standard definition, it has consistently been shown that the prognosis is dismal in patients who are refractory to the first induction cycle as indicated by persistence of a substantial amount of bone marrow blasts at day 15 or who do not achieve at least a partial remission at day 21 to 35.17-19  Taking this into account, response-adapted induction therapy with dose intensification during the second induction cycle is frequently12,19,20  implemented in cases of insufficient response to first induction therapy as in our patient. In patient 1, the early assessment at day 15, which we routinely do in our practice, opened the possibility to intensify treatment in a timely manner. Based on our recent analysis intensification using either high-dose cytarabine in combination with gemtuzumab ozogamicin and all-trans retinoic acid or a regimen based on high-dose cytarabine plus fludarabine significantly improved the CR rate.21  The latter is in line with the report from the Medical Research Council showing an earlier achievement of CR with FLAG-IDA (fludarabine, high-dose cytarabine, idarubicin, granulocyte colony-stimulating factor [G-CSF]) compared with standard 7+3 induction therapy.22  Our patient responded rather well to FLA-IDA with 8% residual blasts and proceeded directly to an allogeneic HSCT. However, if the marrow had been hypoplastic with 5% to 10% blast cells, we would have repeated the induction cycle with the identical chemotherapy as soon as clinically feasible (ie, in the absence of uncontrolled infection), as these patients do as well with conventional chemotherapy as with high-dose cytarabine based regimens.23  Once these patients have achieved CR after the second induction cycle, they have the same long-term outcome as patients who achieve complete blast clearance with the first induction cycle.14  The question of whether a second FLA-IDA before an allogeneic HSCT would have been beneficial cannot be answered based on currently available data. However, data from matched related donor (MRD) assessment before allogeneic HSCT indicating that a lower leukemia burden with a negative MRD assessment before allogeneic HSCT is associated with a better outcome after transplant24,25  might be in favor of a second cycle. After early relapse, our patient received sorafenib, a multi–tyrosine kinase inhibitor (TKI) in relapse after allogeneic HSCT. The use of a sorafenib in this situation holds some promise, as sorafenib may synergize with allogeneic immune effects to induce remissions.26  Furthermore, there appear to be synergistic effects between sorafenib and AZA.16 

A 58-year-old female who was treated with standard intense therapy (ICE) in our institution for core-binding factor AML with inversion 16 (inv16) (c-kit receptor tyrosine kinase wild type) in an interventional treatment trial. She achieved CR after 1 cycle of induction therapy but relapsed at 3 months after completing her consolidation therapy with 3 cycles of high-dose cytarabine. At the time of relapse the patient was pancytopenic with an absolute neutrophil count of 100/µL. An echocardiogram revealed a reduced cardiac ejection fraction of 44%. Based on high second CR rates in patients with inv(16)-AML even in the situation of an early relapse, we recommended an intensive salvage therapy in this situation. Because of her reduced ejection fraction, the patient received a modified FLA-IDA regimen with liposomal daunorubicin replacing idarubicin. During therapy, the patient became febrile and diagnostic workup showed multiple nodules in a computed tomography scan consistent with aspergillosis of the lung. The patient was treated with antifungal therapy (voriconazole), but her respiratory situation did not improve and she needed mechanical ventilation. Granulocyte transfusions were given to the patient, and she slowly improved and could be extubated. Her neutrophil count normalized on day 32. Bone marrow biopsy revealed a CR morphologically. The patient went on to allogeneic HSCT from a fully HLA-compatible unrelated donor using reduced intensity conditioning according to the fludarabine, amsacrine, cytarabine, and 4 Gy total body irradiation protocol.15  Three years later, the patient is alive in CR.

Discussion for patient 2

This patient showed 2 clinically relevant problems in the setting of her relapsed disease: cardiac insufficiency and uncontrolled infection. Cardiac insufficiency is especially relevant when anthracyclines are considered. Many patients with relapsed AML have already received high cumulative doses of anthracycline. In this situation, replacing conventional anthracycline by liposomal daunorubicin should be considered as it does not reduce its efficacy27,28  while being less cardiotoxic as shown in pediatric AML protocols. Because prolonged neutropenia is a common problem in relapsed/refractory patients, fungal and bacterial infections present a major problem. For some patients not responding to broad-spectrum antibiotic or antifungal therapy, granulocyte infusions can be helpful as a bridge to regeneration.29  Interestingly, patients with inv16 have a higher likelihood of responding to salvage therapy compared with patients in other cytogenetic groups including patients with t(8;21).30 

Looking at the scenarios, it becomes evident that there is not 1 standard therapy for all patients with refractory/early relapse AML. Is there a way to predict the probability of resistant disease at the time of AML diagnosis? Walter et al tried to answer this question based on data from 4601 newly diagnosed AML patients who were treated with standard induction therapy within the National Cancer Research Institute (NCRI), Hemato-Oncologie voor Volwassenen Nederland (HOVON), Schweizerische Arbeitsgemeinschaft für Klinische Krebsforschung (SAKK), Southwest Oncology Group (SWOG), or MD Anderson Cancer Center AML study groups. Prognostic factors for failure to achieve CR included age, performance status, white blood cell count, secondary disease, cytogenetic risk group, and FLT3-ITD/nucleophosmin 1 (NPM1) mutation status. However, the area under the receiver operator characteristic curves was only 0.78 meaning that it was difficult to forecast primary resistance based on these parameters.31  Nonetheless, there are clear associations of distinct genotypes and a high probability of resistant disease to induction therapy, for example, AML characterized by a monosomal karyotype, an inv(3)/t(3;3) or a p53-alteration.32  Not only the likelihood of refractory/early relapse AML but also the prognosis for refractory/early relapsed patients is dependent on cytogenetic and molecular features. Several scoring systems have been introduced for patients with refractory/relapsed AML in order to identify patients with an improved outcome including the one from the HOVON and the Groupe Ouest-Est d'Etude des Leucémies Aigues et autres Maladies du Sang (GOELAMS) study group (Tables 1 and 2).33 ,34,35  Cytogenetics other than favorable, mutated FLT3 (FLT3-ITD), higher age, previous HSCT, and a short duration of first CR in relapsed patients were all adverse risk factors. Again, our patients had different adverse risk factors. Although patient 1 was young, her molecular profile revealed a FLT3-ITD as a risk factor. In patient 2, the inv16 was a favorable factor in the relapse situation. Similarly, relapsed patients with the genotype double-mutated CEBPA also have a high likelihood of a second CR and a favorable long-term survival after allogeneic SCT after relapse.36  When we diagnose primary refractory/early relapse AML, we decide primarily whether the patient is a candidate for allogeneic HSCT.12  At this point, allogeneic HSCT from a matched related or unrelated donor is the treatment strategy with the highest probability of cure,37,38  although survival does not exceed 20% to 35% after 4 years.12,39  If not already done at the time of primary diagnosis, we initiate a rapid donor search. Risk factors prompting us to look for suitable donors already at the time of diagnosis include unfavorable cytogenetics based on European Leukemia Net recommendations2  and molecular markers including FLT3-ITD (especially if allelic ratios of >0.51).32,40,41  For us, there is no clear-cut age limit for allogeneic HSCT. The Hematopoietic Cell Transplant–Comorbidity Index introduced by Sorror et al in 2005 (and adjusted for age in 2014) gives a good guidance for comorbidity-associated risk assessment and should be calculated for all patients when considering transplantation.42,43 

Table 1

Prognostic scoring systems for patients with refractory/relapsed AML: GOELAMS score

FactorPoints
CR1 duration  
 ≥12 mo 
 ≤12 mo (refractory/early relapse) 
FLT3-ITD status  
 Negative 
 Positive 
Cytogenetics*  
 Favorable/intermediate 
 High risk 
FactorPoints
CR1 duration  
 ≥12 mo 
 ≤12 mo (refractory/early relapse) 
FLT3-ITD status  
 Negative 
 Positive 
Cytogenetics*  
 Favorable/intermediate 
 High risk 

For information on the GOELAMS score, see Chevallier et al.33  Prognostic groups: good (0 points; OS 58%, event-free survival [EFS] 45% at 2 y); intermediate (1 point; OS 37%, EFS 31% at 2 y); poor (2-3 points; OS 12%, EFS 12% at 2 y).

*

The cytogenetic risk group is defined according to MRC criteria.35 

Table 2

Prognostic scoring systems for patients with refractory/relapsed AML: European Prognostic Index score

FactorPoints
CR1 duration  
 ≥18 mo 
 7-18 mo 
 ≤6 months 
Cytogenetics at diagnosis  
 t(16;16) or inv16 
 t(8;21) 
 Other 
Age at relapse  
 ≤35 y 
 36-45 y 
 >45 y 
SCT before first relapse  
 No 
 Yes 
FactorPoints
CR1 duration  
 ≥18 mo 
 7-18 mo 
 ≤6 months 
Cytogenetics at diagnosis  
 t(16;16) or inv16 
 t(8;21) 
 Other 
Age at relapse  
 ≤35 y 
 36-45 y 
 >45 y 
SCT before first relapse  
 No 
 Yes 

For information on the EPI score, see Breems et al.34  Prognostic groups: favorable (1-6 points; OS of 70% at 1 y and 46% at 5 y); intermediate (7-9 points; OS of 49% at 1 y and 18% at 5 y); poor (10-14 points; OS of 16% at 1 y and 4% at 5 y).

SCT, stem cell transplantation.

If the patient is not a candidate for allogeneic HSCT (eg, because of comorbidities or patient choice), we look at other treatment modalities including new drugs and/or palliative therapy with the aim to prolong the patient’s life with a meaningful quality of life (Figure 1). Outside trials, we start with low-dose cytarabine mainly to control leukocyte counts combined with best supportive care (blood transfusions, antibiotic/antifungal treatment). In some of our frailest patients, hydroxurea or 6-mercaptopurine to control hyperleukocytosis is currently the only treatment option. Palliative care services should be integrated in patients’ care. Calculations for the gain in quality-adjusted life-years for different treatment options do not exist in Germany and are therefore not considered in our deliberations for treatment allocation.

Figure 1

Algorithm for patients with early relapsed/refractory AML. When diagnosing primary refractory AML/early relapse patients need to be evaluated for allogeneic HSCT and/or a suitable trial. DLI, donor lymphocyte infusion; HCT-CI, Hematopoietic Cell Transplant–Comorbidity Index; LDAC, low-dose cytarabine; MEC, mitoxantrone, etoposide, cytarabine. #, § indicate that clinical trials should consider the patient’s performance status; that is, fit patients should receive more intense study medications (in most cases including chemotherapy) compared with unfit patients.

Figure 1

Algorithm for patients with early relapsed/refractory AML. When diagnosing primary refractory AML/early relapse patients need to be evaluated for allogeneic HSCT and/or a suitable trial. DLI, donor lymphocyte infusion; HCT-CI, Hematopoietic Cell Transplant–Comorbidity Index; LDAC, low-dose cytarabine; MEC, mitoxantrone, etoposide, cytarabine. #, § indicate that clinical trials should consider the patient’s performance status; that is, fit patients should receive more intense study medications (in most cases including chemotherapy) compared with unfit patients.

Close modal

If allogeneic HSCT is considered as best treatment option for the patient, we need to select the most promising salvage regimen to induce remission of the disease as a “bridge to transplantation.” The aim of the salvage therapy is to reduce the leukemic burden, as one of the most significant factors for all survival end points after allogeneic HSCT is the disease status before allogeneic HSCT. The lower the leukemia burden prior to transplantation the better the outcome.12,44  Although salvage chemotherapy leads to CR rates of 40% to 60%, if CR duration was 1 year or longer, this rate drops to 10% to 15% in cases of shorter CR duration37  with the exception of AML with inv(16) or double-mutated CEBPA.30,36  Are there any superior salvage/second-line chemotherapies? Salvage therapy often includes additional drugs that have not already been used during the first induction cycle. Although a number of trials have examined different combination salvage therapies (Table 3),45-67  there is still no commonly accepted standard in this situation. Whenever possible, we include patients in a clinical trial for relapsed/refractory AML patients. Outside trials, we use FLA-IDA as a salvage regimen.20,45,68,69  If there are no infectious complications we do not add G-CSF to this regimen as there is no data supporting its use. However, idarubicin as an anthracycline appears to be an important drug in this combination as FLA alone (fludarabine, high-dose cytosine) was inferior to cytosine arabinoside, daunorubicin, and etoposide as reinduction in a study for relapsed/refractory patients.70  CR rates can be expected with this regimen in the range of 30% to 50%.68  Other regimens are comparable in their effectiveness in this situation. For instance, daunorubicin has been replaced by mitoxantrone as an alternative anthracycline in combination with cytarabine and etoposide (MEC).46  High-dose cytarabine in combination with mitoxantrone is also a common regimen in refractory/relapsed AML.47  Adding other purine analogs to salvage chemotherapy is also practiced in this situation, but these drugs can increase toxicity. The combination of clofarabine with high-dose cytarabine improved the response rate compared with high dose cytarabine alone but not OS.48  Cladribine is also an important purine analog used in the treatment of relapsed childhood AML,71  although not fulfilling its promise in relapsed adult AML.72  Sapacitabine is an oral purine analog that was investigated in elderly AML patients with newly diagnosed AML or relapsed AML showing some efficacy.73  When elacytarabine was compared with 7 other commonly used AML salvage therapies according to investigator´s choice, outcome was not significantly different between patients receiving elacytarabine or treatment in the control arm.49  Other modalities of reducing treatment toxicity is liposomal delivery of chemotherapy. CPX-351, a liposomal formulation of cytarabine:daunorubicin, has shown good efficacy in treatment related AML.74  A recent phase 2 study evaluated CPX-351 vs intense salvage therapy in relapsed patients. Here, a benefit for CPX-351 was seen for relapsed patients with a poor-risk profile34  (see Tables 1 and 2) with regard to response rates (39.3% vs 27.6%), EFS, and OS.50 

Table 3

Trials with salvage regimens in refractory/relapsed AML

ReferenceStudy designRegimensNumber of patientsRefractory/ relapsedMedian age (y)% CR
51 Phase 2 HiDAC vs HiDAC + DXR or DNR 78 42/36 37 63 vs 65 (REF: 20 vs 56) 
52 Phase 2 MTZ, etoposide 61 21/20 47 43 
53 Phase 2 MTZ, etoposide, IDAC (MEC) 32 18/14 24 66 
54 Phase 2 MTZ, etoposide, IDAC (MEC) 74 0/30 37 55 
55 Phase 2 IDAC + IDA + etoposide 97 36/61 37 43 (REF: 29) 
56 Phase 3, randomized MTZ, etoposide, AraC + G-CSF vs MTZ, etoposide, AraC 50 6/44 43 vs 47 54 vs 42 
57 Phase 3, randomized HiDAC vs HiDAC + etoposide 131 n.g. n.g. 31 vs 38 
46 Phase 2 Etoposide, MTZ, AraC (EMA) 133 22/111 43 60 (REF: 44; REL: 76) 
47 Phase 3, randomized HiDAC + MTZ vs IDAC + MTZ 186 27/159 50 47 (REF <60 y: 46 vs 26) 
58 Phase 3, randomized HiDAC vs HiDAC + MTZ 162 56/106 48 vs 53 32 vs 44 
59 Phase 3, randomized Etoposide, MTZ, AraC (EMA) + GM-CSF vs EMA 192 120/72 47 vs 46 65 vs 59 
REF: 51 vs 46; REL: 89 vs 81 
45 Phase 2 FLAG-IDA 46 10/36 41 52 
60 Phase 2 Cladribine, HiDAC, MTZ 118 78/40 45 58 (REF:51; REL: 54) 
61 Phase 2 FLAG-IDA ± GO 71 10/61 48 29 (+GO) vs 39 (−GO) (ORR 56 vs 52) 
62 Phase 2, randomized IDAC + GO vs IDAC + liposomal DNR vs AraC, CTX, topotecan 82 29/53 60 vs 52 vs 53 12 vs 7 vs 4 
63 Phase 1/2 HiDAC + clofarabine + G-CSF 50 (46 eval.) 18/32 53 46 (ORR 61) (REL: 32; REF: 67) 
64 Phase 2 IDAC + clofarabine 47 20/27 51 51 (REF: 45) 
65 Phase 2 BIDFA ± GO 93 n.g. 62 23 (ORR: 27) 
48 Phase 3, randomized IDAC + clofarabine vs IDAC 326 171/148 67 35 vs 18 (ORR: 47 vs 23) 
REF: 46 vs 23; REL: 49 vs 23 
66 Phase 2 Homoharringtonine, AraC, aclarubicine 46 11/35 37 80 (REF: 67; REL: 96) 
67 Phase 3, randomized SHAI vs SHAI + fludarabine IDAC + GO vs IDAC + liposomal DNR vs AraC, CTX, topotecan 326 n.g. 57 vs 52 35 vs 44 (ORR: 42 vs 54) 
62 Phase 2, randomized 
28 Phase 2 Fludarabine, HiDAC, liposomal DNR MTZ, etoposide 41 11/30 60 0 vs 73 
49 Phase 3, randomized Elacytarabine vs others 381 140/241 59 vs 60 15 vs 12 (ORR: 23 vs 21) 
50 Phase 2 CPX-351 vs first salvage therapy 125 125 52 vs 56 37.0 vs 31.8 
ReferenceStudy designRegimensNumber of patientsRefractory/ relapsedMedian age (y)% CR
51 Phase 2 HiDAC vs HiDAC + DXR or DNR 78 42/36 37 63 vs 65 (REF: 20 vs 56) 
52 Phase 2 MTZ, etoposide 61 21/20 47 43 
53 Phase 2 MTZ, etoposide, IDAC (MEC) 32 18/14 24 66 
54 Phase 2 MTZ, etoposide, IDAC (MEC) 74 0/30 37 55 
55 Phase 2 IDAC + IDA + etoposide 97 36/61 37 43 (REF: 29) 
56 Phase 3, randomized MTZ, etoposide, AraC + G-CSF vs MTZ, etoposide, AraC 50 6/44 43 vs 47 54 vs 42 
57 Phase 3, randomized HiDAC vs HiDAC + etoposide 131 n.g. n.g. 31 vs 38 
46 Phase 2 Etoposide, MTZ, AraC (EMA) 133 22/111 43 60 (REF: 44; REL: 76) 
47 Phase 3, randomized HiDAC + MTZ vs IDAC + MTZ 186 27/159 50 47 (REF <60 y: 46 vs 26) 
58 Phase 3, randomized HiDAC vs HiDAC + MTZ 162 56/106 48 vs 53 32 vs 44 
59 Phase 3, randomized Etoposide, MTZ, AraC (EMA) + GM-CSF vs EMA 192 120/72 47 vs 46 65 vs 59 
REF: 51 vs 46; REL: 89 vs 81 
45 Phase 2 FLAG-IDA 46 10/36 41 52 
60 Phase 2 Cladribine, HiDAC, MTZ 118 78/40 45 58 (REF:51; REL: 54) 
61 Phase 2 FLAG-IDA ± GO 71 10/61 48 29 (+GO) vs 39 (−GO) (ORR 56 vs 52) 
62 Phase 2, randomized IDAC + GO vs IDAC + liposomal DNR vs AraC, CTX, topotecan 82 29/53 60 vs 52 vs 53 12 vs 7 vs 4 
63 Phase 1/2 HiDAC + clofarabine + G-CSF 50 (46 eval.) 18/32 53 46 (ORR 61) (REL: 32; REF: 67) 
64 Phase 2 IDAC + clofarabine 47 20/27 51 51 (REF: 45) 
65 Phase 2 BIDFA ± GO 93 n.g. 62 23 (ORR: 27) 
48 Phase 3, randomized IDAC + clofarabine vs IDAC 326 171/148 67 35 vs 18 (ORR: 47 vs 23) 
REF: 46 vs 23; REL: 49 vs 23 
66 Phase 2 Homoharringtonine, AraC, aclarubicine 46 11/35 37 80 (REF: 67; REL: 96) 
67 Phase 3, randomized SHAI vs SHAI + fludarabine IDAC + GO vs IDAC + liposomal DNR vs AraC, CTX, topotecan 326 n.g. 57 vs 52 35 vs 44 (ORR: 42 vs 54) 
62 Phase 2, randomized 
28 Phase 2 Fludarabine, HiDAC, liposomal DNR MTZ, etoposide 41 11/30 60 0 vs 73 
49 Phase 3, randomized Elacytarabine vs others 381 140/241 59 vs 60 15 vs 12 (ORR: 23 vs 21) 
50 Phase 2 CPX-351 vs first salvage therapy 125 125 52 vs 56 37.0 vs 31.8 

AraC, cytarabine; BIDFA, twice-daily fludarabine and cytarabine; CTX, cyclophosphamide; DNR, daunorubicin; DXR, doxorubicin; GO, gemtuzumab ozagamicin; HiDAC, high-dose cytarabine; IDAC, intermediate-dose cytarabine; MTZ, mitoxantron; ORR, overall response rate; REF, refractory; REL, relapsed; SHAI, sequential high-dose cytarabine + idarubicin.

Whenever possible, we aim for allogeneic transplantation for patients in second CR because there are essentially no cures with chemotherapy alone. However, if patients do not achieve a significant cytoreduction in the bone marrow or have more than 25% blasts in bone marrow, we usually abstain from transplantation and recommend hypomethylating agents or trial participation (and reevaluation after these therapies). The source of the graft (peripheral blood stem cells vs bone marrow) as well as MRD or matched unrelated donor do not impact significantly on outcome.12,75  If no HLA-matched donor is available, alternative graft sources including cord blood or stem cells from a haploidentical donor should be considered in these high risk patients. However, in a head to head comparison results had been inferior with cord blood or haploidentical donors compared with MRD and matched unrelated donor.12  For patients in second CR, we tolerate less well-matched donors because there are essentially no cures with chemotherapy alone in second CR.

It is unlikely that patients with refractory AML will be cured solely by changing and improving current chemotherapy regimens. The candidate targets for novel therapeutic approaches in AML are diverse (Figure 2). Some of these novel approaches have already been studied even in phase 3 trials, others are just entering the early clinical trial phase or are under development. Although outside clinical trials the majority of these drugs are currently not available, we would still like to discuss these drugs because relapsed/refractory patients can be included in trials with these drugs and some of them are likely to be licensed soon. The list of targets and approaches is long and they can be classified in the following groups: epigenetic modifiers (demethylating agents, histone deacetylase inhibitors), antibody-based therapies (eg, GO), TKIs (eg, TKI against FLT3-ITD), small-molecule inhibitors of kinases involved in cell division (eg, Polo-like kinase 1), inhibitors of mutated enzymes (eg, inhibitors for IDH1, IDH2), aminopeptidase inhibitors (eg, Tosedostat), DOT1-like inhibitors (eg, EPZ-5676) for MLL-rearranged (MLL-r) leukemia, cell-based therapies including chimeric antigen receptor therapy, and immunomodulating agents.

Figure 2

The candidate targets in AML. cKIT, c-kit receptor tyrosine kinase; IDH, isocitrate dehydrogenase; ras, rat sarcoma.

Figure 2

The candidate targets in AML. cKIT, c-kit receptor tyrosine kinase; IDH, isocitrate dehydrogenase; ras, rat sarcoma.

Close modal

The individual approach for each patient depends on several factors: the patient’s molecular profile (eg, mutations in IDH1/IDH2, FLT3-ITD, MLL-r), trial availability, the patient’s performance status, and so forth.

Targeting FLT3

FLT3 is an important target for patients with an activating FLT3 mutation. Although 30% of younger AML patients show a FLT3-ITD at the time of diagnosis, the percentage of patients rises in the population of patients with relapsed/refractory disease, as FLT3-ITD is highly associated with refractory/relapsed disease especially with a high allelic ratio. Several TKIs (eg, sorafenib, midostaurin, quizartinib, crenolanib) have been introduced to the treatment of these patients. It is important to keep in mind that these TKIs vary significantly in their specificity and their activity against resistance-conferring kinase domain mutations in FLT3.76  This may translate to differences in their clinical efficacy. Thus, it is unlikely that the question of the efficacy of TKIs will be solved in the very near future. FLT3 inhibitors as monotherapy have only led to transient responses.77  For some TKIs results from a phase 3 trial are available, whereas for others phase 1 and 2 trials are just being initiated. Should our 37-year-old patient in case 1 have received targeted therapy with a TKI directed against FLT3 at the time of induction therapy or soon after? The answer is complex and cannot be definitely given at this point. In a randomized placebo controlled phase 3 trial, no benefit was shown for elderly newly diagnosed AML patients treated with sorafenib in combination with intense therapy compared with patients receiving placebo.78  Importantly, there was also no outcome benefit seen in the very small subgroup of patients with FLT3-ITD treated with sorafenib, and patients in the sorafenib arm showed a higher treatment-related mortality.78  However, a recently presented study in newly diagnosed younger AML patients showed a benefit for EFS and relapse-free survival if sorafenib was added to standard therapy. This effect was independent of FLT3 mutational status.79  Other TKIs against FLT3 include crenolanib, quizartinib (AC220), lestaurtinib, PLX 3397, ASP 2215, and so forth. They are currently being studied and hold promise because of the high selectivity for FLT3. Quizartinib has already been shown to be effective in patients with relapsed or refractory AML with FLT3-ITD and also some patients with wild-type FLT3.80  Lestaurtinib was evaluated against placebo in relapsed FLT3-ITD positive patients after receiving chemotherapy. Here, no benefit in survival or response was observed in the lestaurtinib treated patients.81  The expectations toward efficacy of a TKI against FLT3 might be scaled down by the fact that FLT3-ITD is likely not an early mutation in clonal evolution82  and thus less promising as a target.

Targeting IDH1/IDH2

Since the discovery of IDH1 and IDH2 mutations in ∼10% of AML patients,83-87  inhibitors of IDH1 and IDH2 are already being introduced to clinical trials. Mutant IDH1 and IDH2 appear to be ideal pharmacologic targets as enzymes can be more easily targeted compared with other mutated structures in AML. AG-221, an oral IDH2 inhibitor, is the first of its kind being already studied in a phase 1 dose escalation clinical trial. Here, AG-221 was studied as a monotherapy in IDH2 mutated patients and the clinical response rate was promising in the first interim analysis.88  The introduction of IDH1 inhibitors will follow.

Targeting MLL

A novel approach for AML with recurrent translocations at the 11q23 locus, referred to as MLL-rearranged (MLL-r) leukemia, is the DOT1L inhibitor EPZ-567689  and palbociclib as a CDK6 inhibitor.90  Both are currently tested in clinical trials (NCT01684150, EudraCT2014-003647-34).

Targeting CD33

GO presents a combination of calicheamicin and a recombinant humanized IgG4 antibody directed against CD33. Several phase 1/2 trials have looked at its utility in primary refractory/relapsed disease but larger phase 3 trials in this setting are missing. CR rates between 32% and 55% were observed for combinations of GO with chemotherapy.91-93  Interestingly, data from the MRC 15 trial showed that the addition of GO to induction therapy might be of significant benefit for patients with CBF AML.94  Although GO is not easily available outside clinical trials, we try to get this drug based on published evidence in elderly relapsed patients with CD33 expression of the blasts as a bridge to transplantation.95  GO has also been used in combination with demethylating agents such as vorinostat96  or AZA.97  These preliminary studies are especially promising for patients not tolerating intense chemotherapy, and modifications of the antibody and its linker might lead to further improvement. For patients who receive GO with the goal of allogeneic HSCT especially sinusoidal obstructive syndrome (SOS) of the liver is a concern.98  No clear risk factors for the occurrence of SOS during SCT could be identified, but the rate of death related to SOS was found to be small.98 

Other targets

The quinolone derivative vosaroxin inhibits the topoisomerase II and acts independently of the p53 mutational status. This drug was recently evaluated in combination with cytarabine (1 g/m2 on days 1-5) in a phase 3 study in patients with refractory or relapsed AML. A benefit in OS of 1.4 months (7.5 months vs 6.1 months) was observed in the overall cohort that just missed significance.99  Inhibitors of cell division such as the Polo-like kinase 1 inhibitor volasertib showed promise for AML patients.100  The oral aminopeptidase inhibitor tosedostat was studied in a multicenter phase 2 trial (NCT00780598) in elderly patients with refractory/relapsed AML with an overall response rate of 22%.

Immunologic approaches

Immune based approaches including chimeric antigen receptor therapies are still very immature and the immense operating expenses do not allow a general approach at this point. However, already well known immunomodulating agents such as lenalidomide might play a role in future AML therapy, especially in low proliferative disease.101 

Finally, epigenetic modifiers could help patients with relapsed/refractory patients. Their role might be increased by combination therapy with novel agents (tosedostat, midostaurin) or as a bridge to allogeneic transplantation for less fit patients with the perspective of receiving reduced intensity conditioning. This is based on the assumption that these drugs sensitize tumor cells to cytotoxic agents by reexpression of epigenetically silenced tumor suppressor genes.102 

In summary, the landscape of novel agents is very exciting and diverse. Although some agents might only be applicable for molecularly defined subgroups of AML patients (eg, IDH1/2 mutations, FLT3-ITD, MLL-r), other agents hold promise for a broad unselected group of relapsed/refractory AML patients.

The prognosis of refractory/early relapse AML patients remains poor even with allogeneic SCT. Our insight into the molecular landscape of AML has dramatically increased with the introduction of next-generation sequencing as it has allowed us to identify novel genetic alterations including recurrent driver mutations. Functional analysis of these genetic aberrations has helped us to unravel the process of leukemogenesis further. For patients with refractory/early relapsed AML treatments arising out of these efforts present the most promising approaches.

This study was supported by grants from Deutsche Krebshilfe (110284, 110287, 110292 and 111267); the Deutsche José Carreras Leukämie-Stiftunge.V (grant DJCLS R13/14), the German Federal Ministry of Education and Research (grant 01EO0802) (IFB-Tx), Deutsche Forschungsgemeinschaft (grants HE 5240/5-1 and HE 5240/6-1), and Dieter-Schlag Stiftung.

Contribution: F.T., R.F.S., M.H., and A.G. wrote the manuscript; and all authors read and agreed to the final version of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Arnold Ganser, Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str 1, 30625 Hannover, Germany; e-mail: ganser.arnold@mh-hannover.de.

1
Cheson
 
BD
Bennett
 
JM
Kopecky
 
KJ
, et al. 
International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia
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 [published correction appears in J Clin Oncol. 2004;22(3):576].
J Clin Oncol
2003
, vol. 
21
 
24
(pg. 
4642
-
4649
)
2
Döhner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
European LeukemiaNet
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
3
Estey
 
EH
Treatment of relapsed and refractory acute myelogenous leukemia.
Leukemia
2000
, vol. 
14
 
3
(pg. 
476
-
479
)
4
Weltermann
 
A
Fonatsch
 
C
Haas
 
OA
, et al. 
Impact of cytogenetics on the prognosis of adults with de novo AML in first relapse.
Leukemia
2004
, vol. 
18
 
2
(pg. 
293
-
302
)
5
Fernandez
 
HF
Sun
 
Z
Yao
 
X
, et al. 
Anthracycline dose intensification in acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1249
-
1259
)
6
Löwenberg
 
B
Ossenkoppele
 
GJ
van Putten
 
W
, et al. 
Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON); German AML Study Group (AMLSG); Swiss Group for Clinical Cancer Research (SAKK) Collaborative Group
High-dose daunorubicin in older patients with acute myeloid leukemia [published correction appears in N Engl J Med. 2010;362(12):1155].
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1235
-
1248
)
7
Luskin
 
MR
Lee
 
J-W
Fernandez
 
HF
, et al. 
High dose daunorubicin improves survival in AML up to age 60, across all cytogenetic risk groups including patients with unfavorable cytogenetic risk, and FLT3-ITD mutant AML: updated analyses from Eastern Cooperative Oncology Trial E1900 [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 373
8
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
A randomised comparison of daunorubicin 90mg/m2 vs 60mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients [published online ahead of print April 1, 2015].
Blood
 
doi:10.1182/blood-2015-01-623447
9
Gardin
 
C
Chevret
 
S
Pautas
 
C
, et al. 
Superior long-term outcome with idarubicin compared with high-dose daunorubicin in patients with acute myeloid leukemia age 50 years and older.
J Clin Oncol
2013
, vol. 
31
 
3
(pg. 
321
-
327
)
10
Pautas
 
C
Merabet
 
F
Thomas
 
X
, et al. 
Randomized study of intensified anthracycline doses for induction and recombinant interleukin-2 for maintenance in patients with acute myeloid leukemia age 50 to 70 years: results of the ALFA-9801 study.
J Clin Oncol
2010
, vol. 
28
 
5
(pg. 
808
-
814
)
11
Prebet
 
T
Bertoli
 
S
Delaunay
 
J
, et al. 
Anthracycline dose intensification improves molecular response and outcome of patients treated for core binding factor acute myeloid leukemia.
Haematologica
2014
, vol. 
99
 
10
(pg. 
e185
-
e187
)
12
Schlenk
 
RF
Döhner
 
K
Mack
 
S
, et al. 
Prospective evaluation of allogeneic hematopoietic stem-cell transplantation from matched related and matched unrelated donors in younger adults with high-risk acute myeloid leukemia: German-Austrian trial AMLHD98A.
J Clin Oncol
2010
, vol. 
28
 
30
(pg. 
4642
-
4648
)
13
Ossenkoppele
 
G
Löwenberg
 
B
How I treat the older patient with acute myeloid leukemia.
Blood
2015
, vol. 
125
 
5
(pg. 
767
-
774
)
14
Rowe
 
JM
Kim
 
HT
Cassileth
 
PA
, et al. 
Adult patients with acute myeloid leukemia who achieve complete remission after 1 or 2 cycles of induction have a similar prognosis: a report on 1980 patients registered to 6 studies conducted by the Eastern Cooperative Oncology Group.
Cancer
2010
, vol. 
116
 
21
(pg. 
5012
-
5021
)
15
Schmid
 
C
Schleuning
 
M
Schwerdtfeger
 
R
, et al. 
Long-term survival in refractory acute myeloid leukemia after sequential treatment with chemotherapy and reduced-intensity conditioning for allogeneic stem cell transplantation.
Blood
2006
, vol. 
108
 
3
(pg. 
1092
-
1099
)
16
Ravandi
 
F
Alattar
 
ML
Grunwald
 
MR
, et al. 
Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation.
Blood
2013
, vol. 
121
 
23
(pg. 
4655
-
4662
)
17
Wheatley
 
K
Burnett
 
AK
Goldstone
 
AH
, et al. 
A simple, robust, validated and highly predictive index for the determination of risk-directed therapy in acute myeloid leukaemia derived from the MRC AML 10 trial. United Kingdom Medical Research Council’s Adult and Childhood Leukaemia Working Parties.
Br J Haematol
1999
, vol. 
107
 
1
(pg. 
69
-
79
)
18
Kern
 
W
Haferlach
 
T
Schoch
 
C
, et al. 
Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia: data from the German AML Cooperative Group (AMLCG) 1992 Trial.
Blood
2003
, vol. 
101
 
1
(pg. 
64
-
70
)
19
Schlenk
 
RF
Benner
 
A
Hartmann
 
F
, et al. 
AML Study Group Ulm (AMLSG ULM)
Risk-adapted postremission therapy in acute myeloid leukemia: results of the German multicenter AML HD93 treatment trial.
Leukemia
2003
, vol. 
17
 
8
(pg. 
1521
-
1528
)
20
Heil
 
G
Krauter
 
J
Raghavachar
 
A
, et al. 
Risk-adapted induction and consolidation therapy in adults with de novo AML aged </= 60 years: results of a prospective multicenter trial.
Ann Hematol
2004
, vol. 
83
 
6
(pg. 
336
-
344
)
21
Wattad
 
M
Döhner
 
K
Krauter
 
J
, et al. 
Impact of the composition of salvage regimens on response and overall survival in primary refractory acute myeloid leukemia.
Haematologica
2014
, vol. 
99
 
s1
pg. 
526
  
Abstract S1351
22
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
Optimization of chemotherapy for younger patients with acute myeloid leukemia: results of the medical research council AML15 trial.
J Clin Oncol
2013
, vol. 
31
 
27
(pg. 
3360
-
3368
)
23
Büchner
 
T
Hiddemann
 
W
Wörmann
 
B
, et al. 
Double induction strategy for acute myeloid leukemia: the effect of high-dose cytarabine with mitoxantrone instead of standard-dose cytarabine with daunorubicin and 6-thioguanine: a randomized trial by the German AML Cooperative Group.
Blood
1999
, vol. 
93
 
12
(pg. 
4116
-
4124
)
24
Walter
 
RB
Buckley
 
SA
Pagel
 
JM
, et al. 
Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission.
Blood
2013
, vol. 
122
 
10
(pg. 
1813
-
1821
)
25
Bastos-Oreiro
 
M
Perez-Corral
 
A
Martínez-Laperche
 
C
, et al. 
Prognostic impact of minimal residual disease analysis by flow cytometry in patients with acute myeloid leukemia before and after allogeneic hemopoietic stem cell transplantation.
Eur J Haematol
2014
, vol. 
93
 
3
(pg. 
239
-
246
)
26
Metzelder
 
SK
Schroeder
 
T
Finck
 
A
, et al. 
High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses.
Leukemia
2012
, vol. 
26
 
11
(pg. 
2353
-
2359
)
27
Bieker
 
R
Lerchenmüller
 
C
Wehmeyer
 
J
, et al. 
Phase I study of liposomal daunorubicin in relapsed and refractory acute myeloid leukemia.
Oncol Rep
2003
, vol. 
10
 
4
(pg. 
915
-
920
)
28
De Astis
 
E
Clavio
 
M
Raiola
 
AM
, et al. 
Liposomal daunorubicin, fludarabine, and cytarabine (FLAD) as bridge therapy to stem cell transplant in relapsed and refractory acute leukemia.
Ann Hematol
2014
, vol. 
93
 
12
(pg. 
2011
-
2018
)
29
Bishton
 
M
Chopra
 
R
The role of granulocyte transfusions in neutropenic patients.
Br J Haematol
2004
, vol. 
127
 
5
(pg. 
501
-
508
)
30
Schlenk
 
RF
Benner
 
A
Krauter
 
J
, et al. 
Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup.
J Clin Oncol
2004
, vol. 
22
 
18
(pg. 
3741
-
3750
)
31
Walter
 
RB
Othus
 
M
Burnett
 
AK
, et al. 
Resistance prediction in AML: analysis of 4,601 patients from MRC/NCRI, HOVON/SAKK, SWOG, and MD Anderson Cancer Center.
Leukemia
2015
, vol. 
29
 
2
(pg. 
312
-
320
)
32
Schlenk
 
RF
Döhner
 
H
Genomic applications in the clinic: use in treatment paradigm of acute myeloid leukemia.
Hematology Am Soc Hematol Educ Program
2013
, vol. 
2013
 (pg. 
324
-
330
)
33
Chevallier
 
P
Labopin
 
M
Turlure
 
P
, et al. 
A new Leukemia Prognostic Scoring System for refractory/relapsed adult acute myelogeneous leukaemia patients: a GOELAMS study.
Leukemia
2011
, vol. 
25
 
6
(pg. 
939
-
944
)
34
Breems
 
DA
Van Putten
 
WL
Huijgens
 
PC
, et al. 
Prognostic index for adult patients with acute myeloid leukemia in first relapse.
J Clin Oncol
2005
, vol. 
23
 
9
(pg. 
1969
-
1978
)
35
Grimwade
 
D
Walker
 
H
Oliver
 
F
, et al. 
The Medical Research Council Adult and Children’s Leukaemia Working Parties
The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial.
Blood
1998
, vol. 
92
 
7
(pg. 
2322
-
2333
)
36
Schlenk
 
RF
Taskesen
 
E
van Norden
 
Y
, et al. 
The value of allogeneic and autologous hematopoietic stem cell transplantation in prognostically favorable acute myeloid leukemia with double mutant CEBPA.
Blood
2013
, vol. 
122
 
9
(pg. 
1576
-
1582
)
37
Estey
 
E
Kornblau
 
S
Pierce
 
S
Kantarjian
 
H
Beran
 
M
Keating
 
M
A stratification system for evaluating and selecting therapies in patients with relapsed or primary refractory acute myelogenous leukemia.
Blood
1996
, vol. 
88
 
2
pg. 
756
 
38
Biggs
 
JC
Horowitz
 
MM
Gale
 
RP
, et al. 
Bone marrow transplants may cure patients with acute leukemia never achieving remission with chemotherapy.
Blood
1992
, vol. 
80
 
4
(pg. 
1090
-
1093
)
39
Duval
 
M
Klein
 
JP
He
 
W
, et al. 
Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure.
J Clin Oncol
2010
, vol. 
28
 
23
(pg. 
3730
-
3738
)
40
Damm
 
F
Heuser
 
M
Morgan
 
M
, et al. 
Integrative prognostic risk score in acute myeloid leukemia with normal karyotype.
Blood
2011
, vol. 
117
 
17
(pg. 
4561
-
4568
)
41
Schlenk
 
RF
Kayser
 
S
Bullinger
 
L
, et al. 
German-Austrian AML Study Group
Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation.
Blood
2014
, vol. 
124
 
23
(pg. 
3441
-
3449
)
42
Sorror
 
ML
Maris
 
MB
Storb
 
R
, et al. 
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT.
Blood
2005
, vol. 
106
 
8
(pg. 
2912
-
2919
)
43
Sorror
 
ML
Storb
 
RF
Sandmaier
 
BM
, et al. 
Comorbidity-age index: a clinical measure of biologic age before allogeneic hematopoietic cell transplantation.
J Clin Oncol
2014
, vol. 
32
 
29
(pg. 
3249
-
3256
)
44
Sierra
 
J
Storer
 
B
Hansen
 
JA
, et al. 
Transplantation of marrow cells from unrelated donors for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching, and marrow cell dose.
Blood
1997
, vol. 
89
 
11
(pg. 
4226
-
4235
)
45
Pastore
 
D
Specchia
 
G
Carluccio
 
P
, et al. 
FLAG-IDA in the treatment of refractory/relapsed acute myeloid leukemia: single-center experience.
Ann Hematol
2003
, vol. 
82
 
4
(pg. 
231
-
235
)
46
Archimbaud
 
E
Thomas
 
X
Leblond
 
V
, et al. 
Timed sequential chemotherapy for previously treated patients with acute myeloid leukemia: long-term follow-up of the etoposide, mitoxantrone, and cytarabine-86 trial.
J Clin Oncol
1995
, vol. 
13
 
1
(pg. 
11
-
18
)
47
Kern
 
W
Aul
 
C
Maschmeyer
 
G
, et al. 
Superiority of high-dose over intermediate-dose cytosine arabinoside in the treatment of patients with high-risk acute myeloid leukemia: results of an age-adjusted prospective randomized comparison.
Leukemia
1998
, vol. 
12
 
7
(pg. 
1049
-
1055
)
48
Faderl
 
S
Wetzler
 
M
Rizzieri
 
D
, et al. 
Clofarabine plus cytarabine compared with cytarabine alone in older patients with relapsed or refractory acute myelogenous leukemia: results from the CLASSIC I Trial.
J Clin Oncol
2012
, vol. 
30
 
20
(pg. 
2492
-
2499
)
49
Roboz
 
GJ
Rosenblat
 
T
Arellano
 
M
, et al. 
International randomized phase III study of elacytarabine versus investigator choice in patients with relapsed/refractory acute myeloid leukemia.
J Clin Oncol
2014
, vol. 
32
 
18
(pg. 
1919
-
1926
)
50
Cortes
 
JE
Goldberg
 
SL
Feldman
 
EJ
, et al. 
Phase II, multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML.
Cancer
2015
, vol. 
121
 
2
(pg. 
234
-
242
)
51
Herzig
 
RH
Lazarus
 
HM
Wolff
 
SN
Phillips
 
GL
Herzig
 
GP
High-dose cytosine arabinoside therapy with and without anthracycline antibiotics for remission reinduction of acute nonlymphoblastic leukemia.
J Clin Oncol
1985
, vol. 
3
 
7
(pg. 
992
-
997
)
52
Ho
 
AD
Lipp
 
T
Ehninger
 
G
, et al. 
Combination of mitoxantrone and etoposide in refractory acute myelogenous leukemia—an active and well-tolerated regimen.
J Clin Oncol
1988
, vol. 
6
 
2
(pg. 
213
-
217
)
53
Amadori
 
S
Arcese
 
W
Isacchi
 
G
, et al. 
Mitoxantrone, etoposide, and intermediate-dose cytarabine: an effective and tolerable regimen for the treatment of refractory acute myeloid leukemia.
J Clin Oncol
1991
, vol. 
9
 
7
(pg. 
1210
-
1214
)
54
Spadea
 
A
Petti
 
MC
Fazi
 
P
, et al. 
Mitoxantrone, etoposide and intermediate-dose Ara-C (MEC): an effective regimen for poor risk acute myeloid leukemia.
Leukemia
1993
, vol. 
7
 
4
(pg. 
549
-
552
)
55
Carella
 
AM
Carlier
 
P
Pungolino
 
E
, et al. 
The GIMEMA Cooperative Group
Idarubicin in combination with intermediate-dose cytarabine and VP-16 in the treatment of refractory or rapidly relapsed patients with acute myeloid leukemia.
Leukemia
1993
, vol. 
7
 
2
(pg. 
196
-
199
)
56
Ohno
 
R
Naoe
 
T
Kanamaru
 
A
, et al. 
Kohseisho Leukemia Study Group
A double-blind controlled study of granulocyte colony-stimulating factor started two days before induction chemotherapy in refractory acute myeloid leukemia.
Blood
1994
, vol. 
83
 
8
(pg. 
2086
-
2092
)
57
Vogler
 
WR
McCarley
 
DL
Stagg
 
M
, et al. 
A phase III trial of high-dose cytosine arabinoside with or without etoposide in relapsed and refractory acute myelogenous leukemia. A Southeastern Cancer Study Group trial.
Leukemia
1994
, vol. 
8
 
11
(pg. 
1847
-
1853
)
58
Karanes
 
C
Kopecky
 
KJ
Head
 
DR
, et al. 
A phase III comparison of high dose ARA-C (HIDAC) versus HIDAC plus mitoxantrone in the treatment of first relapsed or refractory acute myeloid leukemia Southwest Oncology Group Study.
Leuk Res
1999
, vol. 
23
 
9
(pg. 
787
-
794
)
59
Thomas
 
X
Fenaux
 
P
Dombret
 
H
, et al. 
Granulocyte-macrophage colony-stimulating factor (GM-CSF) to increase efficacy of intensive sequential chemotherapy with etoposide, mitoxantrone and cytarabine (EMA) in previously treated acute myeloid leukemia: a multicenter randomized placebo-controlled trial (EMA91 Trial).
Leukemia
1999
, vol. 
13
 
8
(pg. 
1214
-
1220
)
60
Wierzbowska
 
A
Robak
 
T
Pluta
 
A
, et al. 
Polish Adult Leukemia Group
Cladribine combined with high doses of arabinoside cytosine, mitoxantrone, and G-CSF (CLAG-M) is a highly effective salvage regimen in patients with refractory and relapsed acute myeloid leukemia of the poor risk: a final report of the Polish Adult Leukemia Group.
Eur J Haematol
2008
, vol. 
80
 
2
(pg. 
115
-
126
)
61
Martin
 
MG
Augustin
 
KM
Uy
 
GL
, et al. 
Salvage therapy for acute myeloid leukemia with fludarabine, cytarabine, and idarubicin with or without gemtuzumab ozogamicin and with concurrent or sequential G-CSF.
Am J Hematol
2009
, vol. 
84
 
11
(pg. 
733
-
737
)
62
Litzow
 
MR
Othus
 
M
Cripe
 
LD
, et al. 
Eastern Cooperative Oncology Group Leukemia Committee
Failure of three novel regimens to improve outcome for patients with relapsed or refractory acute myeloid leukaemia: a report from the Eastern Cooperative Oncology Group.
Br J Haematol
2010
, vol. 
148
 
2
(pg. 
217
-
225
)
63
Becker
 
PS
Kantarjian
 
HM
Appelbaum
 
FR
, et al. 
Clofarabine with high dose cytarabine and granulocyte colony-stimulating factor (G-CSF) priming for relapsed and refractory acute myeloid leukaemia.
Br J Haematol
2011
, vol. 
155
 
2
(pg. 
182
-
189
)
64
Scappini
 
B
Gianfaldoni
 
G
Caracciolo
 
F
, et al. 
Cytarabine and clofarabine after high-dose cytarabine in relapsed or refractory AML patients.
Am J Hematol
2012
, vol. 
87
 
12
(pg. 
1047
-
1051
)
65
Jabbour
 
E
Garcia-Manero
 
G
Cortes
 
J
, et al. 
Twice-daily fludarabine and cytarabine combination with or without gentuzumab ozogamicin is effective in patients with relapsed/refractory acute myeloid leukemia, high-risk myelodysplastic syndrome, and blast- phase chronic myeloid leukemia.
Clin Lymphoma Myeloma Leuk
2012
, vol. 
12
 
4
(pg. 
244
-
251
)
66
Yu
 
W
Mao
 
L
Qian
 
J
, et al. 
Homoharringtonine in combination with cytarabine and aclarubicin in the treatment of refractory/relapsed acute myeloid leukemia: a single-center experience.
Ann Hematol
2013
, vol. 
92
 
8
(pg. 
1091
-
1100
)
67
Fiegl
 
M
Unterhalt
 
M
Kern
 
W
, et al. 
German AML Cooperative Group (AMLCG)
Chemomodulation of sequential high-dose cytarabine by fludarabine in relapsed or refractory acute myeloid leukemia: a randomized trial of the AMLCG.
Leukemia
2014
, vol. 
28
 
5
(pg. 
1001
-
1007
)
68
Jackson
 
G
Taylor
 
P
Smith
 
GM
, et al. 
A multicentre, open, non-comparative phase II study of a combination of fludarabine phosphate, cytarabine and granulocyte colony-stimulating factor in relapsed and refractory acute myeloid leukaemia and de novo refractory anaemia with excess of blasts in transformation.
Br J Haematol
2001
, vol. 
112
 
1
(pg. 
127
-
137
)
69
Luo
 
S
Cai
 
F
Jiang
 
L
, et al. 
Clinical study of Mito-FLAG regimen in treatment of relapsed acute myeloid leukemia.
Exp Ther Med
2013
, vol. 
5
 
3
(pg. 
982
-
986
)
70
Milligan
 
DW
Wheatley
 
K
Littlewood
 
T
Craig
 
JI
Burnett
 
AK
NCRI Haematological Oncology Clinical Studies Group
Fludarabine and cytosine are less effective than standard ADE chemotherapy in high-risk acute myeloid leukemia, and addition of G-CSF and ATRA are not beneficial: results of the MRC AML-HR randomized trial.
Blood
2006
, vol. 
107
 
12
(pg. 
4614
-
4622
)
71
Krance
 
RA
Hurwitz
 
CA
Head
 
DR
, et al. 
Experience with 2-chlorodeoxyadenosine in previously untreated children with newly diagnosed acute myeloid leukemia and myelodysplastic diseases.
J Clin Oncol
2001
, vol. 
19
 
11
(pg. 
2804
-
2811
)
72
Van Den Neste
 
E
Martiat
 
P
Mineur
 
P
, et al. 
2-Chlorodeoxyadenosine with or without daunorubicin in relapsed or refractory acute myeloid leukemia.
Ann Hematol
1998
, vol. 
76
 
1
(pg. 
19
-
23
)
73
Kantarjian
 
H
Faderl
 
S
Garcia-Manero
 
G
, et al. 
Oral sapacitabine for the treatment of acute myeloid leukaemia in elderly patients: a randomised phase 2 study.
Lancet Oncol
2012
, vol. 
13
 
11
(pg. 
1096
-
1104
)
74
Lancet
 
JE
Cortes
 
JE
Hogge
 
DE
, et al. 
Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML.
Blood
2014
, vol. 
123
 
21
(pg. 
3239
-
3246
)
75
Holtick
 
U
Albrecht
 
M
Chemnitz
 
JM
, et al. 
Bone marrow versus peripheral blood allogeneic haematopoietic stem cell transplantation for haematological malignancies in adults.
Cochrane Database Syst Rev
2014
, vol. 
4
 pg. 
CD010189
 
76
Smith
 
CC
Lasater
 
EA
Lin
 
KC
, et al. 
Crenolanib is a selective type I pan-FLT3 inhibitor.
Proc Natl Acad Sci USA
2014
, vol. 
111
 
14
(pg. 
5319
-
5324
)
77
Knapper
 
S
Burnett
 
AK
Littlewood
 
T
, et al. 
A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy.
Blood
2006
, vol. 
108
 
10
(pg. 
3262
-
3270
)
78
Serve
 
H
Krug
 
U
Wagner
 
R
, et al. 
Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial.
J Clin Oncol
2013
, vol. 
31
 
25
(pg. 
3110
-
3118
)
79
Röllig
 
C
Müller-Tidow
 
C
Hüttmann
 
A
, et al. 
Sorafenib versus placebo in addition to standard therapy in younger patients with newly diagnosed acute myeloid leukemia: results from 267 patients treated in the randomized placebo-controlled SAL-Soraml trial [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 6
80
Cortes
 
JE
Kantarjian
 
H
Foran
 
JM
, et al. 
Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status.
J Clin Oncol
2013
, vol. 
31
 
29
(pg. 
3681
-
3687
)
81
Levis
 
M
Ravandi
 
F
Wang
 
ES
, et al. 
Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse.
Blood
2011
, vol. 
117
 
12
(pg. 
3294
-
3301
)
82
Welch
 
JS
Ley
 
TJ
Link
 
DC
, et al. 
The origin and evolution of mutations in acute myeloid leukemia.
Cell
2012
, vol. 
150
 
2
(pg. 
264
-
278
)
83
Mardis
 
ER
Ding
 
L
Dooling
 
DJ
, et al. 
Recurring mutations found by sequencing an acute myeloid leukemia genome.
N Engl J Med
2009
, vol. 
361
 
11
(pg. 
1058
-
1066
)
84
Ward
 
PS
Patel
 
J
Wise
 
DR
, et al. 
The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate.
Cancer Cell
2010
, vol. 
17
 
3
(pg. 
225
-
234
)
85
Paschka
 
P
Schlenk
 
RF
Gaidzik
 
VI
, et al. 
IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication.
J Clin Oncol
2010
, vol. 
28
 
22
(pg. 
3636
-
3643
)
86
Wagner
 
K
Damm
 
F
Göhring
 
G
, et al. 
Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor.
J Clin Oncol
2010
, vol. 
28
 
14
(pg. 
2356
-
2364
)
87
Thol
 
F
Damm
 
F
Wagner
 
K
, et al. 
Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia.
Blood
2010
, vol. 
116
 
4
(pg. 
614
-
616
)
88
Stein
 
EM
Altman
 
JK
Collins
 
R
, et al. 
AG-221, an oral, selective, first-in-class, potent inhibitor of the IDH2 mutant metabolic enzyme, induces durable remissions in a phase I study in patients with IDH2 mutation positive advanced hematologic malignancies [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 115
89
Daigle
 
SR
Olhava
 
EJ
Therkelsen
 
CA
, et al. 
Potent inhibition of DOT1L as treatment of MLL-fusion leukemia.
Blood
2013
, vol. 
122
 
6
(pg. 
1017
-
1025
)
90
Placke
 
T
Faber
 
K
Nonami
 
A
, et al. 
Requirement for CDK6 in MLL-rearranged acute myeloid leukemia.
Blood
2014
, vol. 
124
 
1
(pg. 
13
-
23
)
91
Stone
 
RM
Moser
 
B
Sanford
 
B
, et al. 
Cancer and Leukemia Group B
High dose cytarabine plus gemtuzumab ozogamicin for patients with relapsed or refractory acute myeloid leukemia: Cancer and Leukemia Group B study 19902.
Leuk Res
2011
, vol. 
35
 
3
(pg. 
329
-
333
)
92
Chevallier
 
P
Delaunay
 
J
Turlure
 
P
, et al. 
Long-term disease-free survival after gemtuzumab, intermediate-dose cytarabine, and mitoxantrone in patients with CD33(+) primary resistant or relapsed acute myeloid leukemia.
J Clin Oncol
2008
, vol. 
26
 
32
(pg. 
5192
-
5197
)
93
Farhat
 
H
Reman
 
O
Raffoux
 
E
, et al. 
Fractionated doses of gemtuzumab ozogamicin with escalated doses of daunorubicin and cytarabine as first acute myeloid leukemia salvage in patients aged 50-70-year old: a phase 1/2 study of the acute leukemia French association.
Am J Hematol
2012
, vol. 
87
 
1
(pg. 
62
-
65
)
94
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
95
Bornhäuser
 
M
Illmer
 
T
Oelschlaegel
 
U
, et al. 
Gemtuzumab ozogamicin as part of reduced-intensity conditioning for allogeneic hematopoietic cell transplantation in patients with relapsed acute myeloid leukemia.
Clin Cancer Res
2008
, vol. 
14
 
17
(pg. 
5585
-
5593
)
96
Walter
 
RB
Medeiros
 
BC
Gardner
 
KM
, et al. 
Gemtuzumab ozogamicin in combination with vorinostat and azacitidine in older patients with relapsed or refractory acute myeloid leukemia: a phase I/II study.
Haematologica
2014
, vol. 
99
 
1
(pg. 
54
-
59
)
97
Nand
 
S
Othus
 
M
Godwin
 
JE
, et al. 
A phase 2 trial of azacitidine and gemtuzumab ozogamicin therapy in older patients with acute myeloid leukemia.
Blood
2013
, vol. 
122
 
20
(pg. 
3432
-
3439
)
98
Tallman
 
MS
McDonald
 
GB
DeLeve
 
LD
, et al. 
Incidence of sinusoidal obstruction syndrome following Mylotarg (gemtuzumab ozogamicin): a prospective observational study of 482 patients in routine clinical practice.
Int J Hematol
2013
, vol. 
97
 
4
(pg. 
456
-
464
)
99
Ravandi
 
F
Ritchie
 
E
Sayar
 
H
, et al. 
Improved survival in patients with first relapsed or refractory acute myeloid leukemia (AML) treated with vosaroxin plus cytarabine versus placebo plus cytarabine: results of a phase 3 double-blind randomized controlled multinational study (VALOR) [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract LBA-6
100
Döhner
 
H
Lübbert
 
M
Fiedler
 
W
, et al. 
Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy.
Blood
2014
, vol. 
124
 
9
(pg. 
1426
-
1433
)
101
Blum
 
W
Klisovic
 
RB
Becker
 
H
, et al. 
Dose escalation of lenalidomide in relapsed or refractory acute leukemias.
J Clin Oncol
2010
, vol. 
28
 
33
(pg. 
4919
-
4925
)
102
Plumb
 
JA
Strathdee
 
G
Sludden
 
J
Kaye
 
SB
Brown
 
R
Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter.
Cancer Res
2000
, vol. 
60
 
21
(pg. 
6039
-
6044
)
Sign in via your Institution