Abstract
Together, acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) make up approximately one-third of all pediatric cancer diagnoses. Despite remarkable improvement in the treatment outcomes of these diseases over the past several decades, the prognosis for certain high-risk groups of leukemia and for relapsed disease remains poor. However, recent insights into different types of ‘driver’ lesions of leukemogenesis, such as the aberrant activation of signaling pathways and various epigenetic modifications, have led to the discovery of novel agents that specifically target the mechanism of transformation. In parallel, emerging approaches in cancer immunotherapy have led to newer therapies that can exploit and harness cytotoxic immunity directed against malignant cells. This review details the rationale and implementation of recent and specifically targeted therapies in acute pediatric leukemia. Topics covered include the inhibition of critical cell signaling pathways [BCR-ABL, FMS-like tyrosine kinase 3 (FLT3), mammalian target of rapamycin (mTOR), and Janus-associated kinase (JAK)], proteasome inhibition, inhibition of epigenetic regulators of gene expression [DNA methyltransferase (DNMT) inhibitors, histone deacetylase (HDAC) inhibitors, and disruptor of telomeric signaling-1 (DOT1L) inhibitors], monoclonal antibodies and immunoconjugated toxins, bispecific T-cell engaging (BiTE) antibodies, and chimeric antigen receptor-modified (CAR) T cells.
Keywords: ALL, AML, blinatumomab, CAR T-cells, carfilzomib, EPZ-5676, FLT3, moxetumomab
Introduction
Remarkable progress has been made in the past several decades in the treatment of childhood acute lymphoblastic leukemia (ALL), with 5-year survival rates now approaching 90% [Hunger et al. 2012]. This success has been achieved with a risk stratification design incorporating minimal residual disease (MRD) status, and the intensification of cytotoxic chemotherapy for those at highest risk [Pui et al. 2011a]. However, up to 20% of children with ALL will be refractory to treatment or relapse following treatment, and the survival rate for relapsed ALL remains poor. In addition, although the majority of children with acute myeloid leukemia (AML) will achieve remission with conventional chemotherapy, less than 60% will be long-term survivors. Cytotoxic therapy intensification has been maximized in the treatment of AML, highlighting the need for additional, targeted novel therapies in this disease [Pui et al. 2011b]. Targeted therapies in pediatric leukemia are largely unproven to date, with the clear exceptions of tyrosine kinase inhibitors (TKIs) in BCR-ABL (Philadelphia chromosome) positive leukemia [Schultz et al. 2009], and all-trans retinoic acid (ATRA) in acute promyelocytic leukemia (APML) with PML-RARa fusions [Tallman, 2004]. Challenges have included the effective incorporation of novel agents into current chemotherapy regimens, the development of resistance to targeted therapies, and failure to eradicate the leukemia stem cell (LSC) population in an individual disease. In this article, we summarize recent and developing novel drugs for pediatric acute leukemia, review open and recently completed clinical trials using these agents (listed in Table 1), and discuss plans to implement these therapies into pediatric ALL or AML treatment regimens.
Table 1.
Drug | Target | Disease | Phase | Clinical trial identifier(s) | |
---|---|---|---|---|---|
Dasatinib | BCR-ABL | ALL (Ph+) | II | NCT01460160 | *completed |
COG AALL1122 | |||||
Nilotonib | BCR-ABL | ALL, CML (Ph+) | I | NCT01077544 | |
CAMN107A2120 | |||||
Lestaurtinib | FLT3 | Infant ALL | III | NCT00557193 | *completed |
AML | I | COG AALL0631 | |||
NCT00469859 | *completed | ||||
COG AAML06P1 | |||||
Midostaurin | FLT3 | Infant ALL, AML | I/II | NCT00866281 | |
Quizartinib | FLT3 | ALL, AML | I | NCT01411267 | *completed |
TACL 2009-004 | |||||
Sorafenib | FLT3, other RTKs | AML | III | NCT01371981 | |
Infant ALL, AML, MDS | I | COG AAML1031 | |||
ALL, AML | I | NCT00908167 | |||
RELHEM | |||||
NCT00665990 | |||||
ANGIO1 | |||||
Rapamycin | mTOR | ALL, NHL | I | NCT01658007 | |
ALL | III | 2012-0361 | |||
NCT00382109 | *completed | ||||
COG ASCT0431 | |||||
Temsirolimus | mTOR | ALL, NHL | I | NCT01403415 | |
COG ADVL1114 | |||||
Everolimus | mTOR | ALL | I | NCT01523977 | |
ALL | I/II | 11-237 | |||
NCT00968253 | |||||
2009-0100 | |||||
Ruxolitinib | JAK 1/2 | ALL, AML | I | NCT01164163 | *completed |
COG ADVL1011 | |||||
Bortezomib | Proteasome | ALL, AML | I | NCT00077467 | *completed |
AML | II | COG ADVL0317 | |||
ALL | II | NCT00666588 | *completed | ||
AML | III | COG AAML07P1 | |||
NCT00873093 | *completed | ||||
COG AALL07P1 | |||||
NCT01371981 | |||||
COG AAML1031 | |||||
Azacitidine | DNMT | ALL, AML | I | NCT01861002 | *completed |
TACL 2011-002 | |||||
Vorinostat | HDAC | ALL | I/II | NCT01483690 | |
TACL 2009-003 | |||||
Panobinostat | HDAC | ALL, AML, HL, NHL | I | NCT01321346 | |
TACL 2009-012 | |||||
EPZ-5676 | DOT1L | ALL, AML (MLL-r) | IIb | NCT02141828 | |
Gemtuzomab | CD33 | AML | III | NCT00372593 | *completed |
COG AAML0531 | |||||
Epratuzumab | CD22 | CD22+ ALL | I/II | NCT00098839 | *completed |
ALL | II | ADVL04P2 | |||
NCT01802814 | |||||
IntReALL SR 2010 | |||||
Inotuzumab | CD22 | ALL | I/II | NCT01134575 | *completed |
2009-0872 | |||||
Moxetumomab | CD22 | ALL or NHL | I | NCT00659425 | |
II | CAT-8015-1004 | ||||
ALL | NCT02227108 | ||||
CAT-8015-1036 | |||||
Blinatumomab | CD3, CD19 | ALL | I/II | NCT01471782 | |
ALL | III | MT103-205 | |||
NCT02101853 | |||||
COG AALL1331 | |||||
CAR T-cells | CD19 | CD19+ ALL /lymphoma | I/IIa | NCT01626495 | |
CD19+ ALL /lymphoma | II | 10-007706 | |||
CD19+ ALL | I | NCT01593696 | |||
CD19+ ALL | I | 120112, 12-C-0112 | |||
CD19+ ALL | I/II | NCT01860937 | |||
13-052 | |||||
NCT01683279 | |||||
PLAT-01 | |||||
NCT02028455 | |||||
PLAT-02 |
ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; Ph+, Philadelphia chromosome positive; CML, chronic myeloid leukemia; MDS, myelodysplastic syndrome; NHL, non-Hodgkin lymphoma; HL, Hodgkin lymphoma; MLL-r, mixed lineage leukemia gene rearranged; DNMT, DNA methyltransferase; HDAC, histone deacetylase; DOT1L, Disruptor of telomerase-1; CAR T-cells, chimeric antigen receptor.
Tyrosine kinases (TKIs)
BCR-ABL
Among the successful stories of targeted therapies in pediatric acute leukemia is the introduction of the TKI imatinib into upfront therapy for Philadelphia chromosome positive (Ph+) ALL patients. The Children’s Oncology Group (COG) trial AALL0031 (2002–2006) incorporated imatinib into an upfront, intensive chemotherapy backbone for Ph+ ALL pediatric patients. Initial results from this trial demonstrated a 3-year event-free survival (EFS) of 88%, doubling that of historical controls [Schultz et al. 2009]. In addition, longer follow-up revealed that Ph+ ALL patients receiving chemotherapy with continuous imatinib had similar 5-year EFS rates to patients undergoing either related or unrelated donor hematopoietic stem-cell transplant (HSCT; 71%, 64% and 63%, respectively; p = 0.77) [Schultz et al. 2014]. Retrospective analysis of patients who relapsed after treatment demonstrated remission reinduction rates similar to other high-risk non-Ph+ ALL patients treated on contemporaneous trials, allowing these patients to proceed to HSCT as salvage therapy [Schultz et al. 2014]. Imatinib was FDA-approved for the treatment of Ph+ ALL in children in 2013. However, a well-known mechanism of resistance to TKI therapy is the outgrowth of resistant clones, often mediated through the development of point mutations in the kinase domain of ABL. In a recent review of 272 adult patients with relapsed Ph+ ALL, 70% harbored a kinase domain point mutation, including T315I, E255K and Y253H [Soverini et al. 2014]. Interestingly, kinase point mutations seem to be less common in pediatric Ph+ ALL at relapse, although they have been demonstrated to occur [Chang et al. 2012].
Dasatinib, a second-generation TKI, replaced imatinib in the most recent COG trial AALL1122 for Ph+ ALL patients, after COG phase I/II trial AALL0622 demonstrated good tolerability and rapid efficacy of dasatinib in combination with chemotherapy. There is in vitro evidence that dasatinib has superior central nervous system (CNS) penetration compared with imatinib [Porkka et al. 2008]. Dasatinib is effective against many resistant mutations, with the exception of point mutation T315I [Talpaz et al. 2006]. Nilotinib is another second-generation TKI that has been less studied in both adults and pediatrics, but some reports show efficacy against certain dasatinib-resistant mutations, although not T315I [Jabbour et al. 2008; Sekimizu et al. 2013]. A phase II COG study is investigating the efficacy of nilotinib in pediatric chronic myeloid leukemia (CML), and a multi-institutional phase I study of nilotinib is open for pediatric patients with CML or relapsed/refractory Ph+ ALL (Table 1). The third-generation TKI, ponatinib, is active against the increasingly clinically significant mutation T315I, but toxicities of arterial thrombosis risk were documented [Cortes et al. 2013], temporarily halting its development in clinical trials. As of January 2014, ponatinib is FDA-approved for adults with Ph+ leukemia resistant to other TKIs, now carrying the additional warning of thrombosis. Other classes of kinase inhibitors are being explored in adult Ph+ leukemia in an attempt to prevent the development of resistance, such as the Janus-associated kinase (JAK) inhibitor, ruxolitinib, in combination with nilotinib [ClinicalTrials.gov identifiers: NCT01702064 and NCT01914484].
FMS-like tyrosine kinase 3 (FLT3)
FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase expressed on human CD34+ hematopoietic stem and early progenitor cells, and FLT3 signaling is central to cell proliferation and differentiation [Small et al. 1994]. FLT3 is aberrantly expressed on the majority of leukemic blasts regardless of CD34 expression [Carow et al. 1996]. Of note, the most consistently overexpressed gene in mixed lineage leukemia-rearranged (MLL-r) infant ALL is wild-type FLT3 [Armstrong et al. 2002]. Also, mutations of FLT3 occur in 20–25% of pediatric AML patients, and result in ligand-independent constitutive activation of the receptor [Kondo et al. 1999; Meshinchi et al. 2001]. Roughly two-thirds of these mutations are internal tandem duplications (ITD) of the juxtamembrane domain of the gene, and the remaining one-third are point mutations of the tyrosine kinase domain (TKD) [Meshinchi et al. 2001; Yamamoto et al. 2001]. Multiple studies have documented decreased overall survival and increased rate of relapse in FLT3-ITD mutant AML [Iwai et al. 1999; Kondo et al. 1999; Meshinchi et al. 2001]. In one study, children with ITD mutations had 8-year overall survival (OS) and EFS rates of 13% and 7%, respectively, compared with an OS of 50% and EFS of 44% for children without ITD mutations [Meshinchi et al. 2001]. A large retrospective review determined that an ITD allelic ratio of 0.4 or higher identified the highest risk group with the worst prognosis, whereas children with allelic ratios <0.4 had similar outcomes to those with wild type FLT3 [Meshinchi et al. 2006]. These data provide strong rationale for the use of FLT3 inhibitors in pediatric acute leukemia.
The FLT3 inhibitor lestaurtinib (CEP-701) has shown modest efficacy in adult trials as monotherapy [Smith et al. 2004; Knapper et al. 2006] or in combination with chemotherapy [Levis et al. 2011]. In pediatrics, a phase I trial of lestaurtinib with chemotherapy for relapsed/refractory AML has been completed, though clinical data are not yet published. However, at each dose level, five of six patients tested had >80% inhibition of FLT3 phosphorylation at the majority of lestaurtinib trough time points. Recently completed COG phase III trial AALL0631 investigated lestaurtinib in combination with chemotherapy for newly diagnosed infant ALL. Intermediate-risk (MLL-r and >90 days old) and high-risk (MLL-r and <90 days old) infants were randomized to receive lestaurtinib after induction chemotherapy. Trial efficacy results are pending.
Midostaurin (PKC412) is a multi-TKI that has activity against FLT3, and early phase adult clinical trials showed hematological responses in patients with mutant FLT3 [Stone et al. 2005; Fischer et al. 2010]. In pediatrics, a phase I/II clinical trial open in Europe and some US centers is currently recruiting MLL-r infant ALL and FLT3-mutant AML patients to receive midostaurin as a single agent. Laboratory correlatives include evaluation of FLT3 phosphorylation before and after receiving drug.
Quizartinib, or AC220, is a far more potent and selective FLT3 inhibitor than lestaurtinib and midostaurin. In a phase II study of quizartinib as monotherapy in relapsed/refractory adults with FLT3-ITD mutant AML, a complete response (CR) or a complete response with incomplete blood count recovery (CRi) was induced in 9 of 17 patients (53%) [Levis et al. 2012]. Current adult studies are investigating quizartinib in combination with chemotherapy. In pediatrics, a recently completed Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) pilot study investigated quizartinib in combination with cytarabine and etoposide in patients with MLL-r ALL or relapsed/refractory AML. Laboratory correlatives showed near-complete inhibition of FLT3 phosphorylation; importantly, four of six patients with FLT3-ITD mutant AML achieved a CR or CRi, and the other two ITD patients had stable disease [Cooper et al. 2013].
Sorafenib, a multi-TKI with activity against FLT3, induced clinical responses specifically in ITD-mutant cases of relapsed/refractory AML in three independent phase I adult trials [Zhang et al. 2008; Crump et al. 2010; Pratz et al. 2010]. A phase I/II trial in newly diagnosed adult AML combined sorafenib with cytarabine and idarubicin and impressively, 38 of 51 (75%) patients achieved a CR, including 14 of 15 FLT3-ITD patients [Ravandi et al. 2010]. In 12 pediatric patients with relapsed/refractory AML, 5 of 5 FLT3-ITD and 3 of 7 wild type FLT3 patients achieved a CR or CRi with sorafenib in combination with clofarabine and cytarabine [Inaba et al. 2011]. Given these encouraging reports, the current COG phase III trial for newly diagnosed pediatric AML nonrandomly assigns ITD-mutant patients with high allelic ratios (>0.4) to receive sorafenib in combination with chemotherapy, and includes a maintenance phase with single agent sorafenib for 1 year. There are also two phase I studies open at St. Jude Children’s Research Hospital, combining sorafenib with chemotherapy in patients with refractory hematological malignancies.
Serine/threonine kinases
mTOR
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase, and is centrally integrated in several key signal transduction pathways critical to cell growth and proliferation. Aberrant activation of the mTOR pathway has been demonstrated in multiple tumor types, and inhibition of mTOR by the macrolide rapamycin (sirolimus) or one of its analogs (temsirolimus, everolimus) has shown antitumor activity in preclinical models and in early phase clinical trials.
Constitutive activation of the mTOR pathway has been demonstrated in the majority of cases of AML [Min et al. 2003; Xu et al. 2003; Recher et al. 2005b]. Rapamycin treatment causes G0/G1 cell cycle arrest in AML cell lines, and inhibits the clonogenic properties of AML patient samples without significantly affecting healthy donor CD34+ bone marrow (BM) cells [Recher et al. 2005a]. Rapamycin induced clinical responses in 4 of 9 relapsed/refractory adult AML patients when given as a single agent for a 28-day course [Recher et al. 2005b]. A phase I/II study of single-agent everolimus in adults with relapsed/refractory leukemia demonstrated good tolerability and a modest clinical response [Yee et al. 2006]. Currently, there are no ongoing trials investigating mTOR inhibitors in childhood AML.
Rapamycin has also been shown to have activity in pre-B cell ALL cell lines, primary patient samples, and a xenograft model [Brown et al. 2003; Maude et al. 2012]. mTOR inhibitors also synergize with methotrexate in pre-B ALL cells [Teachey et al. 2008]. In addition, rapamycin was shown to sensitize steroid-resistant malignant lymphoid cells to glucocorticoid-induced apoptosis via modulation of anti-apoptotic MCL1 [Wei et al. 2006], and could therefore have a role in overcoming glucocorticoid resistance, a known poor prognostic factor in ALL. These studies provide strong rationale to pursue mTOR inhibition in combination with chemotherapy in pediatric ALL, and there are multiple ongoing early phase clinical trials investigating these agents. A single-institution pilot trial at Cincinnati Children’s hospital combines rapamycin with the mitoxantrone arm of UKALLR3 reinduction chemotherapy [Parker et al. 2010] in patients with relapsed ALL up to 30 years of age. The phase I COG study ADVL1114 is investigating temsirolimus in combination with a similar reinduction chemotherapy backbone in relapsed pediatric ALL patients. A multi-institutional phase I trial combines everolimus with four-drug reinduction for pediatric ALL patients in first bone marrow relapse. Finally, a single-institution phase I/II trial investigating everolimus in combination with hyper-CVAD chemotherapy is underway for patients aged 10 years and older with relapsed/refractory ALL.
mTOR inhibitors such as rapamycin (sirolimus) are also effective in graft versus host disease (GVHD). The recently completed phase III COG trial, ASCT0431, investigated whether the addition of sirolimus to GVHD prophylaxis in children with ALL would decrease relapse rates as well as acute GVHD (aGVHD). Results showed that although sirolimus decreased aGVHD, survival was not improved [Pulsipher et al. 2014].
JAK/STAT
Hyperactive signaling of the Janus-associated kinase (JAK)/signal transducer and activator of transcription (STAT) pathway has been documented in several types of high-risk leukemia. Activating mutations of JAK2 are well documented in AML [Daver and Cortes, 2012]. Aberrant activation of JAK signaling has also been described in Philadelphia chromosome-like (Ph-like) ALL, a recently defined subtype of ALL with gene expression patterns similar to Ph+ cases but lacking the BCR-ABL fusion [Mullighan et al. 2009]. Xenograft models of eight cases of Ph-like ALL demonstrated decreased leukemic burden when treated with a selective JAK1/2 inhibitor, ruxolitinib, and six of these xenograft models harbored either a JAK2 mutation or CRLF2 rearrangement [Maude et al. 2012]. The remaining two Ph-like ALL patient samples contained some other activating signature of hyperactive JAK/STAT signaling, despite lacking a point mutation. This suggests that a JAK2 activation footprint may be more significant than the presence of a mutation, in terms of predicting response to JAK2 inhibition. Interestingly, the mTOR pathway is also often aberrantly activated in Ph-like pre-B ALL patients, and single agent rapamycin demonstrated activity in all eight of these xenograft models [Maude et al. 2012]. Combining mTOR inhibitors with JAK2 inhibition, or combining JAK2 inhibition with cytotoxic chemotherapy, has not yet been studied in pediatric leukemia. The phase I COG study ADVL1011 recently investigated the safety and dosing of single-agent ruxolitinib in children with relapsed/refractory hematologic malignancies and solid tumors, and results are pending.
Furthermore, a detailed genomic analysis of 154 patients identified as Ph-like ALL by gene expression profiling was recently reported [Roberts et al. 2014]. Greater than 90% of patients in this cohort were found to have a kinase-activating lesion. Rearrangements involving ABL1, ABL2, and JAK2 were among the most common alterations, and fusion protein expression caused cell proliferation and activated STAT5 signaling. Importantly, Ph-like leukemia cells with fusions involving ABL 1/2 were sensitive to dasatinib in vitro, and cells with JAK2 rearrangements were sensitive to ruxolitinib. Trials are needed to determine whether identifying Ph-like patients and incorporating targeted TKIs into therapy will improve outcomes in this patient population.
JAK2 inhibitors were also identified in a high-throughput screen of kinase inhibitors as potential therapeutic agents in resistant FLT3-mutant AML [Weisberg et al. 2012]. The combination of JAK inhibitors with the FLT3 inhibitor PKC412 in AML cell lines demonstrated synergistic cytotoxicity and inhibition of downstream pathway signaling, specifically overcoming resistance to PKC412 in the presence of stroma [Weisberg et al. 2012]. An adult phase III study is investigating pacritinib, a dual JAK2/FLT3 inhibitor, in myelofibrosis [ClinicalTrials.gov identifier: NCT02055781].
Proteasome inhibitors
Proteasome inhibitors impair tumor growth through a variety of mechanisms [Rajkumar et al. 2005] and inactivate nuclear factor kB (NF-kB) by blocking the degradation of IkBa, a negative regulator of NF-kB. NF-kB is a transcriptional activator with anti-apoptotic properties, and is thought to be a key survival factor in various malignancies [Wang et al. 1996]. NF-kB is aberrantly constitutively activated in primary AML samples, specifically in the quiescent CD34+/CD38− LSC compartment [Guzman et al. 2001]. NF-kB activity is also increased in leukemia cells after treatment with chemotherapy [Maestre et al. 2001]. Treatment of AML samples with a proteasome inhibitor led to down-regulation of NF-kB and its downstream targets, and induced apoptosis in CD34+ leukemia cells, but not in normal CD34+ bone marrow [Guzman et al. 2001]. Combining a proteasome inhibitor with idarubicin induced a rapid apoptotic response in primary AML samples in vitro, and effectively ablated the ability of the LSC population to engraft in a xenograft model, though normal hematopoietic cells were viable and able to engraft [Guzman et al. 2002]. This data led to the clinical investigation of a proteasome inhibitor, bortezomib, hypothesizing that selective targeting of the LSC population would result in more effective and durable responses in combination with standard treatment.
Bortezomib
Although bortezomib treatment inhibits NF-kB activity, it has little antileukemia activity as a single agent [Horton et al. 2007]. Adult trials have demonstrated that bortezomib may be safely combined with AML chemotherapy, although there have been rare cases of acute respiratory distress syndrome (ARDS) in combination with high-dose cytarabine [Attar et al. 2008]. The phase II COG protocol AAML07P1 combined bortezomib with standard AML chemotherapy and though results are not yet published, no children developed ARDS. Thus, while the available data on bortezomib in pediatric AML are limited, in vitro and adult data demonstrate that bortezomib may be combined safely with AML chemotherapy and may augment the efficacy of standard AML therapy. Phase III COG trial AAML1031 is evaluating bortezomib in a randomized fashion in newly diagnosed pediatric AML, in combination with standard chemotherapy.
A phase I TACL trial determined that bortezomib is well tolerated in combination with four-drug reinduction chemotherapy in relapsed/refractory pediatric ALL [Messinger et al. 2010]. The expanded phase II portion of this trial evaluated 22 relapsed ALL pediatric patients and demonstrated a 73% overall response rate, including 14 CRs and 2 CRis [Messinger et al. 2012]. The recently completed phase II COG study AALL07P1 combined bortezomib with reinduction chemotherapy for relapsed ALL. End of induction CR was achieved in 42 of 61 (68%) of pre-B ALL patients, as well as 11 of 17 (65%) T-cell ALL patients (unpublished data, Horton, COG open meeting, 2013), prompting the planned incorporation of bortezomib in a randomized fashion in the next phase III COG study for newly diagnosed T-cell ALL, AALL1231.
Carfilzomib
Carfilzomib is another proteasome inhibitor that is structurally and mechanistically distinct from bortezomib, demonstrating less reactivity against non-proteosomal proteases [Arastu-Kapur et al. 2011] and achieving higher levels of proteasome inhibition than bortezomib in preclinical models [Yang et al. 2011]. In early phase trials of adults with multiple myeloma, carfilzomib showed good tolerability both alone and in combination with chemotherapy, and clinical responses were achieved in both bortezomib-naïve and bortezomib pretreated patients [Siegel et al. 2012; Wang et al. 2013]. Carfilzomib induced apoptosis in a variety of pediatric tumor cell lines, and synergized with etoposide and cyclophosphamide [Jayanthan et al. 2013; Ruan et al. 2013]. Based on this encouraging preclinical data, a phase I Pediatric Oncology Experimental Therapeutics Investigator’s Consortium (POETIC) study is planned, evaluating the tolerability of carfilzomib with cyclophosphamide and etoposide in relapsed/refractory pediatric leukemia and solid tumors. A TACL phase I/II study is also planned for children with relapsed/refractory leukemia, investigating carfilzomib in combination with UKALLR3 reinduction chemotherapy.
Epigenetic targeting
Epigenetic modifications, including methylation of CpG islands in gene promoter regions and chromatin modifications by histone acetylation, play a critical role in gene expression. Transcriptional silencing of tumor suppressor genes can lead to malignant transformation in many cancers, including leukemia [Herman et al. 1997; Seedhouse et al. 2003]. These modifications can be targeted by DNA methyltransferase (DNMT) inhibition or histone deacetylase (HDAC) inhibition in an attempt to reverse the epigenetic silencing of crucial regulatory genes and modify the malignant phenotype [Gore, 2005].
DNMT
The two azanucleosides, 5-azacytidine (azacitidine) and 5-aza-2′-deoxycytidine (decitabine), are cytosine analogs that incorporate into nucleic acids, thereby affecting multiple molecular pathways. After incorporation into DNA, both drugs covalently ‘trap’ DNMTs, which are then degraded [Santi et al. 1984]. There is in vitro evidence that these drugs are effective in inducing DNA demethylation and epigenetic gene reactivation [Stresemann et al. 2006], although differential demethylating effects on various human AML cell lines is seen [Stresemann et al. 2008].
Both azacitidine and decitabine are effective in the treatment of adult patients with MDS [Silverman et al. 2002; Kantarjian et al. 2006], and have been FDA-approved for this disease. Decitabine has also been approved in Europe for the treatment of adult AML. Combining DNMT inhibitors with HDAC inhibitors is being studied extensively in adult MDS and AML, with promising initial results [Garcia-Manero et al. 2006; Gore et al. 2006; Soriano et al. 2007]. Some studies suggest that despite good clinical responses, no correlation is seen between response and methylation or gene expression patterns [Fandy et al. 2009]. However, another study compared gene expression and methylation patterns of paired diagnostic-relapsed ALL samples, and demonstrated a relapse-specific signature associated with chemoresistance [Bhatla et al. 2012]. Treating primary relapsed samples or leukemia cell lines in vitro with the HDAC inhibitor vorinostat alone or in combination with decitabine restored expression of genes that were preferentially silenced at relapse, and seemed to ‘reprogram’ the abnormal relapsed gene expression profile. Furthermore, these drugs demonstrate synergistic cytotoxicity when given prior to chemotherapy [Bhatla et al. 2012]. A recently completed phase I TACL trial combined azacitidine with chemotherapy for relapsed/refractory pediatric ALL or AML, and an ongoing phase I/II TACL trial is evaluating the efficacy of epigenetic therapy with decitabine and vorinostat followed by reinduction chemotherapy in relapsed/refractory pediatric ALL.
HDAC
HDACs remove acetyl groups from lysine residues on histones, causing decreased accessibility of chromatin and thus, transcriptional repression and epigenetic silencing [Lucas et al. 2010]. In vitro experiments demonstrate efficacy of HDAC inhibitors in both AML and ALL. One investigation demonstrated synergy of vorinostat and methotrexate in ALL cell lines by increasing the expression of the synthetase that metabolizes methotrexate to its active form, leading to increased cytotoxicity [Leclerc et al. 2010]. Another study demonstrated restoration of BH3-only BCL2 family member (BIM) expression after vorinostat treatment in glucocorticoid-resistant ALL xenografts, resensitizing this disease to glucocorticoids [Bachmann et al. 2010]. In an AML cell line, treatment with vorinostat resulted in increased expression of p21, leading to p53-independent cell cycle arrest [Vrana et al. 1999]. HDAC inhibitors have also been shown to abolish the fusion proteins AML1-ETO and PML-RARa in AML [Kramer et al. 2008]. Stumpel and colleagues demonstrated that treatment with HDAC inhibitors induced cytotoxicity in t(4;11) MLL-r primary infant ALL cells, and HDAC inhibitor treatment was shown to neutralize the MLL-AF4 fusion protein [Stumpel et al. 2012]. These data and others led to the investigation of HDAC inhibitors in acute leukemia.
A phase I adult trial with vorinostat monotherapy demonstrated some antileukemia effect and good tolerability [Garcia-Manero et al. 2008]. Subsequently, a phase I pediatric trial with vorinostat in solid tumors and leukemia also demonstrated tolerability, although patients with refractory leukemia required a lower dose [Fouladi et al. 2010]. As stated above, vorinostat is currently being investigated in a phase II pediatric clinical trial in combination with decitabine in relapsed ALL, in an attempt to ‘reprogram’ the gene expression signature and restore sensitivity to chemotherapy. In addition, a phase I TACL trial recently opened and is investigating the tolerability of the newer HDAC inhibitor, panobinostat, in refractory pediatric hematological malignancies.
DOT1L
Approximately 60–80% of infant leukemia harbor rearrangements of the MLL gene (MLL-r) [Ayton and Cleary, 2001] and have a poor overall prognosis with standard chemotherapy approaches. Similar gene expression patterns have been described in MLL-r leukemia despite the variety of possible fusion proteins [Tsutsumi et al. 2003], including upregulation of homeobox (Hox) gene expression [Krivtsov et al. 2006]. It has been demonstrated that various MLL-fusion proteins form a complex with Disruptor of telomeric signaling-1 (DOT1L) [Zhang et al. 2006; Buttner et al. 2010]. DOT1L is the sole methyltransferase that methylates lysine 79 of histone 3 (H3K79), causing transcriptional activation [Lacoste et al. 2002]. The interaction of MLL-fusion proteins with DOT1L can lead to the mistargeting of DOT1L and subsequent methylation of H3K79 at inappropriate gene promoter sites, likely playing an important role in leukemogenic transformation [Okada et al. 2005; Mueller et al. 2007; Krivtsov et al. 2008]. For example, hypermethylation of H3K79 is seen at the promoter of HoxA9 in MLL-r leukemia [Krivtsov et al. 2008; Yokoyama et al. 2010].
In vivo and in vitro experiments have demonstrated that H3K79 methylation by DOT1L is in fact required for both initiation and maintenance of MLL-AF9 induced leukemogenesis [Chang et al. 2010; Bernt et al. 2011; Nguyen et al. 2011]. A potent small molecule inhibitor of DOT1L, EPZ004777, selectively killed MLL-r cells while having little effect on non-MLL translocated cells, and led to increased survival in an MLL-AF9 xenograft model [Daigle et al. 2011]. Similar results were then demonstrated in MLL-AF10 and MLL-AF6 cells [Chen et al. 2013; Deshpande et al. 2013]. Furthermore, treatment with EPZ004777 led to decreased expression of HoxA9 and Meis1 in all MLL-rearranged leukemia cells. These data provide strong rationale for targeting DOT1L in MLL-r leukemia. A phase I/II trial of DOT1L inhibitor EPZ-5676 is currently underway for adults with advanced MLL-r hematological malignancies [ClinicalTrials.gov identifier: NCT01684150], and a phase I multi-institutional trial of EPZ-5676 recently opened for pediatric patients with relapsed/refractory MLL-r leukemia.
Antibody-based immunotherapy
CD33
CD33 cell surface antigen is expressed in 88% of childhood AML blasts [Creutzig et al. 1995]. CD33 is also present on normal maturing hematopoietic progenitor cells, but is not present on hematopoietic stem cells or other normal tissue, making it an attractive target [Sievers et al. 2001]. Gemtuzumab (CMA-676 or GO) is an anti-CD33 antibody conjugated to calicheamicin, a potent antitumor antibiotic that cleaves double-stranded DNA at specific sequences, leading to apoptosis [Zein et al. 1988; Hinman et al. 1993; Sievers et al. 1999]. COG phase III study AAML0531 incorporated GO in a randomized fashion in combination with standard chemotherapy in de novo AML. Results showed a modest improvement in 3-year EFS in the GO arm (53% versus 47%, p = 0.05) and a trend toward improved OS [Aplenc et al. 2013]. However, GO was withdrawn from the US market in 2010 after adults receiving GO in induction therapy experienced increased mortality, despite recently published studies of GO in children [Burnett et al. 2011; Cooper et al. 2012; Hasle et al. 2012] and adults [Castaigne et al. 2012] showing that a dose of 3 mg/m2 has been well tolerated. Furthermore, use of GO as consolidation therapy after HSCT in AML patients [Roman et al. 2005], or as part of a conditioning regimen for HSCT [Satwani et al. 2012] in early phase studies demonstrated safety in both settings. Reports also demonstrate reduction of MRD status with GO in otherwise refractory pediatric AML patients, thereby optimizing disease status prior to HSCT [Rubnitz et al. 2010; O’Hear et al. 2013]. These reports provide good rationale to consider ‘resurrecting’ GO [Ravandi et al. 2012].
CD22
CD22 is expressed in 96% of pre-B ALL patients on at least 90% of blasts [Gudowius et al. 2006]. Epratuzumab is a humanized monoclonal anti-CD22 antibody, and cells demonstrate rapid intracellular localization of the antibody complex after binding [Carnahan et al. 2003]. COG phase I/II study ADVL04P2 combined epratuzumab with reinduction chemotherapy in children with CD22+ relapsed ALL, and response was assessed after block one. In the phase II portion of the study, 65% of 98 evaluable patients achieved a CR, and 46% of these were MRD negative [Raetz et al. 2011]. This was significantly higher than the 25% of CR patients achieving negative MRD with chemotherapy alone on predecessor study AALL01P2 (p = 0.001). The upcoming European phase III IntReALL study will randomize pediatric ALL patients in standard risk first relapse to receive epratuzumab.
Since CD22 is internalized upon binding of monoclonal antibody, it is attractive for targeted delivery of immunotoxin [Press et al. 1989]. Inotuzumab ozogamicin (CMC-544 or IO) is an anti-CD22 humanized monoclonal antibody conjugated to calicheamicin, similar to anti-CD33 GO [DiJoseph et al. 2004]. IO was originally developed for use in CD22-expressing non-Hodgkin lymphoma (NHL), and two early phase trials of IO in adults with refractory NHL showed promising clinical responses as a single agent [Advani et al. 2010] and in combination with rituximab [Fayad et al. 2013]. Preclinical evidence with pre-B ALL cell lines and mouse models demonstrated that IO has good cytotoxic effect against CD22+ lymphoblasts in vitro, and abrogates xenograft tumors and disseminated leukemia in vivo [Dijoseph et al. 2007]. IO demonstrated clinical activity in a phase II study of adults and children with relapsed/refractory CD22+ pre-B ALL at dosing of 1.3–1.8 mg/m2 every 3–4 weeks [Kantarjian et al. 2012]. Of 49 patients, 28 (57%) achieved CR, CRi or complete response with incomplete platelet recovery (CRp; platelets <100 × 109/L). An additional 41 patients were enrolled at 1.8 mg/m2 divided into a weekly dosing regimen (0.8, 0.5, and 0.5 mg/m2/week) and had a similar overall response rate of 59% [Kantarjian et al. 2013]. The most common toxicities were grade 1–2 fever, hypotension, elevated transaminases and hyperbilirubinemia. A retrospective review of the five pediatric patients on this trial demonstrated good tolerability and activity at both dosing schedules [Rytting et al. 2014]. Three of five patients achieved a CR or CRp and went to HSCT, including one patient who had achieved an MRD negative status after two cycles of the weekly dosing schedule. Current trials are investigating IO in adults with ALL in relapsed disease (phase III trial [ClinicalTrials.gov identifier: NCT01564784]), in the elderly population (phase I/II trial [ClinicalTrials.gov identifier: NCT01371630]), and prior to allogeneic transplant (phase I/II trial [ClinicalTrials.gov identifier: NCT01664910]). There are no open pediatric trials investigating IO at the time of this review.
Moxetumomab pasudotox (MP, previously CA-8015 or HA22) is a second-generation anti-CD22 immunotoxin, composed of the variable fragment of the antibody (Fv) conjugated to a protein derivative (PE38) of Pseudomonas exotoxin A [Pastan et al. 2006]. BL22, the first-generation predecessor of moxetumomab, demonstrated cytotoxicity against malignant CD22+ cells in vitro [Kreitman et al. 2000] and clinical activity in a phase I trial of pediatric ALL patients [Wayne et al. 2010]. Likewise, moxetumomab demonstrated good in vitro activity against relapsed, steroid-resistant, and de novo pediatric pre-B ALL samples [Mussai et al. 2010]. Immunotoxin-resistant ALL cells were found to be heavily hypermethylated at the DPH4 promoter, rendering these cells refractory to moxetumomab, and suggesting a role for concurrent demethylating agents [Wei et al. 2012]. A phase I study of moxetumomab in CD22+ pediatric malignancies is ongoing, and preliminary results report 4 CRs in 17 evaluable patients (24%), 1 partial response (6%) and 7 patients (41%) with >50% reduction in peripheral blood blasts or improvement in blood counts [Wayne et al. 2011]. Toxicities were generally mild and reversible, although capillary leak syndrome was dose limiting in several patients. Fourteen percent of patients developed neutralizing antibodies, necessitating further study of the potential challenges of this drug. A phase II multicenter study of moxetumomab recently opened in August 2014 for relapsed/refractory pre-B ALL or B-cell lymphoblastic lymphoma.
CD19
Blinatumomab is a CD19/CD3 bispecific T-cell engaging (BiTE) antibody that binds to CD3+ T-cells and colocalizes them with CD19+ B-cells, thereby activating the T-cells and inducing perforin-mediated death of the targeted B-cells [Loffler et al. 2000]. After promising results in a phase I trial of adults with NHL [Bargou et al. 2008], blinatumomab was moved in a phase II study in adult pre-B ALL. This study yielded striking results; of 20 evaluable patients with detectable MRD by polymerase chain reaction (PCR), 16 (12 primary refractory and 4 relapsed) achieved an MRD negative remission. Eight of these 16 patients received a HSCT and remained in remission post-transplant. Common adverse events were pyrexia, chills and lymphopenia with hypogammaglobulinemia [Topp et al. 2011]. At a median follow-up of 33 months, relapse-free survival in these patients was 61% overall, with 6 relapses reported [Topp et al. 2012]. Of 4 earlier relapses (at 3–7 months), two had CD19-negative marrow disease, one had an isolated CNS relapse, and one had a testicular relapse. Interestingly, the CNS and testes are relatively T-cell deplete compartments. The two later relapses (at 19 and 31 months) were both CD19+ [Topp et al. 2012]. Importantly, four responders received no subsequent therapy yet remain in remission, suggesting that as a single agent, blinatumomab can induce a durable remission in MRD-positive disease. Remarkably, the effector memory T-cell subset was amplified in all patients, which could partially explain the duration of response in these patients, though amplification occurred in non-responders as well [Klinger et al. 2012]. The US Intergroup ECOG is currently investigating blinatumomab in upfront adult pre-B ALL in a phase III clinical trial [ClinicalTrials.gov identifier: NCT02003222].
In children, Handgretinger and colleagues evaluated blinatumomab monotherapy in a small case series of heavily pretreated, post-HSCT pediatric ALL patients. All three of three children achieved negative MRD with tolerable and similar toxicities. Two of three ultimately relapsed, one after a second transplant, and the other after four courses of blinatumomab. The third patient received a second transplant and remained in a CR at 23 months [Handgretinger et al. 2011]. A multicenter phase I trial in 34 children with relapsed/refractory pre-B ALL administered blinatumomab as a continuous 4-week infusion, and established the maximum tolerated dose (MTD) as 15 µg/m2/day [Zugmaier et al. 2013]. The dose-limiting toxicity was cytokine-release syndrome. Across all dose levels, the overall response was 41% [Zugmaier et al. 2013]. A randomized COG phase III study for pre-B ALL children in first relapse is scheduled to open in late 2014, combining blinatumomab with UKALLR3 reinduction chemotherapy.
Cellular-based immunotherapy
Chimeric antigen receptor-modified T-cells (CAR T-cells) with CD19 specificity are generating excitement as a novel therapy for B-cell malignancies. CAR T cells are patient-derived T-cells, transduced to express a chimeric antigen receptor, which includes an anti-CD19 antibody fragment fused to a T-cell intracellular signaling domain [Barrett et al. 2014]. Second-generation CAR T cells also encode for a costimulatory domain, such as CD28 or members of the tumor necrosis factor receptor family such as CD27, CD137 (4-1BB) and CD134 (OX40). The costimulatory domains activate the CAR T-cells, allowing for targeting and lysis of CD19+ cells. Clinical trials with second-generation CD19 CAR T-cells in adult chronic lymphocytic leukemia (CLL) demonstrated massive T-cell expansion in vivo, lysis of CD19+ tumor cells, and aplasia of normal CD19+ B cells [Kalos et al. 2011]. Importantly, these relapsed and refractory CLL patients demonstrated durable remissions after infusions of CD19 CAR T-cells.
Several institutions in the United States have open pediatric trials of CD19 CAR T-cells in pre-B cell ALL. The first two pediatric patients treated at Children’s Hospital of Philadelphia (CHOP) with refractory/relapsed pre-B ALL achieved a CR within a month of CAR T-cell infusion [Grupp et al. 2013]. In both patients, dramatic expansion of CAR T-cells was documented in the peripheral blood, peaking around day 10. Surprisingly, CAR T-cells were detected in the CNS in both patients. Toxicity related to a cytokine-release syndrome after CAR T-cell infusions is significant, and in severe cases can be reminiscent of macrophage-activation syndrome. However, anticytokine therapy with the anti-IL6 monoclonal antibody, tocilizumab, can rapidly induce clinical improvement in this setting [Grupp et al. 2013]. As expected, complete B-cell aplasia occurs, and the duration of this effect is undefined and must be supported with intravenous immunoglobulin (IVIG) administration. Of the original two pediatric patients, one remains in remission without any subsequent disease-directed therapy. The other patient relapsed in 2 months with a CD19-negative clone, signifying the strong selective pressure of this therapy.
At the American Society of Hematology (ASH) 2013 annual meeting, the CHOP group presented their updated outcomes on this trial, having now enrolled 16 pediatric and 4 adult patients with CD19+ leukemia. A total of 14 of 17 evaluable patients (82%) achieved a CR at 1 month. Three of these 14 CR patients subsequently relapsed. Aside from the patient mentioned above, the other two relapsed patients did not have a CD19 negative clone. All patients have demonstrated some sort of cytokine release syndrome corresponding with the peak T-cell expansion in vivo [Frey et al. 2013]. Seven of 20 required treatment for respiratory or hemodynamic instability, and all improved rapidly after tocilizumab given with or without steroids. In all patients with durable responses, CAR T-cells were detectable by flow cytometry for a range of 1–15 months. Given these encouraging results, multicenter pediatric trials are now being planned to investigate CD19 CAR T-cells in a phase II setting.
Finally, a phase I clinical trial of CD22 CAR T-cells for pediatric patients with relapsed/refractory ALL is currently in development, based on promising preclinical data [Haso et al. 2013].
Conclusion
There is compelling rationale for the ongoing discovery and implementation of novel and targeted agents in pediatric leukemia. Future goals of treatment for pediatric ALL and AML, as well as other forms of cancer, are to personalize treatment to the specific molecular aberrancies driving each individual’s disease, while decreasing the intensity and toxicities of conventional chemotherapy regimens. Pediatric leukemia is a disease prototype for this area of translational and clinical research, given the relatively smaller number of cumulative ‘hits’ necessary for malignant transformation. However, there are significant challenges to the development of novel agents. The process of drug development, including enrolling adequate numbers of patients to complete clinical trials, will be particularly challenging when only specific subgroups of leukemia patients are appropriate to study for each new agent. As mentioned previously, incorporating a new agent in the safest and most effective manner into existing chemotherapy regimens is complex, and this complexity will undoubtedly increase as combinations of targeted agents are proposed; particularly since another challenge is the development of resistance that occurs upon applying strong selective pressure with targeted agents. Finally, newer targeted agents must be effective in eradicating not only the bulk leukemia population, but also the elusive and often quiescent leukemic stem cell (LSC) population, in order to effectively prevent relapse and improve long-term survival. Despite these challenges, the ongoing discovery of oncogenic lesions in pediatric leukemia and the exciting parallel development of diverse novel therapeutic agents provide true promise for improving outcomes in high-risk and relapsed pediatric acute leukemia.
Footnotes
Conflict of interest statement: The authors declare that there is no conflict of interest.
Funding: This work was supported by St. Baldrick’s Foundation Fellowship Awards (C.E.A.), Leukemia and Lymphoma Society Scholar Award (P.B.), and American Cancer Society Research Scholar Grant (P.B.).
Contributor Information
Colleen E. Annesley, Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
Patrick Brown, Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB-I 2M46, Baltimore, MD 21231, USA.
References
- Advani A., Coiffier B., Czuczman M., Dreyling M., Foran J., Gine E., et al. (2010) Safety, pharmacokinetics, and preliminary clinical activity of inotuzumab ozogamicin, a novel immunoconjugate for the treatment of B-cell non-Hodgkin’s lymphoma: results of a phase I study. J Clin Oncol 28: 2085–2093. [DOI] [PubMed] [Google Scholar]
- Aplenc R., Alonzo T., Sung L., Meshinchi S., Gerbing R., Raimondi S., et al. (2013) Gemtuzumab Ozogamicin (GO) in children with De Novo Acute Myeloid Leukemia (AML) improves Event-Free Survival (EFS) by reducing relapse risk – results from the randomized Phase III Children’s Oncology Group (COG) trial, AAML0531. Blood 122: 355–355. [Google Scholar]
- Arastu-Kapur S., Anderl J., Kraus M., Parlati F., Shenk K., Lee S., et al. (2011) Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a link to clinical adverse events. Clin Cancer Res 17: 2734–2743. [DOI] [PubMed] [Google Scholar]
- Armstrong S., Staunton J., Silverman L., Pieters R., den Boer M., Minden M., et al. (2002) MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Gen 30: 41–47. [DOI] [PubMed] [Google Scholar]
- Attar E., De Angelo D., Supko J., D’Amato F., Zahrieh D., Sirulnik A., et al. (2008) Phase I and pharmacokinetic study of bortezomib in combination with idarubicin and cytarabine in patients with acute myelogenous leukemia. Clin Cancer Res 14: 1446–1454. [DOI] [PubMed] [Google Scholar]
- Ayton P., Cleary M. (2001) Molecular mechanisms of leukemogenesis mediated by MLL fusion proteins. Oncogene 20: 5695–5707. [DOI] [PubMed] [Google Scholar]
- Bachmann P., Piazza R., Janes M., Wong N., Davies C., Mogavero A., et al. (2010) Epigenetic silencing of BIM in glucocorticoid poor-responsive pediatric acute lymphoblastic leukemia, and its reversal by histone deacetylase inhibition. Blood 116: 3013–3022. [DOI] [PubMed] [Google Scholar]
- Bargou R., Leo E., Zugmaier G., Klinger M., Goebeler M., Knop S., et al. (2008) Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321: 974–977. [DOI] [PubMed] [Google Scholar]
- Barrett D., Singh N., Porter D., Grupp S., June C. (2014) Chimeric antigen receptor therapy for cancer. Ann Rev Med 65: 333–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernt K., Zhu N., Sinha A., Vempati S., Faber J., Krivtsov A., et al. (2011) MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 20: 66–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhatla T., Wang J., Morrison D., Raetz E., Burke M., Brown P., et al. (2012) Epigenetic reprogramming reverses the relapse-specific gene expression signature and restores chemosensitivity in childhood B-lymphoblastic leukemia. Blood 119: 5201–5210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown V., Fang J., Alcorn K., Barr R., Kim J., Wasserman R., et al. (2003) Rapamycin is active against B-precursor leukemia in vitro and in vivo, an effect that is modulated by IL-7-mediated signaling. Proc Natl Acad Sci USA 100: 15113–15118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burnett A., Hills R., Milligan D., Kjeldsen L., Kell J., Russell N., et al. (2011) Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial. J Clin Oncol 29: 369–377. [DOI] [PubMed] [Google Scholar]
- Buttner N., Johnsen S., Kugler S., Vogel T. (2010) Af9/Mllt3 interferes with Tbr1 expression through epigenetic modification of histone H3K79 during development of the cerebral cortex. Proc Natl Acad Sci USA 107: 7042–7047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carnahan J., Wang P., Kendall R., Chen C., Hu S., Boone T., et al. (2003) Epratuzumab, a humanized monoclonal antibody targeting CD22: characterization of in vitro properties. Clin Cancer Res 9: 3982S–3990S. [PubMed] [Google Scholar]
- Carow C., Levenstein M., Kaufmann S., Chen J., Amin S., Rockwell P., et al. (1996) Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias. Blood 87: 1089–1096. [PubMed] [Google Scholar]
- Castaigne S., Pautas C., Terre C., Raffoux E., Bordessoule D., Bastie J., et al. (2012) Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet 379: 1508–1516. [DOI] [PubMed] [Google Scholar]
- Chang B., Willis S., Stork L., Hunger S., Carroll W., Camitta B., et al. (2012) Imatinib resistant BCR-ABL1 mutations at relapse in children with Ph+ ALL: a Children’s Oncology Group (COG) study. Br J Haematol 157: 507–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang M., Wu H., Achille N., Reisenauer M., Chou C., Zeleznik-Le N., et al. (2010) Histone H3 lysine 79 methyltransferase Dot1 is required for immortalization by MLL oncogenes. Cancer Res 70: 10234–10242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen L., Deshpande A., Banka D., Bernt K., Dias S., Buske C., et al. (2013) Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1l. Leukemia 27: 813–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cooper T., Franklin J., Gerbing R., Alonzo T., Hurwitz C., Raimondi S., et al. (2012) AAML03P1, a pilot study of the safety of gemtuzumab ozogamicin in combination with chemotherapy for newly diagnosed childhood acute myeloid leukemia: a report from the Children’s Oncology Group. Cancer 118: 761–769. [DOI] [PubMed] [Google Scholar]
- Cooper, T., Malvar J., Cassar J., Eckroth E., Sposto R., Gaynon P., Dubois S., et al. (2013) A phase I study of AC220 (Quizartinib) in combination with cytarabine and etoposide in relapsed/refractory childhood ALL and AML: a therapeutic advances in childhood leukemia & lymphoma (TACL) study. Blood 122: 624–624.23908442 [Google Scholar]
- Cortes J., Kim D., Pinilla-Ibarz J., le Coutre P., Paquette R., Chuah C., et al. (2013) A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N Engl J Med 369: 1783–1796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Creutzig U., Harbott J., Sperling C., Ritter J., Zimmermann M., Loffler H., et al. (1995) Clinical significance of surface antigen expression in children with acute myeloid leukemia: results of study AML-BFM-87. Blood 86: 3097–3108. [PubMed] [Google Scholar]
- Crump M., Hedley D., Kamel-Reid S., Leber B., Wells R., Brandwein J., et al. (2010) A randomized phase I clinical and biologic study of two schedules of sorafenib in patients with myelodysplastic syndrome or acute myeloid leukemia: a NCIC (National Cancer Institute of Canada) Clinical Trials Group Study. Leukemia Lymphoma 51: 252–260. [DOI] [PubMed] [Google Scholar]
- Daigle S., Olhava E., Therkelsen C., Majer C., Sneeringer C., Song J., et al. (2011) Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20(1): 53–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daver N., Cortes J. (2012) Molecular targeted therapy in acute myeloid leukemia. Hematology 17(Suppl. 1): S59–S62. [DOI] [PubMed] [Google Scholar]
- Deshpande A., Chen L., Fazio M., Sinha A., Bernt K., Banka D., et al. (2013) Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l. Blood 121:2533–2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiJoseph J., Armellino D., Boghaert E., Khandke K., Dougher M., Sridharan L., et al. (2004) Antibody-targeted chemotherapy with CMC-544: a CD22-targeted immunoconjugate of calicheamicin for the treatment of B-lymphoid malignancies. Blood 103: 1807–1814. [DOI] [PubMed] [Google Scholar]
- Dijoseph J., Dougher M., Armellino D., Evans D., Damle N. (2007) Therapeutic potential of CD22-specific antibody-targeted chemotherapy using inotuzumab ozogamicin (CMC-544) for the treatment of acute lymphoblastic leukemia. Leukemia 21(11): 2240–2245. [DOI] [PubMed] [Google Scholar]
- Fandy T., Herman J., Kerns P., Jiemjit A., Sugar E., Choi S., et al. (2009) Early epigenetic changes and DNA damage do not predict clinical response in an overlapping schedule of 5-azacytidine and entinostat in patients with myeloid malignancies. Blood 114: 2764–2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fayad L., Offner F., Smith M., Verhoef G., Johnson P., Kaufman J., et al. (2013) Safety and clinical activity of a combination therapy comprising two antibody-based targeting agents for the treatment of non-Hodgkin lymphoma: results of a phase I/II study evaluating the immunoconjugate inotuzumab ozogamicin with rituximab. J Clin Oncol 31: 573–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fischer T., Stone R., Deangelo D., Galinsky I., Estey E., Lanza C., et al. (2010) Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol 28: 4339–4345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fouladi M., Park J., Stewart C., Gilbertson R., Schaiquevich P., Sun J., et al. (2010) Pediatric phase I trial and pharmacokinetic study of vorinostat: a Children’s Oncology Group phase I consortium report. J Clin Oncol 28: 3623–3629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frey N., Aplenc R., Barrett D., Chew A., Kalos M., Levine B., et al. (2013) T cells engineered with a chimeric antigen receptor (CAR) targeting CD19 (CTL019) produce significant in vivo proliferation, complete responses and long-term persistence without GVHD in children and adults with relapsed, refractory ALL. Blood 122: 67. [Google Scholar]
- Garcia-Manero G., Assouline S., Cortes J., Estrov Z., Kantarjian H., Yang H., et al. (2008) Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood 112: 981–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Manero G., Kantarjian H., Sanchez-Gonzalez B., Yang H., Rosner G., Verstovsek S., et al. (2006) Phase 1/2 study of the combination of 5-aza-2’-deoxycytidine with valproic acid in patients with leukemia. Blood 108: 3271–3279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gore S. (2005) Combination therapy with DNA methyltransferase inhibitors in hematologic malignancies. Nat Clin Pract Oncol 2(Suppl. 1): S30–S35. [DOI] [PubMed] [Google Scholar]
- Gore S., Baylin S., Sugar E., Carraway H., Miller C., Carducci M., et al. (2006) Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res 66: 6361–6369. [DOI] [PubMed] [Google Scholar]
- Grupp S., Kalos M., Barrett D., Aplenc R., Porter D., Rheingold S., et al. (2013) Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gudowius S., Recker K., Laws H., Dirksen U., Troger A., Wieczorek U., et al. (2006) Identification of candidate target antigens for antibody-based immunotherapy in childhood B-cell precursor ALL. Klin Padiatrie 218: 327–333. [DOI] [PubMed] [Google Scholar]
- Guzman M., Neering S., Upchurch D., Grimes B., Howard D., Rizzieri D., et al. (2001) Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 98: 2301–2307. [DOI] [PubMed] [Google Scholar]
- Guzman M., Swiderski C., Howard D., Grimes B., Rossi R., Szilvassy S., et al. (2002) Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl Acad Sci USA 99: 16220–16225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Handgretinger R., Zugmaier G., Henze G., Kreyenberg H., Lang P., von Stackelberg A. (2011) Complete remission after blinatumomab-induced donor T-cell activation in three pediatric patients with post-transplant relapsed acute lymphoblastic leukemia. Leukemia 25: 181–184. [DOI] [PubMed] [Google Scholar]
- Hasle H., Abrahamsson J., Forestier E., Ha S., Heldrup J., Jahnukainen K., et al. (2012) Gemtuzumab ozogamicin as postconsolidation therapy does not prevent relapse in children with AML: results from NOPHO-AML 2004. Blood 120: 978–984. [DOI] [PubMed] [Google Scholar]
- Haso W., Lee D., Shah N., Stetler-Stevenson M., Yuan C., Pastan I., et al. (2013) Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood 121: 1165–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herman J., Civin C., Issa J., Collector M., Sharkis S., Baylin S. (1997) Distinct patterns of inactivation of p15INK4B and p16INK4A characterize the major types of hematological malignancies. Cancer Res 57: 837–841. [PubMed] [Google Scholar]
- Hinman L., Hamann P., Wallace R., Menendez A., Durr F., Upeslacis J. (1993) Preparation and characterization of monoclonal antibody conjugates of the calicheamicins: a novel and potent family of antitumor antibiotics. Cancer Res 53: 3336–3342. [PubMed] [Google Scholar]
- Horton T., Pati D., Plon S., Thompson P., Bomgaars L., Adamson P., et al. (2007) A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children’s Oncology Group study. Clin Cancer Res 13: 1516–1522. [DOI] [PubMed] [Google Scholar]
- Hunger S., Lu X., Devidas M., Camitta B., Gaynon P., Winick N., et al. (2012) Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children’s oncology group. J Clin Oncol 30: 1663–1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inaba H., Rubnitz J., Coustan-Smith E., Li L., Furmanski B., Mascara G., et al. (2011) Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia. J Clin Oncol 29: 3293–3300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwai T., Yokota S., Nakao M., Okamoto T., Taniwaki M., Onodera N., et al. (1999) Internal tandem duplication of the FLT3 gene and clinical evaluation in childhood acute myeloid leukemia. The Children’s Cancer and Leukemia Study Group, Japan. Leukemia 13: 38–43. [DOI] [PubMed] [Google Scholar]
- Jabbour E., Kantarjian H., Jones D., Reddy N., O’Brien S., Garcia-Manero G., et al. (2008) Characteristics and outcome of chronic myeloid leukemia patients with F317L BCR-ABL kinase domain mutation after therapy with tyrosine kinase inhibitors. Blood 112: 4839–4842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jayanthan A., Ruan Y., Hagerty M., Shah R., Truong T., Lewis V., et al. (2013) In vitro growth inhibition, target modulation and drug synergy in pediatric leukemia by the novel proteasome inhibitor carfilzomib. Blood 122: 2673–2673.24004666 [Google Scholar]
- Kalos M., Levine B., Porter D., Katz S., Grupp S., Bagg A., et al. (2011) T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3(95): 95ra73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantarjian H., Issa J., Rosenfeld C., Bennett J., Albitar M., DiPersio J., et al. (2006) Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer 106: 1794–1803. [DOI] [PubMed] [Google Scholar]
- Kantarjian H., Thomas D., Jorgensen J., Jabbour E., Kebriaei P., Rytting M., et al. (2012) Inotuzumab ozogamicin, an anti-CD22-calecheamicin conjugate, for refractory and relapsed acute lymphocytic leukaemia: a phase 2 study. The Lancet Oncology 13(4): 403–411. [DOI] [PubMed] [Google Scholar]
- Kantarjian H., Thomas D., Jorgensen J., Kebriaei P., Jabbour E., Rytting M., et al. (2013) Results of inotuzumab ozogamicin, a CD22 monoclonal antibody, in refractory and relapsed acute lymphocytic leukemia. Cancer 119: 2728–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klinger M., Brandl C., Zugmaier G., Hijazi Y., ÍBargou R., Topp M., et al. (2012) Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood 119: 6226–6233. [DOI] [PubMed] [Google Scholar]
- Knapper S., Burnett A., Littlewood T., Kell W., Agrawal S., Chopra R., et al. (2006) 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 108: 3262–3270. [DOI] [PubMed] [Google Scholar]
- Kondo M., Horibe K., Takahashi Y., Matsumoto K., Fukuda M., Inaba J., et al. (1999) Prognostic value of internal tandem duplication of the FLT3 gene in childhood acute myelogenous leukemia. Med Pediatric Oncol 33: 525–529. [DOI] [PubMed] [Google Scholar]
- Kramer O., Muller S., Buchwald M., Reichardt S., Heinzel T. (2008) Mechanism for ubiquitylation of the leukemia fusion proteins AML1-ETO and PML-RARalpha. FASEB J 22: 1369–1379. [DOI] [PubMed] [Google Scholar]
- Kreitman R., Margulies I., Stetler-Stevenson M., Wang Q., FitzGerald D., Pastan I. (2000) Cytotoxic activity of disulfide-stabilized recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) toward fresh malignant cells from patients with B-cell leukemias. Clin Cancer Res 6: 1476–1487. [PubMed] [Google Scholar]
- Krivtsov A., Feng Z., Lemieux M., Faber J., Vempati S., Sinha A., et al. (2008) H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell 14: 355–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krivtsov A., Twomey D., Feng Z., Stubbs M., Wang Y., Faber J., et al. (2006) Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442: 818–822. [DOI] [PubMed] [Google Scholar]
- Lacoste N., Utley R., Hunter J., Poirier G., Cote J. (2002) Disruptor of telomeric silencing-1 is a chromatin-specific histone H3 methyltransferase. J Biol Chem 277: 30421–30424. [DOI] [PubMed] [Google Scholar]
- Leclerc G., Mou C., Leclerc G., Mian A., Barredo J. (2010) Histone deacetylase inhibitors induce FPGS mRNA expression and intracellular accumulation of long-chain methotrexate polyglutamates in childhood acute lymphoblastic leukemia: implications for combination therapy. Leukemia 24: 552–562. [DOI] [PubMed] [Google Scholar]
- Levis M., Perl A., Dombret H., Dohner H., Steffen B., Rousselot P., et al. (2012) Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/Refractory Acute Myeloid Leukemia After Second-Line Chemotherapy or Hematopoietic Stem Cell Transplantation. ASH Annual Meeting Abstracts 120(21): 673. [Google Scholar]
- Levis M., Ravandi F., Wang E., Baer M., Perl A., Coutre S., et al. (2011) Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood 117: 3294–3301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loffler A., Kufer P., Lutterbuse R., Zettl F., Daniel P., Schwenkenbecher J., et al. (2000) A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood 95: 2098–2103. [PubMed] [Google Scholar]
- Lucas D., Alinari L., West D., Davis M., Edwards R., Johnson A., et al. (2010) The novel deacetylase inhibitor AR-42 demonstrates pre-clinical activity in B-cell malignancies in vitro and in vivo. PloS One 5(6): e10941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maestre N., Tritton T., Laurent G., Jaffrezou J. (2001) Cell surface-directed interaction of anthracyclines leads to cytotoxicity and nuclear factor kappaB activation but not apoptosis signaling. Cancer Res 61: 2558–2561. [PubMed] [Google Scholar]
- Maude S., Tasian S., Vincent T., Hall J., Sheen C., Roberts K., et al. (2012) Targeting JAK1/2 and mTOR in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood 120: 3510–3518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meshinchi S., Alonzo T., Stirewalt D., Zwaan M., Zimmerman M., Reinhardt D., et al. (2006) Clinical implications of FLT3 mutations in pediatric AML. Blood 108: 3654–3661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meshinchi S., Woods W., Stirewalt D., Sweetser D., Buckley J., Tjoa T., et al. (2001) Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia. Blood 97: 89–94. [DOI] [PubMed] [Google Scholar]
- Messinger Y., Gaynon P., Raetz E., Hutchinson R., Dubois S., Glade-Bender J., et al. (2010) Phase I study of bortezomib combined with chemotherapy in children with relapsed childhood acute lymphoblastic leukemia (ALL): a report from the therapeutic advances in childhood leukemia (TACL) consortium. Pediatric Blood Cancer 55: 254–259. [DOI] [PubMed] [Google Scholar]
- Messinger Y., Gaynon P., Sposto R., van der Giessen J., Eckroth E., Malvar J., et al. (2012) Bortezomib with chemotherapy is highly active in advanced B-precursor acute lymphoblastic leukemia: therapeutic advances in childhood leukemia & lymphoma (TACL) study. Blood 120: 285–290. [DOI] [PubMed] [Google Scholar]
- Min Y., Eom J., Cheong J., Maeng H., Kim J., Jeung H., et al. (2003) Constitutive phosphorylation of Akt/PKB protein in acute myeloid leukemia: its significance as a prognostic variable. Leukemia 17(5): 995–997. [DOI] [PubMed] [Google Scholar]
- Mueller D., Bach C., Zeisig D., Garcia-Cuellar M., Monroe S., Sreekumar A., et al. (2007) A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood 110: 4445–4454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mullighan C., Zhang J., Harvey R., Collins-Underwood J., Schulman B., Phillips L., et al. (2009) JAK mutations in high-risk childhood acute lymphoblastic leukemia. Proc Natl Acad Sci USA 106: 9414–9418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mussai F., Campana D., Bhojwani D., Stetler-Stevenson M., Steinberg S., Wayne A., et al. (2010) Cytotoxicity of the anti-CD22 immunotoxin HA22 (CAT-8015) against paediatric acute lymphoblastic leukaemia. Br J Haematol 150: 352–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen A., Taranova O., He J., Zhang Y. (2011) DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis. Blood 117: 6912–6922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Hear C., Inaba H., Pounds S., Shi L., Dahl G., Bowman W., et al. (2013) Gemtuzumab ozogamicin can reduce minimal residual disease in patients with childhood acute myeloid leukemia. Cancer 119: 4036–4043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okada Y., Feng Q., Lin Y., Jiang Q., Li Y., Coffield V., et al. (2005) hDOT1L links histone methylation to leukemogenesis. Cell 121: 167–178. [DOI] [PubMed] [Google Scholar]
- Parker C., Waters R., Leighton C., Hancock J., Sutton R., Moorman A., et al. (2010) Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet 376: 2009–2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pastan I., Hassan R., Fitzgerald D., Kreitman R. (2006) Immunotoxin therapy of cancer. Nat Rev Cancer 6: 559–565. [DOI] [PubMed] [Google Scholar]
- Porkka K., Koskenvesa P., Lundan T., Rimpilainen J., Mustjoki S., Smykla R., et al. (2008) Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia. Blood 112: 1005–1012. [DOI] [PubMed] [Google Scholar]
- Pratz K., Cho E., Levis M., Karp J., Gore S., McDevitt M., et al. (2010) A pharmacodynamic study of sorafenib in patients with relapsed and refractory acute leukemias. Leukemia 24: 1437–1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Press O., Farr A., Borroz K., Anderson S., Martin P. (1989) Endocytosis and degradation of monoclonal antibodies targeting human B-cell malignancies. Cancer Res 49: 4906–4912. [PubMed] [Google Scholar]
- Pui C., Carroll W., Meshinchi S., Arceci R. (2011a) Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J Clin Oncol 29: 551–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pui C., Gajjar A., Kane J., Qaddoumi I., Pappo A. (2011b) Challenging issues in pediatric oncology. Nat Rev Clin Oncol 8: 540–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pulsipher M., Langholz B., Wall D., Schultz K., Bunin N., Carroll W., et al. (2014) The addition of sirolimus to tacrolimus/methotrexate GVHD prophylaxis in children with ALL: a phase 3 children’s oncology group/pediatric blood and marrow transplant consortium trial. Blood 123: 2017–2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raetz E., Cairo M., Borowitz M., Lu X., Devidas M., Reid J., et al. (2011) Reinduction chemoimmunotherapy with epratuzumab in relapsed acute lymphoblastic leukemia (ALL) in children, adolescents and young adults: results from children’s oncology group (COG) study ADVL04P2. ASH Annual Meeting Abstracts 118(21): 573. [Google Scholar]
- Rajkumar S., Richardson P., Hideshima T., Anderson K. (2005) Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol 23: 630–639. [DOI] [PubMed] [Google Scholar]
- Ravandi F., Cortes J., Jones D., Faderl S., Garcia-Manero G., Konopleva M., et al. (2010) Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 28: 1856–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravandi F., Estey E., Appelbaum F., Lo-Coco F., Schiffer C., Larson R., et al. (2012) Gemtuzumab ozogamicin: time to resurrect? J Clin Oncol 30: 3921–3923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Recher C., Beyne-Rauzy O., Demur C., Chicanne G., Dos Santos C., Mas V., et al. (2005a) Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 105: 2527–2534. [DOI] [PubMed] [Google Scholar]
- Recher C., Dos Santos C., Demur C., Payrastre B. (2005b) mTOR, a new therapeutic target in acute myeloid leukemia. Cell Cycle 4: 1540–1549. [DOI] [PubMed] [Google Scholar]
- Roberts K., Li Y., Payne-Turner D., Harvey R., Yang Y., Pei D., et al. (2014) Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med 371: 1005–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roman E., Cooney E., Harrison L., Militano O., Wolownik K., Hawks R., et al. (2005) Preliminary results of the safety of immunotherapy with gemtuzumab ozogamicin following reduced intensity allogeneic stem cell transplant in children with CD33+ acute myeloid leukemia. Clin Cancer Res 11: 7164s–7170s. [DOI] [PubMed] [Google Scholar]
- Ruan Y., Liu D., Jayanthan A., Truong T., Boklan J., Narendran A. (2013) Abstract C205: Cytotoxicity and target modulation in pediatric solid tumors by the proteasome inhibitor carfilzomib. Mol Cancer Ther 12(Suppl. 11): C205–C205. [DOI] [PubMed] [Google Scholar]
- Rubnitz J., Inaba H., Dahl G., Ribeiro R., Bowman W., Taub J., et al. (2010) Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial. Lancet Oncol 11: 543–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rytting M., Triche L., Thomas D., O’Brien S., Kantarjian H. (2014) Initial experience with CMC-544 (inotuzumab ozogamicin) in pediatric patients with relapsed B-cell acute lymphoblastic leukemia. Pediatric Blood Cancer 61: 369–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santi D., Norment A., Garrett C. (1984) Covalent bond formation between a DNA-cytosine methyltransferase and DNA containing 5-azacytosine. Proc Natl Acad Sci USA 81: 6993–6997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Satwani P., Bhatia M., Garvin J., Jr, George D., Dela Cruz F., Le Gall J., et al. (2012) A phase I study of gemtuzumab ozogamicin (GO) in combination with busulfan and cyclophosphamide (Bu/Cy) and allogeneic stem cell transplantation in children with poor-risk CD33+ AML: a new targeted immunochemotherapy myeloablative conditioning (MAC) regimen. Biol Blood Marrow Transplant 18: 324–329. [DOI] [PubMed] [Google Scholar]
- Schultz K., Bowman W., Aledo A., Slayton W., Sather H., Devidas M., et al. (2009) Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a children’s oncology group study. J Clin Oncol 27: 5175–5181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schultz K., Carroll A., Heerema N., Bowman W., Aledo A., Slayton W., et al. (2014) Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: children’s oncology group study AALL0031. Leukemia 28: 1467–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seedhouse C., Das-Gupta E., Russell N. (2003) Methylation of the hMLH1 promoter and its association with microsatellite instability in acute myeloid leukemia. Leukemia 17: 83–88. [DOI] [PubMed] [Google Scholar]
- Sekimizu M., Yamashita Y., Ueki H., Akita N., Hattori H., Maeda N., et al. (2013) Nilotinib monotherapy induced complete remission in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia resistant to imatinib and dasatinib. Leukemia Lymphoma 55: 1652–1653. [DOI] [PubMed] [Google Scholar]
- Siegel D., Martin T., Wang M., Vij R., Jakubowiak A., Lonial S., et al. (2012) A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma. Blood 120: 2817–2825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sievers E., Appelbaum F., Spielberger R., Forman S., Flowers D., Smith F., et al. (1999) Selective ablation of acute myeloid leukemia using antibody-targeted chemotherapy: a phase I study of an anti-CD33 calicheamicin immunoconjugate. Blood 93: 3678–3684. [PubMed] [Google Scholar]
- Sievers E., Larson R., Stadtmauer E., Estey E., Lowenberg B., Dombret H., et al. (2001) Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J Clin Oncol 19: 3244–3254. [DOI] [PubMed] [Google Scholar]
- Silverman L., Demakos E., Peterson B., Kornblith A., Holland J., Odchimar-Reissig R., et al. (2002) Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol 20: 2429–2440. [DOI] [PubMed] [Google Scholar]
- Small D., Levenstein M., Kim E., Carow C., Amin S., Rockwell P., et al. (1994) 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 91: 459–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith B., Levis M., Beran M., Giles F., Kantarjian H., Berg K., et al. (2004) Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 103: 3669–3676. [DOI] [PubMed] [Google Scholar]
- Soriano A., Yang H., Faderl S., Estrov Z., Giles F., Ravandi F., et al. (2007) Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood 110: 2302–2308. [DOI] [PubMed] [Google Scholar]
- Soverini S., De Benedittis C., Papayannidis C., Paolini S., Venturi C., Iacobucci I., et al. (2014) Drug resistance and BCR-ABL kinase domain mutations in Philadelphia chromosome-positive acute lymphoblastic leukemia from the imatinib to the second-generation tyrosine kinase inhibitor era: The main changes are in the type of mutations, but not in the frequency of mutation involvement. Cancer 120: 1002–1009. [DOI] [PubMed] [Google Scholar]
- Stone R., DeAngelo D., Klimek V., Galinsky I., Estey E., Nimer S., et al. (2005) Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood 105: 54–60. [DOI] [PubMed] [Google Scholar]
- Stresemann C., Bokelmann I., Mahlknecht U., Lyko F. (2008) Azacytidine causes complex DNA methylation responses in myeloid leukemia. Mol Cancer Ther 7: 2998–3005. [DOI] [PubMed] [Google Scholar]
- Stresemann C., Brueckner B., Musch T., Stopper H., Lyko F. (2006) Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 66: 2794–2800. [DOI] [PubMed] [Google Scholar]
- Stumpel D., Schneider P., Seslija L., Osaki H., Williams O., Pieters R., et al. (2012) Connectivity mapping identifies HDAC inhibitors for the treatment of t(4;11)-positive infant acute lymphoblastic leukemia. Leukemia 26: 682–692. [DOI] [PubMed] [Google Scholar]
- Tallman M. (2004) Acute promyelocytic leukemia as a paradigm for targeted therapy. Sem Hematol 41(2 Suppl. 4): 27–32. [DOI] [PubMed] [Google Scholar]
- Talpaz M., Shah N., Kantarjian H., Donato N., Nicoll J., Paquette R., et al. (2006) Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med 354: 2531–2541. [DOI] [PubMed] [Google Scholar]
- Teachey D., Sheen C., Hall J., Ryan T., Brown V., Fish J., et al. (2008) mTOR inhibitors are synergistic with methotrexate: an effective combination totreat acute lymphoblastic leukemia. Blood 112: 2020–2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Topp M., Gokbuget N., Zugmaier G., Degenhard E., Goebeler M., Klinger M., et al. (2012) Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 120: 5185–5187. [DOI] [PubMed] [Google Scholar]
- Topp M., Kufer P., Gokbuget N., Goebeler M., Klinger M., Neumann S., et al. (2011) Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 29: 2493–2498. [DOI] [PubMed] [Google Scholar]
- Tsutsumi S., Taketani T., Nishimura K., Ge X., Taki T., Sugita K., et al. (2003) Two distinct gene expression signatures in pediatric acute lymphoblastic leukemia with MLL rearrangements. Cancer Res 63: 4882–4887. [PubMed] [Google Scholar]
- Vrana J., Decker R., Johnson C., Wang Z., Jarvis W., Richon V., et al. (1999) Induction of apoptosis in U937 human leukemia cells by suberoylanilide hydroxamic acid (SAHA) proceeds through pathways that are regulated by Bcl-2/Bcl-XL, c-Jun, and p21CIP1, but independent of p53. Oncogene 18: 7016–7025. [DOI] [PubMed] [Google Scholar]
- Wang C., Mayo M., Baldwin A., Jr (1996) TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB. Science 274: 784–787. [DOI] [PubMed] [Google Scholar]
- Wang M., Martin T., Bensinger W., Alsina M., Siegel D., Kavalerchik E., et al. (2013) Phase 2 dose-expansion study (PX-171-006) of carfilzomib, lenalidomide, and low-dose dexamethasone in relapsed or progressive multiple myeloma. Blood 122: 3122–3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wayne A., Bhojwani D., Silverman L., Richards K., Stetler-Stevenson M., Shah N., et al. (2011) A novel anti-CD22 immunotoxin, moxetumomab pasudotox: phase I study in pediatric acute lymphoblastic leukemia (ALL). ASH Annual Meeting Abstracts 118(21): 248. [Google Scholar]
- Wayne A., Kreitman R., Findley H., Lew G., Delbrook C., Steinberg S., et al. (2010) Anti-CD22 immunotoxin RFB4(dsFv)-PE38 (BL22) for CD22-positive hematologic malignancies of childhood: preclinical studies and phase I clinical trial. Clin Cancer Res 16: 1894–1903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei G., Twomey D., Lamb J., Schlis K., Agarwal J., Stam R., et al. (2006) Gene expression-based chemical genomics identifies rapamycin as a modulator of MCL1 and glucocorticoid resistance. Cancer Cell 10: 331–342. [DOI] [PubMed] [Google Scholar]
- Wei H., Xiang L., Wayne A., Chertov O., FitzGerald D., Bera T., et al. (2012) Immunotoxin resistance via reversible methylation of the DPH4 promoter is a unique survival strategy. Proc Natl Acad Sci USA 109: 6898–6903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weisberg E., Liu Q., Nelson E., Kung A., Christie A., Bronson R., et al. (2012) Using combination therapy to override stromal-mediated chemoresistance in mutant FLT3-positive AML: synergism between FLT3 inhibitors, dasatinib/multi-targeted inhibitors and JAK inhibitors. Leukemia 26: 2233–2244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Q., Simpson S., Scialla T., Bagg A., Carroll M. (2003) Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood 102: 972–980. [DOI] [PubMed] [Google Scholar]
- Yamamoto Y., Kiyoi H., Nakano Y., Suzuki R., Kodera Y., Miyawaki S., et al. (2001) Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 97:2434–2439. [DOI] [PubMed] [Google Scholar]
- Yang J., Wang Z., Fang Y., Jiang J., Zhao F., Wong H., et al. (2011) Pharmacokinetics, pharmacodynamics, metabolism, distribution, and excretion of carfilzomib in rats. Drug Metab Disposition 39: 1873–1882. [DOI] [PubMed] [Google Scholar]
- Yee K., Zeng Z., Konopleva M., Verstovsek S., Ravandi F., Ferrajoli A., et al. (2006) Phase I/II study of the mammalian target of rapamycin inhibitor everolimus (RAD001) in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res 12: 5165–5173. [DOI] [PubMed] [Google Scholar]
- Yokoyama A., Lin M., Naresh A., Kitabayashi I., Cleary M. (2010) A higher-order complex containing AF4 and ENL family proteins with P-TEFb facilitates oncogenic and physiologic MLL-dependent transcription. Cancer Cell 17: 198–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zein N., Sinha A., McGahren W., Ellestad G. (1988) Calicheamicin gamma 1I: an antitumor antibiotic that cleaves double-stranded DNA site specifically. Science 240: 1198–1201. [DOI] [PubMed] [Google Scholar]
- Zhang W., Konopleva M., Shi Y., McQueen T., Harris D., Ling X., et al. (2008) Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia. J Natl Cancer Inst 100: 184–198. [DOI] [PubMed] [Google Scholar]
- Zhang W., Xia X., Reisenauer M., Hemenway C., Kone B. (2006) Dot1a-AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner. J Biol Chem 281: 18059–18068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zugmaier G., Handgretinger R., Locatelli F., Rizzari C., Trippett T., Borkhardt A., et al. (2013) A phase 1/2 study of blinatumomab in pediatric patients with relapsed/refractory B-cell precursor acute lymphoblastic leukemia. Blood 122(21): 70. [Google Scholar]