Open access peer-reviewed chapter

Target Therapy in Acute Myeloid Leukemia

Written By

Vasko Graklanov

Submitted: 29 May 2020 Reviewed: 11 October 2020 Published: 28 October 2020

DOI: 10.5772/intechopen.94422

From the Edited Volume

Acute Leukemias

Edited by Pier Paolo Piccaluga

Chapter metrics overview

632 Chapter Downloads

View Full Metrics

Abstract

Acute myeloid leukemia (AML) is the most common form of acute leukemia in elderly patients. Over the past four decades the basic therapeutic armamentarium was the standard cytotoxic treatment. The new insights in understanding the pathogenesis of AML was the momentum that revolutionized the treatment landscape in AML. The last five years unprecedented growth has been seen in the number of target therapy drugs for the treatment of AML. These new drugs did not just have a clinical benefit as single agents but also have improved AML patient outcomes if combined with conventional cytotoxic therapy. Here, we review recent advances in target-based therapy for patients with AML focusing on their mechanism of action and the results from already published clinical trials.

Keywords

  • acute myeloid leukemia
  • target therapy
  • FLT3 inhibitors
  • IDH inhibitors
  • pro-apoptotic agents
  • smoothened inhibitors
  • checkpoint inhibitors
  • CD33-targeted therapy
  • E-selectin inhibitors
  • Polo-like kinase inhibitors

1. Introduction

Acute myeloid leukemia (AML) is a heterogeneous malignant disease, characterized by uncontrolled proliferation with impaired differentiation of myeloid progenitor cells and aggressive clinical course. In the past two decades the treatment landscape of AML underwent significant changes due to explosive growth in knowledge of the molecular pathways involved in the AML origin and course evolution. This increased new data and understanding of the pathogenesis of AML, facilitated the development of new drugs in the treatment of AML, particularly the creation of drugs that target the disease on a molecular level. Encouraging efficacy of targeted therapy when combined with the traditional chemotherapy have resulted in big improvement to AML treatment and survival. In this chapter, we will discuss the drugs used in treatment of AML, including targeted treatment strategies that have recently entered the clinical practice.

1.1 Signaling and kinase pathway mutations inhibitors

1.1.1 FLT3 tyrosine kinase inhibitors

Full-length human FLT3 was cloned from a pre-B cell library in 1993 [1], from a CD34+ hematopoietic stem cell-enriched library [2], and is located on chromosome 13q12 [3]. FLT3 is a member of class III receptor tyrosine kinases (RTK) [4] and its activation leads to promotion of cell survival, proliferation, and differentiation through various signaling pathways, including PI3K, RAS, and STAT5 [5]. It is present in approximately 20–30% of adult AML patients and 5–15% of pediatric AML patients [6, 7]. FLT3-ITD mutations are associated with higher relapse rate and poorer overall survival, particularly with a high ratio of mutant allelic burden [8, 9]. Several first- and next generation FLT3 inhibitors have been investigated in patients with FLT3-ITD-mutated AML.

1.1.1.1 First-generation FLT3 inhibitors

1.1.1.1.1 Midostaurin

Midostaurin (Rydapt®, Novartis Pharmaceuticals, Inc. (PKC412)) is a multikinase inhibitor, targets wild type FLT3 and mutated FLT3 (ITD and tyrosine kinase domain (TKD)) [10]. Midostaurin also inhibits c-kit, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor (VEGFR), and protein kinase C [11]. A phase III international prospective, multinational, randomized, placebo-controlled, double-blind RATIFY study confirmed that, addition of midostaurin to standard induction chemotherapy could significantly increase OS vs. placebo among AML adults with FLT3 mutation (median OS of 74.7 m vs. 25.6 m, HR = 0.78, n = 717). RATIFY enrolled adults aged 18–59 years with newly diagnosed FLT3-mutated (ITD or TKD) AML. Patients were stratified according to FLT3 mutation type (TKD, ITD with a high AR [>0.7], and ITD with a low AR [0.05–0.7]) The addition of midostaurin demonstrated a significant survival benefit in patients with FLT3-mutated AML compared with placebo, with a 22% reduction in the rate of death compared with placebo (hazard ratio [HR], 0.78 [95% CI, 0.63–0.96]; P = 0.009). Although not specifically mandated, allogeneic stem cell transplantation (allo-SCT) was performed in 25% of patients in first complete remission (CR) and 57% of patients overall. Furthermore, patients receiving an allo-SCT in first CR had better outcome if they were treated with midostaurin during induction therapy (P = 0.08), suggesting that the optimal treatment strategy in FLT3-mutated AML would be to move on to allo-SCT early in first CR [12]. Adverse events (AEs) occurring during treatment with midostaurin were common in patients receiving intensive chemotherapy for AML. The most common nonhematologic grade ≥ 3 AEs were febrile neutropenia, infection, lymphopenia, diarrhea, and rash/desquamation. The rates of grade ≥ 3 AEs were largely similar between the midostaurin and placebo groups, with the exceptions of rash/desquamation and anemia (higher in the midostaurin group) and nausea (nearly twice as common in the placebo group) [12, 13]. Other, less common AEs reported with midostaurin included pulmonary AEs (i.e., pneumonitis or pulmonary infiltrate), cardiac AEs (e.g., prolonged corrected QT interval) and hepatic or renal dysfunction [12, 13, 14].

1.1.1.1.2 Sorafenib

Sorafenib is a potent first-generation multikinase inhibitor with activity against FLT3/ITD receptor but resistance emerges as FLT3-TKD point mutations [15]. It has been evaluated as either single agent or in combination with chemotherapies in numerous phase I and phase II clinical trials [16, 17, 18, 19, 20]. In an early phase clinical trial, sorafenib combined with idarubicin and high dose cytarabine in younger de novo AML patients provided a CR rate of 93% and a 1-year survival rate of 74% in FLT3-ITD positive AML patients [18]. In SORAML study, a placebo-controlled randomized study from Germany in 267 newly diagnosed patients aged 18–60 years. Sorafenib was added to daunorubicin and cytarabine (7 + 3) which resulted in a significantly prolonged 3-year EFS (40 vs. 22%, P = 0.013) and RFS (56 vs. 38%, P = 0.017) without improvement in OS and CR [21]. Another randomized placebo-controlled trial in 201 older patients aged 61–80 years did not demonstrated improvement in EFS, CR, and OS. The results showed higher early mortality (17 versus 7%, P = 0.052) compared with placebo [22]. In multivariate analysis of retrospective study analysing the effect of sorafenib as post-transplant maintenance in adult patients with FLT3-internal tandem duplication (ITD) acute myeloid leukaemia (AML) in 26 sorafenib patients and 55 controls, sorafenib significantly improved OS [Hazard ratio (HR) 0·26, P = 0·021] and PFS (HR 0·25, P = 0·016). Also there was no difference in 2-year non-relapse mortality (9·8% vs. 9·3%, P = 0·82) or 1-year chronic graft-versus-host disease (55·5% vs. 37·2%, P = 0·28) [23]. The most common adverse events occurring during treatment with sorafenib in SORAML study were fever, infections, pneumonia and pain. Grade 3 or worse adverse events that were significantly more common in the sorafenib group than the placebo group were fever, diarrhea, bleeding, cardiac events, hand-foot-skin reaction and rash [21].

1.1.1.1.3 Sunitinib

Sunitinib is an oral multitargeted kinase inhibitor with selectivity for FLT3, PDGFa/b, VEGF receptor, and Kit receptor tyrosine kinases [24]. Sunitinib induces G1 phase arrest, increases pro-apoptotic molecule expression, and decreases anti-apoptotic molecule expression in AML cells [25]. In a study by O’Farrell and colleagues in 29 AML patients each received a single dose of sunitinib, inhibition of FLT3 phosphorylation was observed in 50% of FLT3 wild-type patients and 100% of FLT3 mutated patients [26]. In another phase I study of sunitinib in 15 patients with refractory AML partial responses were achieved in all 4 patients with FLT3 mutations compared with 2 of 10 in patients with wild-type FLT3. All responses were of short duration and the most frequent grade 2 toxicities were edema, fatigue, and oral ulcerations occurring with a regimen of 50 mg/week [27]. In another phase I/II clinical trial, sunitinib combined with intensive chemotherapy included 22 patients older than 60 years with FLT3/ITD-mutated. Thirteen patients, including 8 patients with FLT3/ITD mutation, achieved CR/CRi. The median overall, relapse-free, and event-free survival of the 17 patients were 1.6, 1.0, and 0.4 years, respectively [28].

1.1.1.1.4 Lestaurtinib

Lestaurtinib (CEP-701) is an orally bioavailable first generation FLT3 inhibitor. It is derived from the bacterial fermentation product K-252a as indolocarbazole alkaloid compound. Except inhibition of FLT3 Lestaurtinib also inhibits JAK2, tropomyosin receptor kinases and neurotrophin receptors [29, 30, 31, 32, 33]. In a phase 2 trial lestaurtinib was administered as monotherapy in untreated older patients with AML not considered fit for intensive chemotherapy, irrespective of FLT3 mutation status. This study involved 29 patients with median age of 73 years. Lestaurtinib was administered orally at doses of 60 mg and 80 mg twice daily for 8 weeks. Clinical activity was evident in 8 (30%) patients, including 3 (60%) of 5 FLT3 mutant patients and 5 (23%) of 22 evaluable FLT3 WT patients, the difference in response rates between mutation groups not reaching statistical significance. Lestaurtinib was generally well tolerated. Commonly observed toxicities included mild nausea (8 patients), emesis (5 patients), constipation (5 patients), diarrhea (6 patients), and elevations in alkaline phosphatase concentration (13 patients) [34]. Another phase 1/2 trial in 14 heavily pretreated AML patients treated with CEP-701 at an initial dose of 60 mg orally twice daily, showed clinical evidence of biologic activity and measurable clinical response in 5 patients with significant reductions in bone marrow and peripheral blood blasts and minimal drug related toxicities [35]. A randomized assessment from UK AML 15 and AML 17 trials confirmed no statistically significant benefit observed in the combination of lestaurtinib with standard chemotherapy for newly diagnosed AML patients mostly younger than 60 years.

1.1.1.1.5 Tandutinib

Tandutinib (MLN518) is FLT3, KIT, PDGFR and type III receptor tyrosine kinases inhibitor. Tandutinib induces apoptosis and inhibits FLT3/ITD phosphorylation, cellular proliferation, and signaling of the MAPK and PI3K pathways [36]. In a phase 1 trial tandutinib was given orally (from 50–700 mg twice daily) in 40 patients with either AML or high-risk myelodysplastic syndrome (MDS) with only 8 patients with FLT3-ITD mutations. Even among the patients with FLT3-ITD mutations who were treated at potentially effective doses, response evaluation was often not possible because of rapid disease progression, sudden disease-related clinical deterioration, or tandutinib-related toxicity. Tandutinib treatment was associated muscular weakness, nausea, vomiting and less often diarrhea [37].

1.1.1.2 Second and next generation FLT3 inhibitors

1.1.1.2.1 Quizartinib

Quizartinib (AC220) is a selective and highly potent second-generation class III receptor TKI that selectively inhibits FLT3/STK1, CSF1R/FMS, SCFR/KIT, and PDGFRs in a dose dependent manner [38]. Quizartinib was first tested in phase I dose-escalation trial in patients with relapsed and refractory AML patients irrespective of FLT3 mutation status. Quizartinib was administered orally at escalating doses of 12 to 450 mg/day to 76 patients, with a median of three prior therapies and responses occurred in 23 (30%) of 76 patients, including 10 (13%) complete remissions (CR). The median duration of response was 13.3 weeks and the median survival was 14 weeks. The most common treatment-related adverse events were nausea, vomiting, and prolonged QT interval. The maximum tolerated dose (MTD) was 200 mg/day, and the dose-limiting toxicity was grade 3 QT prolongation [39]. In another 2-part, phase 1, multicenter, open-label, sequential group dose-escalation trial of quizartinib in combination with induction and consolidation chemotherapy in patients with newly diagnosed acute myeloid leukemia a total of 19 patients were enrolled. Sixteen patients (84%) achieved a response; 14 (74%) composite complete response; 2 (11%) morphologic leukemia-free state. Most common grade 3/4 adverse events were febrile neutropenia (47%), neutropenia (42%), thrombocytopenia (32%), and anemia (26%). There were no apparent additional toxicities with addition of quizartinib to chemotherapy although grade ≤ 1 QT prolongation was observed at MTD [40]. In a large phase 2 trial assigning 333 (157 in cohort 1 and 176 in cohort 2) r/r AML patients were enrolled. In cohort 1 56% of FLT3-ITD-positive patients and 36% of FLT3-ITD-negative patients achieved composite complete remission and in cohort 2 46% FLT3-ITD-positive patients achieved composite complete remission whereas 30% of FLT3-ITD-negative patients achieved composite complete remission. Across both cohorts the most common grade 3 or worse treatment-related adverse events were febrile neutropenia, anaemia, thrombocytopenia, QTcF prolongation, neutropenia, leucopenia thrombocytopenia and pneumonia [41]. Preliminary results of a randomized phase 3 study (QuANTUM-R) in patients with FLT3-ITD mutated r/r AML enrolled 367 patients, randomized 2:1 to receive quizartinib or 1 of 3 preselected investigator’s choice therapy.(low-dose cytarabine, mitoxantrone, etoposide, and intermediate-dose cytarabine or fludarabine, cytarabine, and granulocyte-colony stimulating factor with idarubicin). The results showed a significantly improved median OS for quizartinib (6.2 vs. 4.7 months; p = 0.017) and an improved cCR rate (48% vs. 27%, p = 0.0001). Rates of treatment-emergent adverse events were comparable between the 2 arms [42].

1.1.1.2.2 Crenolanib

Crenolanib (CP868596) is a benzamidine quinolone derivative, a second generation RTK inhibiting FLT3-ITD and -TKD mutations. It is potent, selective, and invulnerable to resistance-conferring kinase domain mutation. As a type I pan-FLT3 inhibitor crenolanib inhibits FLT3/WT, FLT3/ITD, FLT3-TKD, PDGFRα/β, KIT, and FLT3/D835 [43]. In nearly one third of AML patients treated with FLT3 inhibitors in different clinical trials a resistance point mutations like D835 and F691 are occurring during disease progression [44, 45]. As a more potent RTK inhibitor crenolanib could inhibit both FLT3/ITD and resistant FLT3/D835 mutants and less disruptive of erythroid colony growth, which may result in relatively lessmyelosuppression [46]. In a phase 1 trial of FLT3-ITD positive AML crenolanib was given to 69 patients divided in three cohorts, cohort A patients with R/R FLT3 AML who had not received prior FLT3 inhibitors, cohort B patients progressing on prior TKIs and cohort C patients who developed FLT3 + AML after prior MDS. Crenolanib therapy resulted in a 39% CRi and 11% PR (6 D835, 9 ITD, 3 ITD + D835) amongst the patients in cohort A with an ORR of 50% [47]. In a phase II trial, the tolerability and efficacy of crenolanib combined with standard 7 + 3 induction chemotherapy was examined in 29 patients with newly diagnosed FLT3 mutant AML. 21 of 29 (72%) patients achieved a CR after one cycle of induction with cytarabine/anthracycline/crenolanib. An additional 3 patients achieved a CR either after re-induction (1 patient) or after treatment with HiDAC or HSCT (1 patient each) [48]. In another study with the same inclusion criteria in which 26 patients were enrolled the most common advere events which led to crenolanib dose reductions were periorbital edema, delayed count recovery, LFT elevation, nausea and rash [49]. Also crenolanib was investigated in a patients with a first relapsed/primary refractory AML. The study enrolled 8 patients, received HAM followed by crenolanib. 6 patients were evaluable for responses with a complete remission rate of 67% (2 CR, 2 CRi), including 2 pts who were refractory to front line chemotherapy. 2 of 3 patients with FLT3 activating mutations (1 with ITD and 1 with D835) achieved complete remission with complete count recovery; the third pt (FLT3-ITD) had 10% residual blasts after 1 cycle of induction [50]. Ohanian et al. studied the effect of crenolanib combined with idarubicin and high-dose Ara-C in 13 pts (median age of 51 years) with multiply relapsed/refractory FLT3+ AML. The ORR in 11 pts evaluable for response was 36% (1 CR, 3 CRi; 2 not evaluable because of early discontinuation of therapy). The median OS for all patients was 259 days; median OS by prior therapies was 259 days for patients with ≤2 prior therapies, and 53 days for patients with ≥3 prior therapies. No dose-limiting toxicities were observed at any of the dose levels explored and there were no dose reductions required. Non-hematologic adverse events assessed as possibly or probably related to crenolanib were all grade 1 in severity, including: nausea, vomiting, diarrhea, and abdominal pain [51].

1.1.1.2.3 Gilteritinib

Gilteritinib (ASP2215) is a selective next-generation novel dual FLT3 (to a lesser extent to FLT3-TKD than -ITD)/AXL inhibitor. Gilteritinib was investigated in 252 patients with relapsed or refractory acute myeloid leukaemia in one of seven dose-escalation (n = 23) or dose-expansion (n = 229) cohorts. At least 90% of FLT3 phosphorylation inhibition was seen by day 8 in most patients receiving a daily dose of 80 mg or higher. In the full analysis 249 patients were included, 8% achieved complete remission, 4% complete remission with incomplete platelet recovery, 18% complete remission with incomplete haematological recovery, and 10% partial remission. The most common grade 3–4 adverse events irrespective of relation to treatment were febrile neutropenia (39%), anaemia (24%), thrombocytopenia (13%), sepsis (11%), and pneumonia (11%) [52]. In another open-label phase 1 study in 24 Japanese patients with relapsed/refractory acute myeloid leukemia the ORR among patients with mutated FLT3 was 80% and among FLT wild-type was 36.4%. The MTD was 200 mg/d, dose-limiting toxicities were grade 3 tumor lysis syndrome and grade 3 elevated blood lactate dehydrogenase, amylase, blood creatine phosphokinase levels, and syncope [53]. The phase 3 ADMIRAL trial assessing oral gilteritinib 120 mg per day versus salvage chemotherapy in adult r/r FLT3 mutated AML patients led to an FDA approval for gilteritinib. 369 adults with FLT3 mutated AML in first relapse or refractory to front-line therapy were enrolled. The 21% of patients who achieved had a median time to response of 3.6 months [54].

1.1.1.2.4 Cabozantinib

Cabozantinib is an oral tyrosine kinase inhibitor of multiple receptor tyrosine kinases and exhibits anti-tumor activity in several cancers [55, 56]. It inhibits FLT3, MET, AXL, vascular endothelial growth factor receptor, and KIT. In a study among 18 patients with relapsed/refractory AML, 5 harboring FLT3/ITD mutations no patients had a marrow response according to formal criteria. 4 patients had peripheral blast reductions, 2 of these 4 patients transiently cleared circulating blasts, 1 patient experienced a reduction in marrow blasts, and 1 had stable disease [57] (Table 1).

Drug ClassMechanism of actionAgent
Signaling and kinase pathway mutations inhibitors (FLT3 tyrosine kinase inhibitors)
First-generation FLT3 inhibitorsInhibition of FLT3
Midostaurin (PKC412)
Sorafenib
Sunitinib
Lestaurtinib
Tandutinib
Second and next generation FLT3 inhibitors
Quizartinib
Crenolanib
Gilteritinib
Cabozantinib
Mutations in epigenetic modifiers: Regulators of DNA methylation and chromatin modification drugs (IDH inhibitors)Inhibition of IDH2Enasidenib
Inhibition of IDH1Ivosidenib
Pro-apoptotic agents (Bcl-2 inhibitors)Inhibition of BCL2Venetoclax
Hedgehog Inhibition (Smoothened inhibitors)inhibition of of HH/GLI signallingGlasdegib
Sonidegib
Vismodegib
Polo-like kinase inhibitorsInhibition of Plk1Volasertib
E-selectin inhibitorsInhibition of E-selectinUproleselan
Checkpoint inhibitorsInhibition of PD-1Nivolumab
Pembrolizumab
Inhibition of CTLA-4Ipilimumab
CD33-targeted therapyInhibition of CD33Gemtuzumab Ozogamicin
Vadastuximab talirine

Table 1.

Selected AML targeted agents.

1.2 Mutations in epigenetic modifiers: regulators of DNA methylation and chromatin modification drugs

1.2.1 IDH inhibitors

For the first time IDH1 mutations in AML were identified in 2009 by sequencing an acute myeloid leukemia genome. IDH is an enzyme that catalyzes the oxidative decarboxylation of isocitrate to alpha-ketoglutarate (α-KG). 5-methylcytosine (5mC) coverts to 5-hydroxymethylcytosine (5hmc) as a result of interaction between α-KG and TET2 which promotes DNA and histone demethylation [58]. Approximately 8–19% of AML cases carry IDH2 mutations, with another 7–14% carrying IDH1 mutations [59]. IDH1/2 are found with higher frequency in older patients and patients with a normal karyotype [60, 61]. IDH1 mutations almost exclusively occur at R132 while IDH2 involve substitutions at R140 or R172 [62]. While IDH2-R172 may represent a distinct genomic subgroup, which mutual exclusivity with NPM1 and with a distinct DNA methylation profile [63]. Some studies showed that IDH1 and IDH2-R172 mutation may predict a worse clinical outcome especially in CN-AML, while the IDH2-R140 concomitant NPM1 mutation may be associated with better prognosis in AML [63, 64, 65]. More further studies are needed due to the conflicting data about the prognostic impact of IDH1/2 mutations in AML.

1.2.1.1 Enasidenib

Enasidenib (AG-221) is the first oral IDH mutation–specific inhibitor. It is a bivalent inhibitor of R140Q and R172K mutated IDH2 and induces terminal differentiation of leukemic blasts into neutrophils in vivo [66]. IDH2 inhibitor enasidenib (AG-221/CC-90007) showed promising activity as a single agent in patients with mutated IDH2 in first-in-human phase 1/2 study with 345 patients enrolled. Median age was 68 years. 214 of 345 patients (62%) with relapsed or refractory (R/R) AML received enasidenib, 100 mg/d. 19.6% attained complete remission, 10.3% proceeded to an allogeneic bone marrow transplant, and the overall response rate was 38.8%. 43.1% red blood cell transfusion–dependent and 40.2% platelet transfusion–dependent patients achieved transfusion independence. Response and survival were comparable among patients with IDH2-R140 or IDH2-R172 mutations. Among all 345 patients, the most common grade 3 or 4 treatment-related adverse events were hyperbilirubinemia (10%), thrombocytopenia (7%), and IDH differentiation syndrome (6%) [67]. These results led to the FDA approval of enasidenib in r/r IDH2 mutated AML patients on 1 August 2017. With regard to predictors of response, the IDH2 mutation allele burden at study entry had no effect on response rate [66]. In a open-label, multicenter, phase 1 study patients 134 newlydiagnosed mIDH1 or mIDH2 AML were treated with induction therapy in combination with either ivosidenib 500 mg once daily (for mIDH1) or enasidenib (mIDH2) 100 mg daily. Among the 77 enasidenib-treated patients evaluable for efficacy, a response of CR, CRi, or CRp was achieved in 73% patients with de novo AML and in 63% patients with sAML. The most frequent co-occurring baseline mutations for patients with IDH2 mutations were DNMT3A, SRSF2 and ASXL1 [68].

1.2.1.2 Ivosidenib

Ivosidenib (AG-120) is a potent and selective IDH1 mutation small-molecule inhibitor. In phase 1, multicenter, open-label, dose-escalation and dose-expansion study 258 patients received ivosidenib orally, daily, in 28-day cycles. In the primary efficacy population (125 patients), the rate of complete remission or complete remission with partial hematologic recovery was 30.4%, the rate of complete remission was 21.6% and the overall response rate was 41.6%. The median durations of these responses were 8.2 months, 9.3 months, and 6.5 months, respectively. No residual detectable IDH1 mutations on digital polymerase-chain-reaction assay were detected in 21% patients who had a complete remission or complete remission with partial hematologic recovery. The most common adverse events (in ≥20% of the patients), irrespective of a relationship to ivosidenib, were diarrhea, leukocytosis, febrile neutropenia, nausea, fatigue, dyspnea, prolongation of the QT interval. Peripheral edema, anemia, pyrexia, cough and and differentiation syndrome [69]. The results of this study led to the FDA approval of ivosidenib in r/r IDH1 mutated AML patients on 2 may 2019. Prescribing information contains a boxed warning about the risk of differentiation syndrome which may be life-threatening or fatal. In a open-label, multicenter, phase 1 study patients 134 newlydiagnosed mIDH1 or mIDH2 AML were treated with induction therapy in combination with either ivosidenib 500 mg once daily (for mIDH1) or enasidenib (mIDH2) 100 mg daily. Among the 41 ivosidenib-treated patients evaluable for efficacy, a response of CR, CRi or CRp was achieved in 93% patients with de novo AML and 46% patients with sAML. Twenty-one patients received ≥1 cycle of consolidation therapy and 11 patients received maintenance after consolidation. Seventeen patients proceeded to HSCT. For patients with IDH1 mutations the most frequent co-occurring baseline mutations were DNMT3A, NPM1 and NRAS. MRD-negative CRs using flow cytometry were observed in 89% of patients with IDH1 positive mutational status [68].

1.3 Pro-apoptotic agents

1.3.1 Bcl-2 inhibitors

Bcl-2 gene is located on chromosome 18q21.33 and it was discovered in 1985 through cloning the breakpoint of a translocation of t(14;18) found in follicular B lymphomas [70]. Bcl-2 is an integral protein of the mitochondrial membrane but has also been identified on endoplasmic reticulum and the nuclear envelope [71]. BCL2 family members are classified into pro and anti-apoptotic proteins. The anti-apoptotic BCL2 family contains 4 proteins: BCL2, BCLXL, BCL-w, and MCL-1. Through direct activation of the effector proteins or antagonizing the effect of antiapoptotic proteins the pro-apoptotic proteins lead to an activation of caspase proteases [72]. Bcl 2 has proven to be major negative regulator in apoptosis, playing key roles in neoplastic transformation and leukemogenesis [73]. Overexpression of anti-apoptotic BCL2 proteins such as BCL2, BCL2L1 and MCL1 is widely associated with tumour initiation, progression and chemo resistance in AML [74].

1.3.1.1 Venetoclax

Venetoclax (ABT-199/GDC- 0199) is a highly selective oral BCL2 inhibitor and does not show significant BCL-XL antagonism [75]. Venetoclax induces apoptosis in AML cell lines, in-vitro patient samples and in mouse xenograft models [75, 76]. In a phase II, single-arm study in 32 patients with high-risk relapsed/refractory AML or unfit for intensive chemotherapy venetoclax was given at a dose of 800 mg daily. The overall response rate was 19%, an additional 19% of patients demonstrated antileukemic activity not meeting IWG criteria (partial bone marrow response and incomplete hematologic recovery). Twelve (38%) patients had IDH 1/2 mutations, of whom 4 (33%) achieved complete response or complete response with incomplete blood count recovery. The responses median progression free interval was 2.5 months. Common adverse events included nausea, diarrhea, febrile neutropenia and hypokalemia. Due to potential tumour lysis syndrome as seen in chronic lymphocytic leukemia, a daily dosing ramp up of venetoclax was executed until 800 mg per day [77]. More effective results were achieved in studies where venetoclax was combined with either low-dose cytarabine (LDAC) or hypomethylating agents (HMAs). In a phase Ib/II study in previously untreated patients with AML venetoclax was combined with low-dose cytarabine in 82 adults 60 years or older. The median age was 74 years, 49% had secondary AML, 29% had prior HMA treatment, and 32% had poor-risk cytogenetic features. 54% achieved complete remission (CR)/CR with incomplete blood count recovery. The median OS was 10.1 months (95% CI, 5.7 to 14.2), and median duration of response (DOR) was 8.1 months (95% CI, 5.3 to 14.9 months). Early (30-day) mortality was 6% [78]. In another phase 1b study of venetoclax plus decitabine or azacitidine in untreated AML patients ≥65 years ineligible for standard induction therapy 145 patients were enrolled. Median age was 74 years, with poor-risk cytogenetics in 49% of patients. With a median time on study of 8.9 months, 67% of patients (all doses) achieved complete remission (CR) + CR with incomplete count recovery (CRi), with a CR + CRi rate of 73% in the venetoclax 400 mg + HMA cohort. No tumor lysis syndrome was observed. Common adverse events (>30%) included nausea, diarrhea, constipation, febrile neutropenia, fatigue, hypokalemia, decreased appetite, and decreased white blood cell count [79]. Due to these marked results venetoclax received FDA approval for combination with low dose cytarabine and HMAs. DiNardo et al. assessed the safety and efficacy of venetoclax in combination with FLAG-IDA in a heavily pre-treated r/r AML patients. Study included 12 patients, of 11 patients, 8 patients (73%) achieved a best response of CR/CRi (7 CR, 1 CRi) with a 6-month survival rate of 67%. Of the 8 responding patients, three patients proceeded to allogeneic SCT [80].

1.4 Hedgehog inhibition

1.4.1 Smoothened inhibitors

The Hedgehog (Hh) family of proteins control cell growth and survival. The Hedgehog signalling pathway (HhP) is essential for embryonic development and usually silenced in adult tissues. Germline mutations that subtly affect Hh pathway activity are associated with developmental disorders, whereas somatic mutations activating the pathway have been linked to multiple forms of human cancer [81, 82, 83]. Aberrant activation of the HhP has been implicated in the maintenance of leukaemia stem cells in several model systems. Overexpression of various HH/GLI components have been found in chemotherapy resistant myeloid blasts and subsequent inhibition of the HH/GLI pathway revised the sensitivity to chemotherapy [84, 85].

1.4.1.1 Glasdegib

Glasdegib (PF-913) is an oral, potent, selective, small molecule inhibitor of HH/GLI signalling, which binds to the smoothened (SMO) receptor [86]. In vitro treatment with Glasdegib induced a decrease in the quiescent cell population and in vivo treatment attenuated the leukemia-initiation potential of AML cells in a serial transplantation mouse model [87]. An open-label, dose-finding, phase 1 study of glasdegib in 47 adult patients with myeloid malignancies (AML, n = 28) found 400 mg once daily as the MTD and a minor response was achieved (over 25% decrease from baseline in BM blasts) or better in more than 30% of AML patients. The most common treatment-related adverse events included dysgeusia, decreased appetite, and alopecia [88]. Based on this study a phase II, randomized, open-label, multicenter study evaluated the efficacy of glasdegib plus low-dose cytarabine (LDAC) in patients with AML or high-risk myelodysplastic syndrome unsuitable for intensive chemotherapy. Glasdegib 100 mg was administered orally in 28-day cycles. Eighty-eight and 44 patients were randomized to glasdegib/LDAC and LDAC, respectively. Median overall survival was 8.8 (6.9–9.9) months with glasdegib/LDAC and 4.9 (3.5–6.0) months with LDAC. Fifteen (17.0%) and 1 (2.3%) patients in the glasdegib/LDAC and LDAC arms, respectively, achieved complete remission (P < 0.05) [89]. Based on this study the FDA approved glasdegib in combination with low dose cytarabine for AML patients unfit for IC. Another phase-II study to evaluate glasdegib from day 3 plus standard-induction chemotherapy for untreated and fit AML or high-risk MDS patients revealed that 46.4% of patients achieved CR. Among all 69 patients, median OS was 14.9 (80% CI 13.4–19.3) months, with 12-month survival probability 66.6%. The most common treatment-related adverse events (≥50% patients) were diarrhea and nausea [90].

1.4.1.2 Sonidegib

Sonidegib (LDE225) is a specific SMO inhibitor and in refractory AML cells increased cell apoptosis and the efficacy of Adriamycin against tumor cells and lowered the expression of the targeted protein [91]. In a phase I/Ib study of azacitidine and sonidegib in myeloid malignancies the best response outcome for untreated AML/MDS patients was 23.1% and for rel/ref 7.1%. However, the rate of SD was remarkably high particularly in the rel/ref AML population at 76%. The most common Gr 3/4 AEs were: thrombocytopenia, neutropenia, anemia and leukopenia [92].

1.4.1.3 Vismodegib

Vismodegib (GDC-0449) safety and efficacy were evaluated in a phase Ib trial in patients with relapsed/refractory acute myeloid leukaemia. All enrolled patients had received prior cancer therapy; most had received more than one therapy, including hypomethylating agents, immunomodulators and targeted signalling pathway inhibitors. 38 received at least one dose of vismodegib but the study was terminated by the sponsor because of lack of efficacy [93].

1.5 Polo-like kinase inhibitors

Polo-like kinases (Plks) are a family of 5 highly conserved serine/threonine protein kinases. They play a key role in mitotic checkpoint regulation and cell division. Plk1 has been shown to be overexpressed in a range of human cancers, including non-small cell lung cancer, prostate, ovarian, breast, and colorectal cancer as well as AML [94, 95, 96].

1.5.1 Volasertib

Volasertib (BI 6727) is a low-molecular-weight, adenosine triphosphate–competitive kinase inhibitor that potently inhibits Plk1 [97]. In a randomized, phase 1/2, open-label, multicenter trial of low-dose cytarabine with or without volasertib in patients with AML ineligible for intensive induction therapy eighty-seven patients (median age 75 years) received LDAC or LDAC + volasertib. The result confirmed greater clinical efficacy in the combination arm, statistically significant in CR (30 vs. 13.3%, P = 0.052). Median overall survival was 8.0 vs. 5.2 months, respectively. LDAC + volasertib led to an increased frequency of adverse events that was most pronounced for neutropenic fever/infections and gastrointestinal events [98].

1.6 E-selectin inhibitors

The endothelial cell adhesion molecule E-selectin is a key component of the bone marrow hematopoietic stem cell (HSC) vascular niche regulating balance between HSC self-renewal and commitment. E-selectin is expressed transiently in the normal vasculature during an inflammatory response and constitutively in the bone marrow. E-selectin directly triggers signaling pathways that promote malignant cell survival and regeneration. In vivo AML blasts with highest E-selectin binding potential are 12-fold more likely to survive chemotherapy and main contributors to disease relapse [99, 100].

1.6.1 Uproleselan

Uproleselan (GMI-1271) is a novel antagonist of E-selectin that down-regulates cell survival pathways and enhances chemotherapy response. In a single arm phase I/II trial of 47 adults with relapsed/refractory AML were treated with GMI-1271 in combination with MEC chemotherapy. GMI-1271 was given 24 hrs prior, then every 12 hrs during and for 48 hrs post induction/consolidation. With a median follow-up of 11 months, the ph 1 median leukemia free survival was not reached and overall survival was 7.6 months. ORR (CR/CRi/MLFS/PR) was evaluable in 21 patients was 50%. Remission rate (CR/CRi) was 45%.Common Gr 3/4 AEs were febrile neutropenia, sepsis, bacteremia, hypoxia. 30 and 60 day mortality were 0 and 7%, respectively [101]. A pivotal phase 2/3 study (NCT03616470) is underway to assess the efficacy and safety of uproleselan with standard salvage chemotherapy in R/R AML. The study is a global, randomized, double-blind, phase 3 trial in adults aged 18–75 years with R/R AML and fit for chemotherapy [102].

1.7 Checkpoint inhibitors

The development of immune checkpoint inhibitors (ICIs) is a revolutionary milestone in the field of immuno-oncology. Immune checkpoint blockade removes inhibitory signals of T-cell activation, which enables tumor-reactive T cells to overcome regulatory mechanisms and mount an effective antitumor response [103, 104, 105]. Recent studies suggest a novel mechanism that tumor cells might evade host immune attack through increased expression of PD-L1 [106]. Immune checkpoint inhibitor treatment involves programmed cell death protein 1 (PD1) and cytotoxic Tlymphocyte- associated antigen 4 (CTLA4), both of which have been used in preclinical AML models [107].

1.7.1 Nivolumab

Nivolumab (BMS-936558, ONO-4538, or MDX1106) is the first-in-human immunoglobulin G4 (IgG4) PD-1 immune checkpoint inhibitor antibody that disrupts the interaction of the PD-1 receptor with its ligands PD-L1 and PD-L2, thereby inhibiting the cellular immune response [108, 109]. In a phase IB/II study of nivolumab in combination with azacytidine in patients (pts) with relapsed AML 51 patients with a median age of 69 years (range 45–90) were included. From 35 patients evaluable for response, 6 (18%) achieved complete remission (CR)/(CRi) (3 CR, 3 CRi), 5 (15%) had hematologic improvement (HI), 9 (26%) had 50% BM blast reduction,3 pts. (9%) had stable disease >6 months, and 12 (34%) had progression. In the subgroup of patients who did not receive HMA prior treatment, the superiority of new regimen was even more evident with ORR at 52–22%. The median overall survival for the 35 evaluable pts was 9.3 months (range, 1.8–14.3). Grade 3/4 and Grade 2 immune mediated toxicities were observed in 7 (14%) and 6 (12%) patients, which included pneumonitis, nephritis, transaminitis, and skin rash. Steroids took effect on 88% of the patients who suffered from drug-related toxicities [110]. A single-arm, phase 2 part of the phase 1–2 study of nivolumab in combination with idarubicin and cytarabine was conducted in 44 patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome. The median overall survival was 18.54 months and median event-free survival was not reached. Among the 44 evaluable patients, the ORR was 77% including 63% CR and 14% CRi. Concerning drug toxicities, the grade 3–4 adverse events were observed in six patients, including rash, colitis, pancreatitis and transaminitis [111]. Using nivolumab in post-transplantation setting showed limited efficacy. Among three relapsed AML patients after allo-HSCT treated with nivolumab, one achieved CR, one experienced stabilization, and the third failed to respond [112].

1.7.2 Pembrolizumab

Pembrolizumab (MK-3475) is another drug that blocks PD-1. In a multicenter phase II study pembrolizumab was administered after high-dose cytarabine salvage chemotherapy in 26 R/R AML patients with median age of 54. The overall response rate was 42% with 9 CR/CRi, one PR, and one patient with morphologic leukemia free state. The median OS was 10.5 months. Most frequently observed grade 3 AEs included hepatitis, rash, and epigastric pain [113]. In another single center, single arm trial of pembrolizumab followed by decitabine in 10 patients R/R AML patients with median age of 62, the ORR was 20% with one patient achieving MRD-negative CR. With a median follow-up of 13 months, the mOS was 7 months [114].

1.7.3 Ipilimumab

Ipilimumab is a human IgG1 monoclonal antibody, CTLA-4 antagonist. In a phase I/Ib, open label, multicenter study of treating patients with relapsed hematological malignancies after allo-SCT with ipilimumab 12 AML patients were enrolled. Complete response was observed in five patients (23%) and the 1-year survival rate was 49%. Immune-related adverse events occurred in three patients. Response was associated with in situ infiltration of CD8+ T cells as well as enrichment of effector T cell subsets [115].

1.8 CD33-targeted therapy

The CD33 antigen is expressed on the blast cells (85–90%) of most cases of acute myeloid leukemia [116, 117, 118]. CD33 seems to be much less expressed on normal hematopoietic stem cells and has decreased expression during the differentiation of the myeloid lineage. Mature granulocytes do not express a significant amount of CD33. That makes CD33 an promising target for AML targeted therapy [119, 120]. The only non-haematopoietic cells expressing CD33 are hepatocytes, which explains to some extent hepatic toxicity induced by anti CD33 antibodies [121, 122].

Advertisement

2. Gemtuzumab ozogamicin (GO)

Gemtuzumab ozogamicin (Mylotarg) is a humanized anti-CD33 IgG4 monoclonal antibody conjugated to a cytotoxic agent N-acetyl gamma calicheamicin via an acid-labile hydrazone linker. After GO binds to CD33, calicheamicin is being released and generates single and double strand breaks with subsequent cellular death [123]. In a phase I dose escalation study of an anti-CD33 calicheamicin immunoconjugate 40 patients with relapsed or refractory CD33(+) AML were treated. Leukemia was eliminated from the blood and marrow of 8 (20%) of the 40 patients and the MTD was determined to be 9 mg/m2 [124]. After the encouraging results from three open label phase II studies, the FDA approved GO for the treatment of patients with CD33-positive AML in first relapse who were ≥ 60 years and not suitable for intensive chemotherapy. 142 patients with AML in first relapse were enrolled in the studies with median age of 61 years. All patients received Mylotarg as a 2-hour intravenous infusion, at a dose of 9 mg/m2, at 2-week intervals for two doses. 30% of patients obtained complete morphological remission. High incidences of myelosuppression, grade 3 or 4 hyperbilirubinemia, and elevated hepatic transaminase levels were registered [125]. In a post-approval phase III trial gemtuzumab ozogamicin was administred during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. 637 patients were randomly assigned to receive daunorubicin, cytarabine, and GO vs. standard induction therapy with daunorubicin and cytarabine alone. The CR rate was 69% for DA + GO and 70% for DA (P = 0.59). In this study, the addition of GO to induction or postconsolidation therapy failed to show improvement in CR rate, disease-free survival, or overall survival. Also addition of GO was associated with a higher early mortality during induction (5.5% vs. 1.4%). Major causes of death were fatal hemorrhage and infection. Based on these negative results GO was withdrawn from the market in 2010 [126]. In a subsequent trials different schedules of GO were investigated. Phase 3, open-label study enrolled 280 patients aged 50–70 years with previously untreated de novo AML. Patients were randomly assigned in a 1:1 ratio to standard treatment (control group) with or without five doses of intravenous gemtuzumab during induction and day 1 of each of the two consolidation chemotherapy courses. Although the CR rates were similar between the two arms, IC plus GO provided a significantly improved median event free survival (EFS) (19.6 vs. 11.9 months, p = 0.00018) and median OS (34 vs. 19.2 months, p = 0.046). Haematological toxicity, particularly persistent thrombocytopenia, was more common in the GO group than in the control group, without an increase in the risk of death from toxicity [127]. A meta-analysis of five open label, phase 3 trials comprising 3325 AML patients found that the addition of gemtuzumab ozogamicin significantly reduced the risk of relapse and improved overall survival at 5 years without increased toxicity for GO treatment [128]. Based on these results FDA approved GO for the treatment of adults with newly diagnosed CD33-positive AML on 1 September 2017, and also approved Mylotarg for the treatment of patients aged 2 years and older with relapsed or refractory CD33-positive AML.

Advertisement

3. Vadastuximab talirine

Vadastuximab talirine (SGN-CD33A) is a novel anti-CD33 mAb conjugated to 2 molecules of pyrrolobenzodiazepine (PBD). After internalization vadastuximab where transported to the lysosomes where the PBD dimer is released via proteolytic cleavage of the linker, crosslinking DNAand leading to apoptosis. Vadastuximab is highly stable in circulation with relatively less off-target toxicity compared to GO [129]. In a dose-escalation phase 1 study 27 treatment naive patients with CD33 positive AML and median age of 74 years were treated with vadastuximab talirine. Of the 26 efficacy evaluable treatment naive patients, 6 patients achieved CR, 8 patients CRi, and 5 patients achieved a morphologic leukemia-free state [130]. In another phase 1 trial of vadastuximab talirine as monotherapy in patients with CD33-positive AML a total of 131 patients, median age, 73 years were enrolled. The CR + CRi rate was 28%, 50% of patients who responded achieved minimal residual disease negativity. Most AEs were consistent with myelosuppression, nonhematological included fatigue, nausea, and diarrhea [131]. Also vadastuximab talirine was added to a 7 + 3 induction therapy in a phase 1b study in 42 patients with a median age of 45.5 years. The CR/CRi rate was 78%. Twenty-three of 31 (74%) patients attaining CR or CRi achieved MRD negative status. No 30-day mortality or significant hepatotoxicity was observed [132]. Fathi et al. combined vadastuximab talirine with hypomethylating agents in patients with CD33-positive AML. Among 53 patients treated, the median age was 75 years. The CR + CRi rate was 70% and 51% of remissions with minimal residual disease-negative status by flow cytometry. The majority of adverse events were a result of myelosuppression, with some causing therapy delays [133]. A phase III trial (CASCADE, NCT02785900) comparing HMA with or without SGN-CD33A in elderly patients with newly diagnosed AML was terminated because of due to safety reasons, specifically a higher rate of deaths, including fatal infections, in the SGN33A arm versus the control arm.

Advertisement

4. Future perspectives

The great advances in understanding molecular mechanisms of AML as well as their prognostic significance have changed the therapeutic armamentarium against AML. The drugs discussed in this chapter and many novel molecules being evaluated in clinical trials are on their way to change the current standard of treatment in AML. Ongoing efforts to understand the heterogeneity of AML, the constantly changing genomic landscape, the mechanisms of resistance/refractoriness will be very important in the development of new drugs. The rational use of these drugs, their potency that might be improved by combining them with other modes of therapy will hopefully increase long-term benefits for patients with AML. Furthermore, the development of novel ultrasensitive methods for minimal residual disease detection will also refine the treatment decision making process and probably improve the survival rates. However, new issues such as extrapolation of the results from the clinical trials enrolling carefully selected patients to general practice, the access and cost of these new drugs must be considered in the treatment decision process. It is a question of time whether personally tailored therapeutic era is going to be the new standard in the treatment of AML. Till then an increased enrolment of patients with AML into clinical trials evaluating the safety and efficacy of these new drugs and combinations is strongly encouraged and recommended.

Advertisement

Declaration of conflicting interest

The authors declare that there is no conflict of interest.

References

  1. 1. Rosnet O, Schiff C, Pébusque MJ, Marchetto S, Tonnelle C, Toiron Y, et al. Human FLT3/FLK2 gene: cDNA cloning and expression in hematopoietic cells. Blood. 1993;82:1110-1119
  2. 2. Small D, Levenstein M, Kim E, Carow C, Amin S, Rockwell P, et al. STK-1, the human homolog of Flk-2/Flt-3, is selectively expressed in CD34+ human bone marrow cells and is involved in the proliferation of early progenitor/stem cells. Proc Natl Acad Sci USA. 1994;91:459-463
  3. 3. Rosnet O, Matteï MG, Marchetto S, Birnbaum D. Isolation and chromosomal localization of a novel FMS-like tyrosine kinase gene. Genomics. 1991;9:380-385. DOI: 10.1016/0888-7543(91)90270-O
  4. 4. Kelly LM, Gilliland DG. Genetics of myeloid leukemias. Annual Review of Genomics and Human Genetics. 2002;3:179-198
  5. 5. Grafone T, Palmisano M, Nicci C, Storti S. An overview on the role of FLT3-tyrosine kinase receptor in acute myeloid leukemia: biology and treatment. Oncology Reviews. 2012;6:e8
  6. 6. Levis M. FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematol Am Soc Hematol Educ Program. 2013;2013:220-226
  7. 7. Pui CH, Carroll WL, Meshinchi S, et al. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. Journal of Clinical Oncology. 2011;29:551-565
  8. 8. Schlenk RF, Kayser S, Bullinger L, Kobbe G, Casper J, Ringhoffer M, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124:3441-3449
  9. 9. Wang Y, Xu Y, Li S, Liu J, Xing Y, Xing H, et al. Targeting FLT3 in acute myeloid leukemia using ligand-based chimeric antigen receptor-engineered T cells. Journal of Hematology & Oncology. 2018;11:60
  10. 10. Wang Y, Yin OQ , Graf P, Kisicki JC, Schran H. Dose- and time-dependent pharmacokinetics of midostaurin in patients with diabetes mellitus. Journal of Clinical Pharmacology. 2008;48:763-775
  11. 11. Stein EM, Tallman MS. Emerging therapeutic drugs for AML. Blood. 2016;127:71-78
  12. 12. Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. The New England Journal of Medicine. 2017;377:454-464
  13. 13. Rydapt (midostaurin) [package insert]. (2017)
  14. 14. Strati H, Kantarjian F, Ravandi A, Nazha G, Borthakur N, Daver, et al. Phase I/II trial of the combination of midostaurin (PKC412) and 5-azacytidine for patients with acute myeloid leukemia and myelodysplastic syndrome. American Journal of Hematology. 2015;90:276-281
  15. 15. Pratz KW et al. A pharmacodynamic study of sorafenib in patients with relapsed and refractory acute leukemias. Leukemia. 2010;24:1437-1444
  16. 16. Borthakur G et al. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica. 2011;96:62-68
  17. 17. Ravandi F et al. Final report of phase II study of sorafenib, cytarabine and idarubicin for initial therapy in younger patients with acute myeloid leukemia. Leukemia. 2014;28:1543-1545
  18. 18. Ravandi F et al. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. Journal of Clinical Oncology. 2010;28:1856-1862
  19. 19. Widemann BC et al. A phase I trial and pharmacokinetic study of sorafenib in children with refractory solid tumors or leukemias: a Children’s Oncology Group Phase I Consortium report. Clinical Cancer Research. 2012;18:6011-6022
  20. 20. Man CH et al. Sorafenib treatment of FLT3-ITD(+) acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation. Blood. 2012;119:5133-5143
  21. 21. Röllig C, Serve H, Hüttmann A, Noppeney R, Müller-Tidow C, Krug U, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. The Lancet Oncology. 2015;16:1691-1699
  22. 22. Serve H, Krug U, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. Journal of Clinical Oncology. 2013;31:3110-3118
  23. 23. Brunner AM, Li S, Fathi AT, Wadleigh M, Ho VT, Collier K, et al. Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3-ITD acute myeloid leukaemia in first complete remission. British Journal of Haematology. 2016;175:496-504
  24. 24. Yee K, Schittenhelm M, O’Farrell A-M, Town A, McGreevey L, Bainbridge T, et al. Synergistic effect of SU11248 with cytarabine or daunorubicin on FLT3 ITD-positive leukemic cells. Blood. 2005;104:4202-4209. DOI: 10.1182/blood-2003-10-3381
  25. 25. Teng CLYC, Hwang WL, Tsai JR, Liu HC, Hwang GY, Hsu SL. Effector mechanisms of sunitinib-induced G1 cell cycle arrest, differentiation, and apoptosis in human acute myeloid leukaemia HL60 and KG-1 cells. Annals of Hematology. 2013;92(3):301-313
  26. 26. O’Farrell AM, Foran JM, Fiedler W, et al. An innovative phase I clinical study demonstrates inhibition of FLT3 phosphorylation by SU11248 in acute myeloid leukemia patients. Clinical Cancer Research. 2003;9:5465-5476
  27. 27. FIedler W, Serve H, Döhner H, et al. A phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease. Blood. 2005;105:986-993
  28. 28. Fiedler WKS, Kebenko M, Janning M, Krauter J, Schittenhelm M, Gotze K, et al. A phase I/II study of sunitinib and intensive chemotherapy in patients over 60 years of age with acute myeloid leukaemia and activating FLT3 mutations. British Journal of Haematology. 2015;169(5):694-700
  29. 29. Marshall JLKH, Deeken J, Bhargava P, Vogelzang NJ, Rizvi N, Luhtala T, et al. Phase I trial of orally administered CEP-701, a novel neurotrophin receptor-linked tyrosine kinase inhibitor. Investigational New Drugs. 2005;23(1):31-37
  30. 30. Levis MAJ, Tse KF, Zheng R, Baldwin BR, Smith BD, Jones-Bolin S, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood. 2002;99(11):3885-3891
  31. 31. Hexner EOMJ, Prchal J, Roboz GJ, Baer MR, Ritchie EK, Leibowitz D, et al. Phase I dose escalation study of lestaurtinib in patients with myelofibrosis. Leukemia & Lymphoma. 2015;56(9):2543-2551
  32. 32. Lee LY, Hernandez D, Rajkhowa T, Smith SC, Raman JR, Nguyen B, et al. Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood. 2017;129:257-260
  33. 33. Hexner EO, Serdikoff C, Jan M, Swider CR, Robinson C, Yang S, et al. Lestaurtinib (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the proliferation of primary erythroid cells from patients with myeloproliferative disorders. Blood. 2008;111:5663-5671
  34. 34. Knapper SBA, Littlewood T, Kell WJ, Agrawal S, Chopra R, Clark R, et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as firstline treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood. 2006;108(10):3262-3270
  35. 35. Smith BDLM, Beran M, Giles F, Kantarjian H, Berg K, Murphy KM, et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood. 2004;103(10):3669-3676
  36. 36. Kelly LMYJ, Boulton CL, Apatira M, Li J, Sullivan CM, Williams I, et al. CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML). Cancer Cell. 2002;1(5):421-432
  37. 37. DeAngelo DJSR, Heaney ML, Nimer SD, Paquette RL, Klisovic RB, Caligiuri MA, et al. Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in patients with acute myelogenous leukemia or high-risk myelodysplastic syndrome: safety, pharmacokinetics, and pharmacodynamics. Blood. 2006;108(12):3674-3681
  38. 38. Kampa-Schittenhelm KMHM, Akmut F, Dohner H, Dohner K, Schittenhelm MM. Quizartinib (AC220) is a potent second generation class III tyrosine kinase inhibitor that displays a distinct inhibition profile against mutant- FLT3, -PDGFRA and -KIT isoforms. Molecular Cancer. 2013;12:19
  39. 39. 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. Journal of Clinical Oncology. 2013;31(29):3681-3687
  40. 40. Altman JK, Foran JM, Pratz KW, Trone D, Cortes JE, Tallman MS. Phase 1 study of quizartinib in combination with induction and consolidation chemotherapy in patients with newly diagnosed acute myeloid leukemia. American Journal of Hematology. 2018;93(2):213-221. DOI: 10.1002/ajh.24974
  41. 41. Cortes J, Perl AE, Dohner H, Kantarjian H, Martinelli G, Kovacsovics T, et al. Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukemia: An open-label, multicentre, single-arm, phase 2 trial. The Lancet Oncology. 2018;19:889-903
  42. 42. Cortes JE, Khaled SK, Martinelli G, Perl AE, Ganguly S, Russell NH, et al. Efficacy and Safety of Single-Agent Quizartinib (Q), a Potent and Selective FLT3 Inhibitor (FLT3i), in Patients (pts) with FLT3-Internal Tandem Duplication (FLT3-ITD)-Mutated Relapsed/Refractory (R/R) Acute Myeloid Leukemia (AML) Enrolled in the Global, Phase 3, Randomized Controlled Quantum-R Trial. Blood. 2018;132:563
  43. 43. Kampa-Schittenhelm KMFJ, Haeusser LA, Illing B, Pavlovsky AA, Blumenstock G, Schittenhelm MM. Crenolanib is a type I tyrosine kinase inhibitor that inhibits mutant KIT D816 isoforms prevalent in systemic mastocytosis and core binding factor leukemia. Oncotarget. 2017;8(47):82897-82909
  44. 44. Levis M, Small D. FLT3: ITDoes matter in leukemia. Leukemia. 2003;17:1738-1752
  45. 45. Galanis A, Ma H, Rajkhowa T, Ramachandran A, Small D, Cortes J, et al. Crenolanib is a potent inhibitor of FLT3 with activity against resistanceconferring point mutants. Blood. 2014;123:94-100
  46. 46. Zimmerman EITD, Buaboonnam J, Hu S, Orwick S, Roberts MS, Janke LJ, et al. Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid leukemia. Blood. 2013;122(22):3607-3615
  47. 47. Cortes JE, Kantarjian HM, Kadia TM, Borthakur G, Konopleva M, Garcia-Manero G, et al. Crenolanib besylate, a type I pan-FLT3 inhibitor, to demonstrateclinical activity in multiply relapsed FLT3-ITD and D835 AML. Journal of Clinical Oncology. 2016;34:7008
  48. 48. Wang ES, MST RMS, Walter RB, Karanes C, Jain V, Collins RH. Low relapse rate in younger patients ≤ 60 years old with newly diagnosed FLT3- mutated acute myeloid leukemia (AML) treated with crenolanib and cytarabine/anthracycline chemotherapy. Blood. 2017;130:566
  49. 49. Wang ESRMS, Tallman MS, Walter RB, Eckardt JR, Collins R. Crenolanib, a type I FLT3 TKI, can be safely combined with cytarabine and anthracycline induction chemotherapy and results in high response rates in patients with newly diagnosed FLT3 mutant acute myeloid leukemia (AML). Blood. 2016;128(22):1071
  50. 50. Iyer SP, Jethava Y, Karanes C, Eckardt JR, Collins R. Safety study of salvage chemotherapy high-dose Ara-C/mitoxantrone (HAM) and type I FLT3-TKI crenolanib in first relapsed/primary refractory AML. Blood. 2016;128(22):3983
  51. 51. Maro Ohanian HMK, Borthakur G, Kadia TM, Konopleva M, Garcia-Manero G, Estrov Z, et al. Efficacy of a type I FLT3 inhibitor, crenolanib, with idarubicin and high-dose Ara-C in multiply relapsed/refractory FLT3+ AML. Blood. 2016;128:2744
  52. 52. Perl AE, Altman JK, Cortes J, Smith C, Litzow M, Baer MR, et al. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloidleukemia: A multicentre, first-in-human, open-label, phase 1-2 study. The Lancet Oncology. 2017;18:1061-1075
  53. 53. Usuki KST, Kobayashi Y, Miyamoto T, Iida H, Morita S, Bahceci E, et al. Clinical profile of gilteritinib in Japanese patients with relapsed/refractory AML an open-label phase 1 study. Cancer Science. 2018;109(10):3235-3244
  54. 54. Perl AE, Cortes JE, Strickland SA, Ritchie EK, Neubauer A, Martinelli G, et al. An open-label, randomized phase 3 study of gilteritinib versus salvage chemotherapy in relapsed or refractory FLT3 mutation-positive acute myeloid leukemia. Journal of Clinical Oncology. 2017;35:TPS7067
  55. 55. Tarabadkar ESTH, Blom A, Parvathaneni U, Olencki T, Nghiem P, Bhatia S. Clinical benefit from tyrosine kinase inhibitors in metastatic Merkel cell carcinoma: a case series of 5 patients. Am J Case Rep. 2018;19:505-511
  56. 56. Osanto S, van der Hulle T. Cabozantinib in the treatment of advanced renal cell carcinoma in adults following prior vascular endothelial growth factor targeted therapy: clinical trial evidence and experience. Therapeutic Advances in Urology. 2018;10(3):109-123
  57. 57. Fathi ATBT, Hernandez D, Amrein PC, Ballen KK, McMasters M, Avigan DE, et al. Cabozantinib is well tolerated in acute myeloid leukemia and effectively inhibits the resistance-conferring FLT3/tyrosine kinase domain/F691 mutation. Cancer. 2018;124(2):306-314
  58. 58. Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. The New England Journal of Medicine. 2009;361(11):1058-1066
  59. 59. Buege MJ, DiPippo AJ, DiNardo CD. Evolving treatment strategies for elderly leukemia patients with IDH mutations. Cancers (Basel). 2018;10(6):1-20
  60. 60. DiNardo C.D., F. Ravandi, S. Agresta, M. Konopleva, K. Takahashi, T. Kadia, et al., Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML, Am. J. Hematol. 90 (2015) 732-736
  61. 61. Paschka P, Schlenk RF, Gaidzik VI, Habdank M, Krönke J, Bullinger L, 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;28:3636-3643
  62. 62. Stein EM. IDH2 inhibition in AML: finally progress? Best Practice & Research Clinical Haematology. 2015;28:112-115
  63. 63. Marcucci G, Maharry K, Wu YZ, Radmacher MD, Mrozek K, Margeson D, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and leukemia group B study. Journal of Clinical Oncology. 2010;28:2348-2355
  64. 64. Green CL, Evans CM, Zhao L, Hills RK, Burnett AK, Linch DC, et al. The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood. 2011;118:409-412
  65. 65. Boissel N, Nibourel O, Renneville A, Gardin C, Reman O, Contentin N, et al. Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: a study by the acute leukemia French association group. Journal of Clinical Oncology. 2010;28:3717-3723
  66. 66. Amatangelo MD, Quek L, Shih A, Stein EM, Roshal M, David MD, et al. Enasidenib induces acute myeloid leukemia cell diferentiation to promote clinical response. Blood. 2017;130:732-741
  67. 67. Stein EM, DiNardo CD, Fathi AT, Pollyea DA, Stone RM, Altman JK, et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood. 2019;133:676-687
  68. 68. Stein EM, DiNardo CD, Fathi AT, Mims AS, Pratz KW, Savona MR, et al. Ivosidenib or Enasidenib Combined with Induction and Consolidation Chemotherapy in Patients with Newly Diagnosed AML with an IDH1 or IDH2 Mutation Is Safe, Effective and Leads to MRD-Negative Complete Remissions. Blood. 2018;132:560
  69. 69. DiNardo CD, Stein EM, de Botton S, Roboz GJ, Altman JK, Mims AS, et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. The New England Journal of Medicine. 2018;378:2386-2398
  70. 70. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM. Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science. 1984;226(4678):1097-1099
  71. 71. Monaghan P, Robertson D, Amos TAS, et al. Ultrastructural localization of Bcl-2 protein. The Journal of Histochemistry and Cytochemistry. 1992;40:1819-1825
  72. 72. Czabotar PE, Lessene G, Strasser A, Adams JM. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Boil. 2014;15:49-63
  73. 73. Testa U, Riccioni R. Deregulation of apoptosis in acute myeloid leukemia. Haematologica. 2007;92(1):81-94
  74. 74. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144:646-674
  75. 75. Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nature Medicine. 2013;19(2):202-208
  76. 76. Pan R, Ruvolo VR, Wei J, Konopleva M, Reed JC, Pellecchia M, et al. Inhibition of Mcl-1 with the pan-Bcl-2 family inhibitor (–)BI97D6 overcomes ABT-737 resistance in acute myeloid leukemia. Blood. 2015;126:363-372
  77. 77. Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological correlates of response in a phase ii study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discovery. 2016;6(10):1106-1117
  78. 78. Wei AH, Strickland SA Jr, Hou JZ, Fiedler W, Lin TL, Walter RB, et al. Venetoclax Combined with Low-Dose Cytarabine for Previously Untreated Patients with Acute Myeloid Leukemia: Results from a Phase 1b/2 Study. J. Clin. Oncol. 2019:Jco1801600
  79. 79. Pollyea DADC, Thirman MJ, Letai A, et al. Results of a phase 1b study of venetoclax plus decitabine or azacitidine in untreated acute myeloid leukemia patients $65 years ineligible for standard induction therapy. J Clin Oncol. 2016;34(suppl 15)
  80. 80. DiNardo CD, Albitar M, Kadia TM, Naqvi K, Vaughan K, Cavazos A, et al. Venetoclax in Combination with FLAG-IDA Chemotherapy (FLAG-V-I) for Fit, Relapsed/Refractory AML Patients: Interim Results of a Phase 1b/2 Dose Escalation and Expansion Study. Blood. 2018;132:4048
  81. 81. Armas-Lopez L, Zuniga J, Arrieta O, Avila-Moreno F. The Hedgehog-GLI pathway in embryonic development and cancer: Implications for pulmonary oncology therapy. Oncotarget. 2017;8:60684-60703
  82. 82. Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI, Eberhart CG, Watkins DN, et al. Medulloblastoma growth inhibition by hedgehog pathway blockade. Science. 2002;297:1559-1561
  83. 83. Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes & Development. 2008;22:2454-2472
  84. 84. Merchant AA, Matsui W. Targeting Hedgehog-a cancer stem cell pathway. Clinical Cancer Research. 2010;16:3130-3140
  85. 85. Queiroz KC, Ruela-de-Sousa RR, Fuhler GM, Aberson HL, Ferreira CV, Peppelenbosch MP, et al. Hedgehog signaling maintains chemoresistance in myeloid leukemic cells. Oncogene. 2010;29:6314-6322
  86. 86. Munchhof MJ, Li Q , Shavnya A, Borzillo GV, Boyden TL, Jones CS, et al. Discovery of PF-04449913, a potent and orally bioavailable inhibitor of smoothened. ACS Medicinal Chemistry Letters. 2012;3:106-111
  87. 87. Fukushima N, Minami Y, Kakiuchi S, et al. Small-molecule Hedgehog inhibitor attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Science. 2016;107(10):1422-1429. DOI: 10.1111/cas.13019
  88. 88. Martinelli G, Oehler VG, Papayannidis C, Courtney R, Shaik MN, Zhang X, et al. Treatment with PF-04449913, an oral smoothened antagonist, in patients with myeloid malignancies: A phase 1 safety and pharmacokinetics study. Lancet Haematol. 2015;2:e339-e346
  89. 89. Cortes JE, Heidel FH, Hellmann A, Fiedler W, Smith BD, Robak T, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33:379-389
  90. 90. Cortes JE, Douglas Smith B, Wang ES, Merchant A, Oehler VG, Arellano M, et al. Glasdegib in combination with cytarabine and daunorubicin in patients with AML or high-risk MDS: Phase 2 study results. American Journal of Hematology. 2018;93:1301-1310
  91. 91. Huang K, Ding B, Zhong Q , Jiang X, Li X, Wang Z, et al. Hh/IGF-1R/PI3K/Akt/MRP1 Pathway Induce Refractory Acute Myeloid Leukemia and Its Targeting Therpy. Blood. 2014;124:3612
  92. 92. Tibes R, Kosiorek HE, Dueck A, Palmer J, Slack JL, Knight EA, et al. Phase I/IB study of azacitidine and hedgehog pathway inhibition with Sonidegib (LDE225) in myeloid malignancies. Blood. 2017;130:2629
  93. 93. Bixby D, Noppeney R, Lin TL, Cortes J, Krauter J, Yee K, et al. Safety and efficacy of vismodegib in relapsed/refractory acute myeloid leukaemia: Results of a phase Ib trial. British Journal of Haematology. 2018
  94. 94. Schöffski P. Polo-like kinase (PLK) inhibitors in preclinical and early clinical development in oncology. The Oncologist. 2009;14(6):559-570
  95. 95. Lens SM, Voest EE, Medema RH. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nature Reviews. Cancer. 2010;10(12):825-841
  96. 96. Renner AG, Dos Santos C, Recher C, et al. Polo-like kinase 1 is overexpressed in acute myeloid leukemia and its inhibition preferentially targets the proliferation of leukemic cells. Blood. 2009;114(3):659-662
  97. 97. Rudolph D, Steegmaier M, Hoffmann M, et al. BI 6727, a Polo-like kinase inhibitor with improved pharmacokinetic profile and broad antitumor activity. Clinical Cancer Research. 2009;15(9):3094-3102
  98. 98. Döhner H, Lübbert M, Fiedler W, Fouillard L, Haaland A, Brandwein JM, et al. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood. 2014;124:1426-1433
  99. 99. Barbier V, Erbani J, Fiveash C, et al. Endothelial E-selectin inhibition improves acute myeloid leukaemia therapy by disrupting vascular niche-mediated chemoresistance. Nat Commun. 2020;11(1):2042. Published 2020 Apr 27
  100. 100. Geoffrey L. Uy, Jun Yin, Heidi D. Klepin, Shira Dinner, Anthony J. Jaslowski, Stephen A. Strickland, Jane L. Liesveld, John C. Byrd, Richard M. Stone; Alliance A041701 - a Randomized Phase 2/3 Study of Conventional Chemotherapy +/− Uproleselan (GMI-1271) in Older Adults with Acute Myeloid Leukemia (AML) Receiving Intensive Induction Chemotherapy. Blood 2019; 134 (Supplement_1): 1366
  101. 101. DeAngelo, D.J., Jonas, B.A., Liesveld, J., O’Dwyer, M., Bixby, D., Advani, A.S., Marlton, P., Magnani,J., Thackray, H.M. & Becker, P.S. (2017a) GMI-1271, a novel E-selectin antagonist, in combination with chemotherapy in relapsed/refractory AML. Journal of Clinical Oncology, 35, 2520
  102. 102. Daniel J. DeAngelo, Harry Paul Erba, Brian Andrew Jonas, Michael O’Dwyer, Paula Marlton, Gerwin A Huls, Jane Liesveld, Brenda W. Cooper, Bhavana Bhatnagar, Michael Armstrong, William Fogler, Mary Chen, John Magnani, Helen M. Thackray, Anjali S. Advani, and Pamela Sue Becker Journal of Clinical Oncology 2019 37:15_suppl, TPS7066-TPS7066
  103. 103. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56-61
  104. 104. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450-461
  105. 105. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews. Cancer. 2012;12:252-264
  106. 106. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nature Medicine. 2002;8(8):793-800. DOI: 10.1038/nm730
  107. 107. Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114:1545-1552
  108. 108. Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, et al. Overall Survival and Long-Term Safety of Nivolumab (Anti-Programmed Death 1 Antibody, BMS-936558, ONO-4538) in Patients With Previously Treated Advanced Non-Small-Cell Lung Cancer. Journal of Clinical Oncology. 2015;33(18):2004-2012
  109. 109. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. The New England Journal of Medicine. 2012;366(26):2443-2454
  110. 110. Daver N, Basu S, Garcia-Manero G, Cortes JE, Ravandi F, Jabbour EJ, et al. Phase IB/II study of nivolumab in combination with azacytidine (AZA) in Patients (pts) with relapsed acute myeloid leukemia (AML). Blood. 2016;128:763
  111. 111. Assi R, Kantarjian HM, Daver NG, Garcia-Manero G, Benton CB, Thompson PA. Results of a Phase 2, open-label study of idarubicin (I), cytarabine (A) and nivolumab (Nivo) in patients with newly diagnosed acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS). Blood. 2018;132(Suppl 1):905. DOI: 10.1002/cncr.31398
  112. 112. Albring J, Inselmann S, Sauer T, Schliemann C, Altvater B, Kailayangiri S, et al. PD-1 checkpoint blockade in patients with relapsed AML after allogeneic stem cell transplantation. Bone Marrow Transplantation. 2017;52:317-320
  113. 113. Zeidner JF, Vincent BG, Ivanova A, Foster M, Coombs CC, Jamieson K, et al. Genomics reveal potential biomarkers of response to pembrolizumab after high dose cytarabine in an ongoing Phase II Trial in relapsed/refractory AML. In: American Society of Hematology Annual Meeting; 2018 December 6-9 (San Diego: Oral and Poster Abstracts Session 616). 2018
  114. 114. Lindblad KE, Thompson J, Gui G, Valdez J, Worthy T, Tekleab H. Pembrolizumab and decitabine for refractory or relapsed acute myeloid leukemia. In: American Society of Hematology Annual Meeting; 2018 December 6-9 (San Diego: Oral and Poster Abstracts Session 616). 2018
  115. 115. Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, Savell A, et al. Leukemia and Lymphoma Society Blood Cancer Research Partnership. N Engl J Med. 2016;375(2):143-153
  116. 116. Griffin JD, Linch D, Sabbath K, Larcom P, Schlossman SF. A monoclonal antibody reactive with normal and leukemic human myeloid progenitor cells. Leukemia Research. 1984;8:521-534
  117. 117. Dinndorf PA, Andrews RG, Benjamin D, Ridgway D, Wolff L, Bernstein ID. Expression of normal myeloid-associated antigens by acute leukemia cells. Blood1986; 67: 1048-1053
  118. 118. Scheinberg DA, Tanimoto M, McKenzie S, Strife A, Old LJ, Clarkson BD. Monoclonal antibody M195: a diagnostic marker for acute myelogenous leukemia. Leukemia. 1989;3:440-445
  119. 119. Andrews RG, Torok-Storb B, Bernstein ID. Myeloid-associated differentiation antigens on stem cells and their progeny identified by monoclonal antibodies. Blood. 1983;62:124-132
  120. 120. Simmons D, Seed B. Isolation of a cDNA encoding CD33, a differentiation antigen of myeloid progenitor cells. The Journal of Immunology. 1988;141:2797-2800
  121. 121. Gao Z, McAlister VC, Williams GM. Repopulation of liver endothelium by bonemarrow- derived cells. The Lancet. 2001;357:932-933
  122. 122. Thol F, Schlenk RF. Gemtuzumab ozogamicin in acute myeloid leukemia revisited. Expert Opinion on Biological Therapy. 2014;14:1185-1195
  123. 123. Nejadmoghaddam MR, Minai-Tehrani A, Ghahremanzadeh R, Mahmoudi M, Dinarvand R, Zarnani AH. Antibody-drug conjugates: possibilities and challenges. Avicenna J Med Biotechnol. 2019;11(1):3-23
  124. 124. Sievers EL, Appelbaum FR, Spielberger RT, Forman SJ, Flowers D, Smith FO, et al. Selective ablation of acute myeloid leukemia using antibody-targeted chemotherapy: a phase I study of an anti-CD33 calicheamicin immunoconjugate. Blood. 1999;93(11):3678-3684
  125. 125. Sievers EL, Larson RA, Stadtmauer EA, Estey E, Lowenberg B, Dombret H, et al. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. Journal of Clinical Oncology. 2001;19(13):3244-3254
  126. 126. Petersdorf SH, Kopecky KJ, Slovak M, Willman C, Nevill T, Brandwein J, et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood. 2013;121(24):4854-4860
  127. 127. Castaigne S, Pautas C, Terre C, Raoux E, Bordessoule D, Bastie JN, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novoacute myeloid leukemia (ALFA-0701): A randomised, open-label, phase 3 study. Lancet. 2012;379:1508-1516
  128. 128. Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M, et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukemia: A meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15:986-996
  129. 129. Kung Sutherland MS, Walter RB, Jeffrey SC, Burke PJ, Yu C, Kostner H, et al. SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drugresistant AML. Blood. 2013;122:1455-1463
  130. 130. Bixby DL, Stein AS, Fathi AT, Kovacsovics TJ, Levy MY, Erba HP, et al. Vadastuximab Talirine monotherapy in older patients with treatment naive CD33-positive acute myeloid leukemia (AML). Blood. 2016;128(22)
  131. 131. Stein EM, Walter RB, Erba HP, Fathi AT, Advani AS, Lancet JE, et al. A phase 1 trial of vadastuximab talirine as monotherapy in patients with CD33-positive acute myeloid leukemia. Blood. 2018;131(4):387-396
  132. 132. Erba HP, Levy MY, Vasu S, Stein AS, Fathi AT, Maris MB, et al. A phase 1b Study of Vadastuximab Talirine in combination with 7+3 induction therapy for patients with newly diagnosed acute myeloid leukemia (AML). Blood. 2016;128(22)
  133. 133. Fathi AT, Erba HP, Lancet JE, Stein EM, Ravandi F, Faderl S, et al. A phase 1 trial of vadastuximab talirine combined with hypomethylating agents in patients with CD33-positive AML. Blood. 2018;132(11):1125-1133

Written By

Vasko Graklanov

Submitted: 29 May 2020 Reviewed: 11 October 2020 Published: 28 October 2020