Although the identification of cancer stem cells as therapeutic targets is now actively being pursued in many human malignancies, the leukemic stem cells in acute myeloid leukemia (AML) are a paradigm of such a strategy. Heterogeneity of these cells was suggested by clonal analyses indicating the existence of both leukemias resulting from transformed multipotent CD33 stem cells as well others arising from, or predominantly involving, committed CD33+ myeloid precursors. The latter leukemias, which may be associated with an intrinsically better prognosis, offer a particularly attractive target for stem cell-directed therapies. Targeting the CD33 differentiation antigen with gemtuzumab ozogamicin was the first attempt of such an approach. Emerging clinical data indicate that gemtuzumab ozogamicin is efficacious not only for acute promyelocytic leukemia but, in combination with conventional chemotherapy, also for other favorable- and intermediate-risk AMLs, providing the first proof-of-principle evidence for the validity of this strategy. Herein, we review studies on the nature of stem cells in AML, discuss clinical data on the effectiveness of CD33-directed therapy, and consider the mechanistic basis for success and failure in various AML subsets.

Normal human hematopoiesis is hierarchically organized, with tissue-specific, quiescent stem cells at the apex that have the ability to perpetuate themselves through self-renewal and generate more mature, transiently amplifying progeny through differentiation.1  Similar to normal hematopoiesis, acute myeloid leukemia (AML) encompasses functionally diverse cells, and origination from a leukemic stem cell (LSC) was initially suspected many decades ago.2  Observations in AML were indeed instrumental for the model of cancer stem cells as cells within a tumor that possess the capacity to self-renew and to cause the heterogeneous lineages of cancer cells that compose the tumor.3 

This model has important clinical implications as it predicts that the inability to eradicate cancer stem cells represents the cause of relapse and therapeutic failure; in turn, effective tumor therapy will require eradication of these cells.2,3  Interest in AML has thus long focused on the nature of LSCs and their specific qualities that predict therapeutic response. The cellular origin of AMLs, however, remains unclear, with ongoing controversy as to whether they arise from transformed hematopoietic stem cells (HSCs) or emerge as a result of genetic events occurring in more mature progenitor cells.2,4,,7  The nature of the cells giving rise to AML may have important biologic, therapeutic, and prognostic implications. Indeed, early recognition that some AMLs may predominantly or entirely involve committed myeloid progenitors led to efforts targeting underlying LSCs with antibodies recognizing the CD33 (SIGLEC-3) differentiation antigen, as exemplified by the development of the immunoconjugate, gemtuzumab ozogamicin (GO; Mylotarg).8  In this review, we summarize studies on stem cells in AML indicating heterogeneous involvement of stem/progenitor populations, discuss emerging data on the effectiveness of CD33-directed therapy, and consider the mechanistic basis for success or failure against individual AML subsets.

There may be no single, unifying cellular origin across the entire spectrum of human AML. Rather, research conducted over the last several decades indicates that AML may arise in (or predominantly involve) either multipotent HSCs or more mature committed myeloid precursors downstream of HSCs. The first hint to this heterogeneity came from early studies on X chromosome inactivation patterns, which identify the clonal cell population in females based on discrimination of the active from the inactive X chromosome and differentiation of each X chromosome's parental origin.9  In some leukemias, these investigations showed dominance of the clonal process in multiple cell lineages (granulocytes, monocytes, erythrocytes, platelets, and occasionally B lymphocytes), reflecting AML origination and expansion at the level of pluripotent stem/progenitor cells.10,11  In other cases, clonal dominance was limited to granulocytes and monocytes,10,11  suggesting that expansion of the malignant clone could occur at the level of committed myeloid precursors.

In the latter leukemias, we hypothesized that CD33 precursors (Figure 1) would be predominantly or completely normal. To test this assumption, we removed CD33+ cells in vitro via CD33-directed complement-mediated lysis or FACS in a small number of patients with such leukemias and placed the remaining CD33 cells in long-term culture together with irradiated allogeneic stroma cells.12,13  Over time, CD33 precursors from some patients indeed generated colony-forming cells (CFCs) with X chromosome inactivation patterns consistent with predominantly nonclonal hematopoiesis12,13 ; because of the inherent limitations of X chromosome inactivation studies,9  we could not distinguish complete from predominant nonclonal derivation. These findings were suggestive of transformation of lineage-committed myeloid precursors; however, restriction of the clonal dominance to granulocytes and monocytes could alternatively result from a mutated pluripotent stem cell that either lost the capacity for differentiation along the other cell lineages14  or only displayed dysregulated growth once single lineage commitment had occurred. Indeed, initial xenotransplantation assays, where only rare, immature CD34+/CD38 cells initiated and sustained leukemia growth in all AML subtypes except possibly acute promyelocytic leukemia (APL) suggested that pluripotent HSCs were generally the target of leukemic transformation.15,16  However, immunophenotypic differences in differentiation markers between normal HSCs and LSCs were noted by several investigators,17,19  and more recent xenotransplantation studies indicate that the transformation process may occur in precursor cells beyond the stem cell stage.20,22  In these investigations, AML was reconstituted in severely immunodeficient mice from cells that appeared phenotypically more mature than pluripotent HSCs and resided in the CD34+/CD38+ or CD34 compartment in some cases.20,21  Moreover, in a recent study suggesting that leukemia cells that engraft in immunodeficient mice and recapitulate human AML resemble hematopoietic progenitor cells phenotypically more closely than HSCs, the LSC-containing cell fraction also most closely resembled normal progenitor populations by gene expression profiling.22  It remains to be determined, however, to what degree this profile reflects the gene expression of the subset of LSCs contained in this cell population. Nevertheless, these more recent studies overall imply a greater variation in the cellular origin of transplantable AML cells than initially perceived and support the concept of LSC heterogeneity.

Figure 1

CD33 and CD34 as differentiation antigens. Simplified hypothetical model of stem and progenitor cells in the human hematopoietic system, showing expression patterns of CD33 and CD34.

Figure 1

CD33 and CD34 as differentiation antigens. Simplified hypothetical model of stem and progenitor cells in the human hematopoietic system, showing expression patterns of CD33 and CD34.

Close modal

Xenotransplantation assays have provided essential insight into AML biology and may currently be considered the gold standard to investigate LSCs on a functional level.2  There are important caveats, however: in particular, many patient specimens fail to engraft and are thus not amenable to study; there is also a bias toward specimens with large numbers of available cells. Thus, it is possible that xenotransplantation models underestimate LSC diversity and provide a skewed, incomplete assessment of these cells. Furthermore, although poorly understood, it is a recurrent observation that the ability to engraft in immunodeficient mice is a nonrandom, inherent property of specimens from patients with worse-prognosis AML (eg, those with unfavorable cytogenetics or aberrations in FLT3).23,,,,,29  Finally, some observations have raised the concern that cells without bona fide human-relevant stem cell properties may be transplantable in such model systems. This possibility is suggested by the recent finding that CD33+ cord blood cells can engraft with multilineage hematopoiesis.30  This is unexpected because previous studies on normal bone marrow showed CD33 expression primarily on multipotent myeloid precursors, unipotent CFCs, as well as maturing granulocytes, and monocytes but, unlike CD34, not on pluripotent HSCs (Figure 1).31,,34  These in vitro investigations were consistent with clinical studies demonstrating delayed but durable multilineage engraftment after transplantation of CD33-depleted autografts in patients with AML,35,36  similarly indicating that normal HSCs lack CD33. Thus, although xenotransplantation models are widely considered the most stringent approach to study the nature of stem cells, some of the findings may need to be interpreted cautiously.

Congruent with studies of human AMLs, experimental AML models in mice demonstrate that HSCs and committed myeloid precursors can be transformed into LSCs in the appropriate context. This was initially suggested by viral transduction studies with AML-associated oncogenic fusion proteins,37,,,41  although, extrapolation of these data might be limited because oncogene overexpression could lead to transformation in a nonphysiologic manner; recent findings using Mll-AF9 have indeed highlighted a critical role of oncogene dosage in hematopoietic cell transformation, with thresholds depending on the developmental stage of the progenitor cell.42  Nevertheless, data from knockin and transgenic mice suggest that committed myeloid progenitors can acquire stem cell properties even at physiologic expression levels of a mutated gene.43,44  The leukemias seen in such models are associated with enhanced self-renewal of committed myeloid progenitors and are transplantable via committed myeloid progenitors.43,44 

Relevant to any discussion of LSCs is the hypothesis that AML is generally the consequence of a multistep process rather than a single catastrophic event.45,46  Support for this assumption comes from mouse models in which single AML-associated somatic mutations lead to AML after long latency or exposure to chemical mutagens, whereas malignancies mimicking human AML can develop after short latencies when such mutations are coexpressed.43,45,,,49  In humans, the requirement for cooperating mutations is most directly suggested by patients with rare germline mutations in myeloid transcription factors who develop AML only after long (up to several decades) latency.50,52  Also informative are cases of core-binding factor (CBF) AMLs, that is, leukemias that are defined by the presence of chromosomal aberrations disrupting one of the CBF transcription factor genes, most commonly t(8;21)(q22;q22) and inv(16)(p13q22), which result in the generation of the RUNX1/RUNX1T1 (also known as AML1-ETO) and CBFB/MYH11 fusion proteins, respectively.53  The need for complementing mutations is indicated by the detection of AML1-ETO transcripts in neonatal Guthrie blood spots in some patients with nonfamilial CBF AML who only much later develop leukemia.54  Finally, epidemiologic data from sporadic AML demonstrate that many patients have more than one recurring nonrandom genetic abnormality, further suggesting that single mutations are insufficient to cause AML.46 

To date, the relative significance of causative mutations and target cell(s) of transformation are unclear, except for a few situations. Examples for the latter are patients with familial leukemia syndromes and probably those with nonfamilial CBF AML in whom AML1-ETO is detected many years before development of AML, suggesting its role as a founder mutation. Also consistent with the notion of a founder mutation, or at least a very early event, is the demonstration of AML1-ETO in other patients in long-term remission after therapy for CBF AML.55,,58  Furthermore, the observation that aberrant CEBPA, DNMT3A, IDH1/2, and possibly NPM1 alleles are typically present at diagnosis and relapse suggests that these could be very early, possibly preleukemic, mutational events.59,,62  In contrast, the mutation status of FLT3, NRAS/KRAS, and WT1 often changes between diagnosis and relapse, and mutations may only be present in a subset of leukemic blasts, indicating a role as secondary events.63,65  This role is supported by our recent studies on specimens from patients with CBF leukemias and concomitant mutations in either FLT3 or KIT, in which we found that many CFCs only harbor the CBF but not tyrosine kinase aberration.66 

Patients with familial leukemia syndromes or in utero origin of nonfamilial AMLs exemplify that complementing mutations leading to AML transformation can be acquired over prolonged time periods. Initial genetic event(s) may thus generate one or more long-lived, “preleukemic” stem cell(s) with establishment of preleukemic clones with self-renewal capacity. Such clones will require acquisition of additional mutation(s) for progression to a leukemic clone and the development of overt AML. The “indolent” persistence of preleukemic clones over time suggests origination in relatively quiescent stem cells with long-term repopulating capacity, with the mutational event affecting cellular differentiation rather than proliferation. It is plausible that one or several subsequent mutation(s) are then acquired in more mature progeny of the preleukemic stem cells. The detection of AML1-ETO transcripts in myeloid cells as well as B lymphocytes in some patients, indisputably demonstrating mutation acquisition at the level of pluripotent HSCs, is consistent with this notion.18  Thus, preleukemic stem cells and LSCs may coexist and vary in their differentiative and proliferative properties and perhaps their response to therapy, that is, persistence of preleukemic stem cells but eradication of fully transformed LSCs. This could explain why AML1-ETO transcripts remain detectable in the blood and, at a low frequency, in various clonogenic progenitors in many patients with AML1-ETO AML in long-term remission,55,,58  whereas a second mutation (eg, KIT) becomes undetectable despite similar detection sensitivities.67  The persistence of preleukemic stem cells is similarly supported by X chromosome inactivation studies showing persistently clonal hematopoiesis in some patients in morphologic remission after treatment for AML.11,68  Although rare in the absence of constitutional skewing of hematopoietic cells, clonal remissions have commonly been found in association with dysplastic changes, consistent with the notion of persistence of a preleukemic stem cells (clone).69,71 

Despite their limitations, observations to date suggest 3 possibly simplified but testable scenarios of AML development (Figure 2). In the first, both the initial and subsequent mutational events occur at the level of pluripotent CD33 precursors; for reasons of simplicity, we will refer to those as “immature” leukemias. In these, clonal dominance could develop in multiple cell lineages or be confined to granulocytes and monocytes if the capacity for differentiation along the other cell lineages is lost, or if unregulated growth is displayed only once single lineage commitment has occurred because of cell context-specific effects of the mutational event(s).11,14  Many of these leukemias may be identifiable in xenotransplantation assays by the ability of CD34+/CD38 cells to recapitulate the disease. In contrast, in “mature” leukemias, at least one relevant mutational event occurs at the level of committed myeloid cells. Specifically, in the second scenario, the initial mutation is acquired in pluripotent HSCs, whereas the collaborating mutational event(s) leading to full AML transformation and subsequent clonal expansion only occur at a later stage, possibly at the level of committed CD33+ myeloid precursors. As discussed in the previous section, at least some of the CBF leukemias may follow this pattern of transformation. Finally, in the third scenario, all mutational events as well as clonal expansion occur at the level of committed CD33+ myeloid precursors. An example may be APL, as small studies indicate expression of APL mainly in the granulocytic/monocytic lineage and predominant involvement of committed CD33+ myeloid progenitors.72 

Figure 2

Proposed models of AML transformation. Three proposed simplified scenarios of step-wise transformation in human AML, leading from a normal cell (green) to a premalignant cell (light red) and, eventually, to a malignant cell (dark red) with clonal expansion: Scenario 1, both the initial transforming event and subsequent mutations leading to clonal expansion occur at the level of multipotent, CD33 precursors; Scenario 2, the initial transforming event occurs at the level of multipotent, CD33 precursors, whereas the collaborating mutational event leading to clonal expansion occurs at the level of CD33+ committed myeloid progenitors; and Scenario 3, both initial and subsequent mutations occur at the level of CD33+ committed myeloid precursors. Similar models have been proposed by others.129 

Figure 2

Proposed models of AML transformation. Three proposed simplified scenarios of step-wise transformation in human AML, leading from a normal cell (green) to a premalignant cell (light red) and, eventually, to a malignant cell (dark red) with clonal expansion: Scenario 1, both the initial transforming event and subsequent mutations leading to clonal expansion occur at the level of multipotent, CD33 precursors; Scenario 2, the initial transforming event occurs at the level of multipotent, CD33 precursors, whereas the collaborating mutational event leading to clonal expansion occurs at the level of CD33+ committed myeloid progenitors; and Scenario 3, both initial and subsequent mutations occur at the level of CD33+ committed myeloid precursors. Similar models have been proposed by others.129 

Close modal

The nature of LSCs likely has significant prognostic implications. For example, the ability of leukemias to engraft in immunodeficient mice correlates with adverse prognosis in most series.24,26,,29  Consistently, an LSC-specific signature derived from engrafting leukemia specimens identifies AML patients with poor long-term survival.73  In turn, a distinct type of LSC may underlie those up to 50% of human AML that fail to engraft in immunodeficient mice and may be more responsive to current therapies, with a paradigm, again, being APL.

Our LSC models emphasize the importance of the differentiative and proliferative potential of the target cells in which the genetic events occur rather than the specific mutation per se; indeed, limited data indicate that similar mutations may lead to distinct outcomes depending on the proliferative potential of the cell in which they arose. For example, Pollard et al demonstrated in a set of FLT3/ITD AMLs that leukemias in which the FLT3 abnormality could be detected in CD34+/CD33 progenitors had a higher relapse risk and worse outcome than leukemias in which FLT3/ITD was only detectable in more mature CD34+/CD33+ cells.74  More recently, Bachas et al demonstrated, among patients initially presenting with various recognized mutations, that the continued presence of these mutations at relapse was associated with a shorter time to relapse, whereas loss of such mutations correlated with a longer time to relapse.65  These observations possibly indicate a worse prognosis if the AML-associated mutations arose in a chemotherapy-resistant, presumably less mature, cellular compartment. The notion that the nature of the stem/progenitor cells acquiring the mutation, rather than the particulars of the mutation itself, determines outcome could explain why prognosis can be similar for many leukemias with dissimilar mutations but different for many leukemias with similar mutations. An alternative explanation, of course, invokes the existence of important yet-to-be identified mutations.

“Mature” leukemias may be particularly suited for stem cell-directed therapies as underlying LSCs may be distinct from normal HSCs and intrinsically less resistant to therapy. LSC-targeted therapy could then (partially) replace other therapeutics and spare patients from some nonspecific toxicity in such lower-risk diseases. As a first attempt to eliminate LSCs in these leukemias, we focused on the myeloid differentiation antigen, CD33, which is expressed on leukemic blasts from 85% to 90% of AML patients.32,75  The utility of CD33-targeted therapeutics was suggested by the observation that ablation of CD33+ cells could restore normal hematopoiesis in vitro in some leukemias with clonal dominance limited to granulocytes/monocytes.8  However, unconjugated anti–CD33 antibodies were largely ineffective in patients with overt CD33+ non-APL AML.76,77  On the other hand, radiolabeled anti–CD33 antibodies were selectively taken up by AML cells and rapidly saturated leukemic blast cells in peripheral blood and bone marrow at intravenous doses of more than or equal to 5 mg/m2,76,78,79  suggesting that addition of a toxic payload might produce better results. In collaboration with industry, we selected N-acetyl γ-calicheamicin dimethyl hydrazide, a stable derivative of a natural enediyne anti–tumor antibiotic initially identified in a screen for potent DNA-damaging agents,80,81  for this purpose. Subsequently, the antibody was humanized to minimize immune responses and conjugated with the calichemicin derivative via an acid-labile linker that provided good potency and selectivity because of its stability in the circulation but rapid toxin release under acidic conditions, such as those in lysosomes (Figure 3).80,82  Preclinically, GO showed selective cytotoxicity against CD33+ AML cells, effectively inhibited CFCs in human AML specimens, and caused regression of CD33+ AML cell line xenografts in athymic mice.82 

Figure 3

Schematic structure of GO. The humanized IgG4 anti–CD33 antibody (hP67.6) contains amino acid sequences that are approximately 98.3% of human origin. Lysine residues on hP67.6 are linked to N-acetyl γ-calicheamicin dimethyl hydrazide via a hybrid 4-(4′-acetylphenoxy)butanoic acid linker. The labile hydrazone bond leading to drug release under acidic conditions, presumably within lysosomes, is shaded in green. GO has approximately 50% of the antibody loaded with 4 to 6 mol of the toxic moiety per mole of antibody; the remaining 50% of antibody is unbound. Reproduced with permission from Macmillan Publishers Ltd.130 

Figure 3

Schematic structure of GO. The humanized IgG4 anti–CD33 antibody (hP67.6) contains amino acid sequences that are approximately 98.3% of human origin. Lysine residues on hP67.6 are linked to N-acetyl γ-calicheamicin dimethyl hydrazide via a hybrid 4-(4′-acetylphenoxy)butanoic acid linker. The labile hydrazone bond leading to drug release under acidic conditions, presumably within lysosomes, is shaded in green. GO has approximately 50% of the antibody loaded with 4 to 6 mol of the toxic moiety per mole of antibody; the remaining 50% of antibody is unbound. Reproduced with permission from Macmillan Publishers Ltd.130 

Close modal

In GO, the unconjugated antibody itself is largely nontoxic83  and primarily facilitates uptake of the calicheamicin derivative into CD33+ cells (Table 1). Once released intracellularly, the enediyne moiety undergoes rearrangement to form a 1,4-benzenoid diradical that initiates single- and double-stranded DNA breaks.81,84  This damage elicits a strong cellular response with cell-cycle arrest and subsequent DNA repair or, if damage is overwhelming, apoptosis and cell death, predominantly via mitochondrial membrane permeabilization and caspase activation.84  The central role of DNA damage in GO-induced cyotoxicity is supported by the observation that cell lines defective in DNA repair are highly sensitive to calicheamicins.85,86 

Table 1

Cellular parameters that affect GO efficacy

FactorComment
Uptake of CD33/GO complexes  
    Receptor-mediated uptake  
        CD33 expression levels Good evidence supporting quantitative relationship from experimental and correlative studies83,90,91,125 ; expression levels associated with cytogenetic risk of AML126  and CD33 SNPs127  
        CD33 saturation In vitro evidence linking reduced CD33 saturation to reduced GO cytotoxicity128  
        CD33 internalization Relatively slow process, controlled by intracellular tyrosine motifs91  and likely tyrosine phosphorylation status of CD33 
        Reexpression of CD33 binding sites Surface CD33 levels return to pretreatment levels within 72 hours after CD33 antibody administration104 ; could contribute to amount of internalized GO, in particular if GO is administered in fractionated doses. 
    Non–receptor-mediated uptake Suggested by experimental studies93 ; clinical role unknown 
Intracellular trafficking of GO Hypothetical 
Activation of GO Low pH in lysosomes required (R.B.W., unpublished data, December 2004) 
Extrusion of GO  
    ABC family of drug transporters Good evidence from experimental and correlative studies for role of P-glycoprotein and multidrug resistance-associated protein 184,89,90 ; role of other transporters unknown 
Induction of cytotoxicity  
    Generation of SS- and DS-DNA breaks Hypersensitivity of cell lines with defects in DNA repair to calicheamicins85,86  
    Mitochondrial pathways of apoptosis Good experimental evidence for role of pro- and antiapoptotic Bcl-2 protein family members84  
    Other downstream pro- or antiapoptotic signaling pathways Not examined in detail 
    Cell-cycle status Limited in vitro data suggesting that resting cells are relatively less susceptible to GO93  
FactorComment
Uptake of CD33/GO complexes  
    Receptor-mediated uptake  
        CD33 expression levels Good evidence supporting quantitative relationship from experimental and correlative studies83,90,91,125 ; expression levels associated with cytogenetic risk of AML126  and CD33 SNPs127  
        CD33 saturation In vitro evidence linking reduced CD33 saturation to reduced GO cytotoxicity128  
        CD33 internalization Relatively slow process, controlled by intracellular tyrosine motifs91  and likely tyrosine phosphorylation status of CD33 
        Reexpression of CD33 binding sites Surface CD33 levels return to pretreatment levels within 72 hours after CD33 antibody administration104 ; could contribute to amount of internalized GO, in particular if GO is administered in fractionated doses. 
    Non–receptor-mediated uptake Suggested by experimental studies93 ; clinical role unknown 
Intracellular trafficking of GO Hypothetical 
Activation of GO Low pH in lysosomes required (R.B.W., unpublished data, December 2004) 
Extrusion of GO  
    ABC family of drug transporters Good evidence from experimental and correlative studies for role of P-glycoprotein and multidrug resistance-associated protein 184,89,90 ; role of other transporters unknown 
Induction of cytotoxicity  
    Generation of SS- and DS-DNA breaks Hypersensitivity of cell lines with defects in DNA repair to calicheamicins85,86  
    Mitochondrial pathways of apoptosis Good experimental evidence for role of pro- and antiapoptotic Bcl-2 protein family members84  
    Other downstream pro- or antiapoptotic signaling pathways Not examined in detail 
    Cell-cycle status Limited in vitro data suggesting that resting cells are relatively less susceptible to GO93  

SNP indicates single-nucleotide polymorphism; SS, single-stranded; and DS, double-stranded.

This mechanism of action implies a critical role for the intracellular accumulation of the calicheamicin-γ1 derivative as well as the cellular response to the toxin's DNA-damaging effect (Table 1). Conceptually, the amount of intracellular, active calicheamicin is affected by cellular uptake, toxin release and activation, as well as drug inactivation/metabolism or extrusion. On the other hand, the toxicity of the calicheamicin moiety will be modulated by the ability of the cell to repair DNA damage and the activity of downstream pro- and antiapoptotic pathways. Large interindividual differences exist in this respect, as indicated by the observation that the sensitivity to the toxic moiety varies more than 100 000-fold between individual human AML cell specimens.87 

Consistent with this mechanism, drug efflux mediated by members of the adenosine triphosphate (ATP) binding cassette (ABC) superfamily of proteins, predominantly P-glycoprotein (ABCB1) and, to a lesser degree, multidrug resistance-associated protein 1 (ABCC1), mediate resistance to GO in vitro and in vivo.84,88,90  Furthermore, experimental studies revealed a striking, quantitative association between CD33 expression/uptake and GO efficacy in engineered human AML cell lines.91,92  In contrast, the clinical impact of DNA repair and downstream pathways has not been examined in detail, but GO efficacy is modulated in vitro by members of the Bcl-2 family of proteins.84  Similarly unknown is optimal timing of the use of GO when combined with other agents. It is conceivable that timing and cell-cycle status matter, however, as in vitro data suggest that resting cells are relatively more resistant to GO.93 

A phase 1 study was conducted in 40 adults with relapsed/refractory CD33+ AML who received up to 3 doses of GO at 2-week intervals.94  Elimination of morphologically detectable leukemia occurred in 8 (20%) of the patients, with 3 (7.5%) achieving a complete remission (CR) as conventionally defined and 2 (5%) achieving a CR with incomplete platelet recovery (CRp). As dose-limiting nonhematologic toxicity was not reached, the highest dose level of 9 mg/m2, which provided almost complete saturation of peripheral blood and marrow CD33 binding sites, was chosen for further study.94  Three open-label, multicenter single-arm phase 2 trials subsequently evaluated GO in a larger cohort of adults with CD33+ de novo AML in first relapse. An interim analysis included 142 patients with a median age of 61 years (range, 22-84 years) who typically received 2 doses of GO 14 days apart; 23 patients (16.2%) achieved a CR, and 19 (13.4%) achieved a CRp, for an overall response rate of 29.6%.95  Based on these results, GO was given accelerated marketing approval in the United States in May 2000 for the treatment of patients older than 60 years with CD33+ AML in first relapse who were not candidates for standard cytotoxic chemotherapy.96  The final report on 277 patients confirmed the early results with demonstration of an overall response rate of 25.6%, although remission durations were relatively short.97 

Several additional phase 2 studies investigated GO monotherapy for treatment of newly diagnosed and/or relapsed/refractory disease in unselected patients with non-APL AML. Together, these studies confirmed single-agent activity of GO in a subset of patients; however, overall response rates have generally not exceeded 25% to 35% and have occasionally been quite disappointing, particularly in heavily pretreated patients.98,,,,103  In these studies, GO has usually been given at 2-week intervals. However, early studies indicated that new CD33 binding sites were continuously reexpressed, and surface CD33 levels returned to pretreatment levels within 72 hours after anti–CD33 antibody administration despite antigen internalization and modulation.104,105  This observation suggests that repeated administration of lower, (near-)saturating doses of GO every 3 days may enhance intracellular accumulation of the calicheamicin-γ1 derivative beyond what can be achieved with 2-weekly administration. The Acute Leukemia French Association group has relatively recently reported the use of GO in fractionated, lower doses with promising efficacy and acceptable safety profile,89,106  but comparative data with the traditional administration scheme are currently not available.

Consistent with preclinical predictions that GO would be most effective for leukemias likely originating in a committed progenitor as suggested for APL, the drug appears to be highly active against this entity.107,109  This was emphasized by the observation that GO monotherapy routinely resulted in molecular remissions that could be durable in patients with molecularly relapsed APL.107,108  In newly diagnosed APL, GO adds to the effectiveness of all-trans retinoic acid and can replace anthracyclines in the curative backbone of treatment.110,111 

In addition to those in APL, many studies have explored GO together with other therapeutics in non-APL AML.98,,,,103  Emerging data from large, phase 3 randomized studies now indicate that substantial numbers of patients with newly diagnosed AML experience improved survival when GO is combined with conventional chemotherapy (Table 2). Particularly important are findings from the Medical Research Council AML15 trial.112  In this trial, 1113 predominantly adult patients were randomized to receive a single dose of GO (3 mg/m2) on day 1 of the first of 2 induction courses with one of 3 induction regimens (cytarabine/daunorubicin/etoposide, daunorubicin/cytarabine, fludarabine/cytarabine/G-CSF/idarubicin). Patients in remission who were not scheduled for hematopoietic cell transplantation (HCT) were then randomized to consolidation with or without the addition of GO. Although adding GO to either induction or consolidation did not affect overall remission rates or survival, protocol-predefined subgroup analyses showed a highly significant interaction between cytogenetics and use of GO in induction therapy. Specifically, with the addition of GO, there was improved overall survival (OS) at 5 years for patients with favorable cytogenetics (79% vs 51%; P = .0003) but no benefit for patients with unfavorable cytogenetics (8% vs 11%; P = .4). There was also an OS benefit for some patients with intermediate-risk disease, as indicated by an internally validated index using cytogenetics, age, and performance status, which predicted that approximately 70% of all intermediate-risk patients would have a 10% improvement in 5-year OS if given GO during induction; within the intermediate-risk subgroup, those 30% predicted to not benefit were older, had higher white blood cell counts, worse performance status, or secondary disease.112 

Table 2

Phase 3 studies of GO in newly diagnosed non-APL AML

StudyDiseaseNAge, y (median)TreatmentResults
MRC AML15112  AML 1113 0-71 (49) ± GO (3 mg/m2) on day 1 of the first of 2 induction courses with either ADE, DA, or FLAG-IDA No difference in ORR, TRM, relapse, or survival. Improved 5-y OS for favorable-risk subgroup (79% vs 51%; P = .0003) with GO; predicted 10% OS benefit for ∼ 70% of patients with intermediate-risk disease 
ALFA 0701113  AML 278 59-66 (62) ± GO (3 mg/m2) on days 1, 4, and 7 of DA induction and day 1 of each of 2 courses of DA consolidation No difference in ORR or mortality. Improved 2-y EFS (40.8% vs 17.1%, P = .0003), DFS (50.3% vs 22.7%, P = .0003) and OS (53.2% vs 41.9%, P = .037) with GO. Survival benefit seen in favorable/intermediate- but not adverse-risk disease 
GOELAMS AML 2006 IR114  AML (intermediate risk) 238 18-60 (50) ± GO (6 mg/m2) on day 1 with DA induction and MA consolidation No difference in ORR, TRM, or 3-y EFS. Improved EFS with GO in patients who did not undergo allogeneic HCT (53.7% vs 27%, P = .0308) 
NCRI AML16115  AML, high-risk MDS 1115 51-84 (67) ± GO (3 mg/m2) on day 1 of the first of 2 induction courses with either DA or DCLo No difference in TRM; trend towards reduced risk of persistent disease with GO (17% vs 21%, P = .06). Reduced 3-y relapse risk (68% vs 76%; P = .007) and superior DFS (21% vs 16%; P = .04) and OS (25% vs 20%; P = .05) with GO 
SWOG S0106116,117  AML 596 18-60 (47) ± GO (6 mg/m2) on day 4 of the first of up to 2 induction courses with DA* Increased TRM in GO arm (5.7% vs 1.4%; P = .01). No difference in ORR, DFS, or OS. Possible trend towards improved OS in favorable-risk subgroup with GO (hazard ratio: 0.49 [0.12–2.04]) 
StudyDiseaseNAge, y (median)TreatmentResults
MRC AML15112  AML 1113 0-71 (49) ± GO (3 mg/m2) on day 1 of the first of 2 induction courses with either ADE, DA, or FLAG-IDA No difference in ORR, TRM, relapse, or survival. Improved 5-y OS for favorable-risk subgroup (79% vs 51%; P = .0003) with GO; predicted 10% OS benefit for ∼ 70% of patients with intermediate-risk disease 
ALFA 0701113  AML 278 59-66 (62) ± GO (3 mg/m2) on days 1, 4, and 7 of DA induction and day 1 of each of 2 courses of DA consolidation No difference in ORR or mortality. Improved 2-y EFS (40.8% vs 17.1%, P = .0003), DFS (50.3% vs 22.7%, P = .0003) and OS (53.2% vs 41.9%, P = .037) with GO. Survival benefit seen in favorable/intermediate- but not adverse-risk disease 
GOELAMS AML 2006 IR114  AML (intermediate risk) 238 18-60 (50) ± GO (6 mg/m2) on day 1 with DA induction and MA consolidation No difference in ORR, TRM, or 3-y EFS. Improved EFS with GO in patients who did not undergo allogeneic HCT (53.7% vs 27%, P = .0308) 
NCRI AML16115  AML, high-risk MDS 1115 51-84 (67) ± GO (3 mg/m2) on day 1 of the first of 2 induction courses with either DA or DCLo No difference in TRM; trend towards reduced risk of persistent disease with GO (17% vs 21%, P = .06). Reduced 3-y relapse risk (68% vs 76%; P = .007) and superior DFS (21% vs 16%; P = .04) and OS (25% vs 20%; P = .05) with GO 
SWOG S0106116,117  AML 596 18-60 (47) ± GO (6 mg/m2) on day 4 of the first of up to 2 induction courses with DA* Increased TRM in GO arm (5.7% vs 1.4%; P = .01). No difference in ORR, DFS, or OS. Possible trend towards improved OS in favorable-risk subgroup with GO (hazard ratio: 0.49 [0.12–2.04]) 

ADE indicates cytarabine/daunorubicin/etoposide; DA, daunorubicin/cytarabine; DClo, daunorubicin/clofarabine; DFS, disease-free survival; EFS, event-free survival; FLAG-Ida, fludarabine/cytarabine/G-CSF/idarubicin; HCT, hematopoietic cell transplantation; MA, mitoxantrone/cytarabine; ORR, overall response rate (CR + CRi); OS, overall survival; and TRM, treatment-related mortality.

*

Daunorubicin was used at 60 mg/m2/d in the control arm and 45 mg/m2/d in the GO arm.

Results from 3 additional European studies support the conclusions from AML15 and extend those findings to older adults. In the Acute Leukemia French Association 0701 trial, 278 evaluable patients with primary non-APL AML 50 to 70 years of age received daunorubicin/cytarabine with or without GO (3 mg/m2) on days 1, 4, and 7; a second course of daunorubicin/cytarabine was given for residual disease on day 15. Patients in remission then received 2 courses of daunorubicin/cytarabine with or without GO (3 mg/m2) on day 1 of each cycle.113  Although there was no statistically significant difference in remission rates (CR + CRp: 81% vs 75%, P = .25) and treatment-related mortality (6% vs 4%, P = .41) between the +GO and −GO arms, the event-free survival at 2 years was significantly superior in the GO arm (40.8% vs 17.1%, P = .0003), as was disease-free survival (50.3% vs 22.7%, P = .0003) and OS (53.2% vs 41.9%, P = .037). Subgroup analysis showed that the event-free survival benefit was associated with favorable/intermediate but not adverse karyotype.113  In the GOELAMS AML 2006 IR study, adults 18 to 60 years of age with de novo AML and intermediate karyotype received daunorubicin/cytarabine with or without GO (6 mg/m2) on day 4; GO was also added to consolidation therapy with mitoxantrone/cytarabine according to the initial randomization.114  Among 238 analyzed patients, there was no difference in remission rates or treatment-related mortality between the 2 treatment arms. Overall, there was also no statistically significant difference in event-free survival (GO vs control: 51% vs 33%) or OS (53% vs 46%) at 3 years. However, subgroup analyses showed that, in patients who did not undergo allogeneic HCT, event-free survival was significantly higher in the GO group (53.7% vs 27%, P = .0308). Finally, in the NCRI AML16 trial, 1115 older adults (51-84 years of age) with AML or high-risk MDS (> 10% marrow blasts) received either daunorubicin/cytarabine or daunorubicin/clofarabine for 2 cycles with or without GO (3 mg/m2) on day 1 of the first induction course; patients in CR were then randomized to receive, or not, a third course of daunorubicin/cytarabine with or without maintenance therapy with azacitidine.115  Similar to the other trials, there was no statistically significant difference in remission rates and treatment-related mortality between the +GO and −GO arms, although the patients in the −GO arm tended to have a higher likelihood of persistent disease (21% vs 17%, P = .06). However, patients receiving GO had a reduced relapse risk (at 3 years: 68% vs 76%; P = .007), resulting in a superior disease-free survival (21% vs 16%; P = .04) and OS (25% vs 20%; P = .05); the benefit of GO appeared greater in patients with de novo disease than those with secondary disease.115  Finally, a meta-analysis of AML15 and AML16 on 2224 patients showed a significant benefit of GO for risk of relapse (odds ratio [OR] = 0.82; 95% CI, 0.72-0.93, P = .002) and survival (OR = 0.88; 95% CI, 0.79-0.98, P = .02); the survival benefit was seen in patients with favorable-risk (OR = 0.47; 95% CI, 0.28-0.77) and intermediate-risk (OR = 0.84; 95% CI, 0.73-0.97) but not adverse-risk (OR = 1.02; 95% CI, 0.81-1.27) disease.115 

The results from these European studies differ from a trial conducted by the Southwest Oncology Group (trial S0106),116  which was developed with the drug sponsor to fulfill their postapproval commitment to the FDA. A total of 637 patients 18 to 60 years of age with de novo AML were accrued to receive up to 2 cycles of induction chemotherapy with daunorubicin/cytarabine with or without a single dose of GO (6 mg/m2) on day 4 of the first induction.116,117  Unlike the European studies in which identical doses of conventional chemotherapeutics were used in both arms, S0106 used a lower daunorubicin dose with GO (45 mg/m2 vs 60 mg/m2) with the intent of evaluating 2 similarly toxic treatment arms.116  After consolidation therapy, patients in remission were eligible for a second randomization (stratified by cytogenetic risk category at diagnosis and use of GO during induction) between GO or observation only. Among 596 evaluable patients, S0106 showed no overall difference in response or survival in induction with the addition of GO, but a trend toward improved OS was seen for patients with CBF leukemias (hazard ratio = 0.49; 0.12-2.04); there was also no benefit for GO in consolidation.116,117  It should be noted that S0106 was not powered to detect important outcome differences in the favorable-risk patients. Given the attempt to achieve equitoxicity with a 25% lower dose of daunorubicin in the GO arm,116  the S0106 trial may thus be confounded toward the null as the dose reduction of daunorubicin might mask a greater benefit of GO. Based on the lack of prespecified overall improvement in outcome, S0106 was prematurely terminated before planned accrual was reached.117  After discussions with the FDA, the failure to detect improved survival together with a trend toward increased treatment-related mortality in S0106 led Pfizer to discontinue commercial availability of GO in the United States in October 2010.118 

Of note, the early studies on the clonal origin of AML indicated that diseases limited primarily to the granulocytic/monocytic lineage occurred predominantly in younger adults and children,11  suggesting that GO-based therapies might be particularly effective for some pediatric patients. So far, only a few clinical studies have explored the use of GO in pediatric AML, primarily in patients with relapsed/refractory disease, and the drug has no established role in childhood AML.102,103  A large trial testing the addition of GO to induction chemotherapy in a randomized fashion was led by the Children's Oncology Group (AAML0531) and has recently completed accrual of more than 1000 participants, with preliminary outcome results expected for 2012 or 2013.

The trials conducted over the last decade demonstrate that GO improves survival for many, but not all, AML patients, supporting the preclinical conclusion that CD33 is a valid target for some disease subforms. The long-term benefit achieved with GO in APL and CBF leukemias and some patients with less favorable prognoses, in particular those with intermediate-risk diseases, is consistent with an ablative effect on disease-relevant CD33+ cells (ie, AML stem or progenitor cells). As discussed previously, we hypothesized that CD33-targeting therapeutics would be most beneficial for patients with “mature” LSCs. The most compelling data to support the notion that CD33-directed therapy might indeed effectively target malignant stem cells in some “mature” leukemias come from the treatment of APL.107,108  Although the high and homogeneous expression of CD33 and low activity of P-glycoprotein may partially explain the exquisite sensitivity of APL toward GO,92,119,120  even monotherapy with an unconjugated humanized anti–CD33 antibody (HuM195) leads to a molecular remissions in approximately half of the APL patients with molecular minimal residual disease after induction chemotherapy.121 

The situation is likely different for CBF leukemias and other AML subforms, in which founder mutations may arise at the level of multipotent CD33 stem cells in at least some of the cases. Although non–receptor-mediated drug uptake of GO has been suggested by small in vitro studies with CD33 acute lymphoblastic leukemia cell lines,93  its contribution to clinical GO efficacy is unknown, and CD33 LSCs are unlikely effective targets for GO. Nevertheless, the clinical benefit of CD33-targeted therapy in these leukemias could be understood if the preleukemic CD33 cells were relatively nonprolific and additional mutations leading to frank leukemic transformation were acquired only at a later stage when the cell expressed CD33 (scenario 2 of the proposed AML transformation models). In this scenario, successful depletion of CD33+ cells would eradicate the fully transformed, proliferative clone and leave the CD33 pre-LSCs behind (ie, reset the clock to a premalignant stage). For a cure, such preleukemic cells may need to be controlled or eliminated by other means (eg, immunologic mechanisms), be it via the patient's own immunosurveillance or via graft-versus-leukemia effects after allogeneic HCT. This may be most successful if the preleukemic cells were undetectable or present at very minimal levels at the time of HCT.

It is important to emphasize that studies to date have not determined whether GO, besides acting on more mature CD33+ progeny, can indeed directly kill CD33+ LSCs in vivo, and whether long-term benefit from GO is related to successful targeting of LSCs. Instead, it is imaginable that the observed clinical long-term efficacy in some leukemias could result from effects on AML cells without stem cell properties. This possibility needs to be considered because “intensification” of the chemotherapeutic regimens via addition of GO (eg, as done in the recent European phase 3 trials) could lead to more robust non-LSC AML cell debulking. The long-term benefit in this situation is conceivable even if successful depletion of CD33+ cells only eliminates the most proliferative cells and leaves fully transformed LSCs behind; the latter might then be eradicated by other chemotherapeutic, radiotherapeutic, or immunologic means, especially if at minimal levels and not overly prolific. This scenario could explain a benefit of GO in leukemias that harbor CD33 LSCs (scheme 1 of the proposed AML transformation models). As an alternative for those leukemias, CD33 LSCs remaining after chemotherapy-induced bulk reduction might enter cell cycle and acquire CD33 and thus be susceptible to CD33 targeting. Finally, the (short-lasting) remissions seen in unselected patients with relapsed/refractory AML could result from debulking of more mature CD33+ AML blasts by GO, even if underlying LSCs remained CD33 and could not be eradicated. There is thus a critical need for further studies to determine the relationship between the origin of LSCs and efficacy of CD33-directed therapy, and formally test our hypothesis that benefit from this therapeutic strategy is mostly confined to patients with “mature” leukemias. These investigations will need to use functional characterizations of cells (eg, via long-term in vitro culture) and carefully consider the possibility of aberrant cell surface marker expression that may limit the conclusions that can be drawn with regard to cell of origin from immunophenotypic studies.

Response to GO parallels sensitivity to conventional chemotherapy, as patients deriving most long-term benefit from GO also have a better response to standard chemotherapeutics. Thus, GO may be used with different goals in mind: first, as best exemplified by the European phase 3 trials, GO could be added to conventional chemotherapy to improve the outcome of standard therapy alone with relatively little increase in toxicity. On the other hand, as suggested by the experience in APL, GO could at least partially replace nontargeted therapeutics in low-risk patients with preservation of treatment efficacy but possibly reduced nonspecific toxicity.

It is plausible that not all CD33+ AML cells, and in particular CD33+ LSCs, may be immediately susceptible to GO. CD33 is challenging to target with immunotoxins because of antigen expression at relatively low abundance (∼ 104 CD33 molecules/cell)122,123  and slow conjugate internalization.91  Furthermore, GO's efficacy is significantly reduced by drug efflux, a concern for any stem cell-directed therapy given increasing evidence linking ABC drug transporters to protection of cancer stem cells.124  Rational strategies to improve immunotherapy with GO would therefore include maneuvers that increase intracellular toxin accumulation, for example by manipulating CD33 expression/turnover or inhibiting drug efflux.92  In addition, approaches that interfere with DNA repair and/or downstream pro-/antiapoptotic signaling may lead to a lowering of the apoptotic threshold and increased cellular toxicity of calicheamicins. An alternative entails the development of novel immunoconjugates using more potent toxins that are not (or less) susceptible to ABC transporter activity. Substitution of the calicheamicin-γ1 derivative with a different toxin may have the potential to reduce the risk of significant liver toxicity, specifically veno-occlusive disease, a risk that appears to have decreased but not entirely disappeared with the lower doses of GO used in the recent phase 3 trials.

The experience with GO is a reminder of the intrinsic heterogeneity in AML. Conceivably reflecting differences in the cellular origin of individual leukemias, this diversity is of particular significance for therapies that aim at eradicating stem cells in AML and implies that no such approach may be universally effective for all leukemias. Nevertheless, the clinical results with GO strongly support the utility of CD33-targeting therapeutics in AML and suggest that the current unavailability of GO is unfortunate. With the emerging data on the benefit of GO for some subsets of AML, reconsideration of the drug's value by the manufacturer and regulatory authorities is warranted. As we improve our abilities to identify those AMLs likely to respond to specific treatments, clinical trials testing these treatments may restrict eligibility to patients with these leukemias. In the future, we may thus have to place greater emphasis on collaborative, perhaps multinational studies, or more likely, accept higher false-positive and false-negative rates or detection of only relatively large differences between treatment and controls.

This work was supported by the National Cancer Institute/National Institutes of Health (grants P30-CA015704-35S6 and U10-CA098543), the Leukemia & Lymphoma Society (Specialized Center of Research grant 7008-08), and the St Baldrick's Foundation.

National Institutes of Health

Contribution: R.B.W. and I.D.B. conceived and wrote the manuscript; and F.R.A. and E.H.E. assisted with writing the manuscript.

Conflict-of-interest disclosure: R.B.W. received research funding from Seattle Genetics Inc. R.B.W. and I.D.B. were consultants for Seattle Genetics Inc. The remaining authors declare no competing financial interests.

Correspondence: Roland B. Walter, Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D2-190, Seattle, WA 98109-1024; e-mail: [email protected].

1
Reya
T
Morrison
SJ
Clarke
MF
Weissman
IL
Stem cells, cancer, and cancer stem cells.
Nature
2001
414
6859
105
111
2
Dick
JE
Stem cell concepts renew cancer research.
Blood
2008
112
13
4793
4807
3
Clarke
MF
Dick
JE
Dirks
PB
et al
Cancer stem cells. Perspectives on current status and future directions: AACR Workshop on cancer stem cells.
Cancer Res
2006
66
19
9339
9344
4
Stubbs
MC
Armstrong
SA
Therapeutic implications of leukemia stem cell development.
Clin Cancer Res
2007
13
12
3439
3442
5
Lane
SW
Gilliland
DG
Leukemia stem cells.
Semin Cancer Biol
2010
20
2
71
76
6
Majeti
R
Monoclonal antibody therapy directed against human acute myeloid leukemia stem cells.
Oncogene
2011
30
9
1009
1019
7
Passegué
E
Jamieson
CH
Ailles
LE
Weissman
IL
Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics?
Proc Natl Acad Sci U S A
2003
100
Suppl 1
11842
11849
8
Bernstein
ID
Monoclonal antibodies to the myeloid stem cells: therapeutic implications of CMA-676, a humanized anti–CD33 antibody calicheamicin conjugate.
Leukemia
2000
14
3
474
475
9
Chen
GL
Prchal
JT
X-linked clonality testing: interpretation and limitations.
Blood
2007
110
5
1411
1419
10
Fialkow
PJ
Singer
JW
Adamson
JW
et al
Acute nonlymphocytic leukemia: heterogeneity of stem cell origin.
Blood
1981
57
6
1068
1073
11
Fialkow
PJ
Singer
JW
Raskind
WH
et al
Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia.
N Engl J Med
1987
317
8
468
473
12
Bernstein
ID
Singer
JW
Andrews
RG
et al
Treatment of acute myeloid leukemia cells in vitro with a monoclonal antibody recognizing a myeloid differentiation antigen allows normal progenitor cells to be expressed.
J Clin Invest
1987
79
4
1153
1159
13
Bernstein
ID
Singer
JW
Smith
FO
et al
Differences in the frequency of normal and clonal precursors of colony-forming cells in chronic myelogenous leukemia and acute myelogenous leukemia.
Blood
1992
79
7
1811
1816
14
McCulloch
EA
Stem cells in normal and leukemic hemopoiesis (Henry Stratton Lecture, 1982).
Blood
1983
62
1
1
13
15
Lapidot
T
Sirard
C
Vormoor
J
et al
A cell initiating human acute myeloid leukaemia after transplantation into SCID mice.
Nature
1994
367
6464
645
648
16
Bonnet
D
Dick
JE
Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.
Nat Med
1997
3
7
730
737
17
Blair
A
Hogge
DE
Ailles
LE
Lansdorp
PM
Sutherland
HJ
Lack of expression of Thy-1 (CD90) on acute myeloid leukemia cells with long-term proliferative ability in vitro and in vivo.
Blood
1997
89
9
3104
3112
18
Miyamoto
T
Weissman
IL
Akashi
K
AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation.
Proc Natl Acad Sci U S A
2000
97
13
7521
7526
19
Chao
MP
Seita
J
Weissman
IL
Establishment of a normal hematopoietic and leukemia stem cell hierarchy.
Cold Spring Harb Symp Quant Biol
2008
73
439
449
20
Taussig
DC
Miraki-Moud
F
Anjos-Afonso
F
et al
Anti–CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells.
Blood
2008
112
3
568
575
21
Taussig
DC
Vargaftig
J
Miraki-Moud
F
et al
Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34− fraction.
Blood
2010
115
10
1976
1984
22
Goardon
N
Marchi
E
Atzberger
A
et al
Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia.
Cancer Cell
2011
19
1
138
152
23
Ailles
LE
Gerhard
B
Kawagoe
H
Hogge
DE
Growth characteristics of acute myelogenous leukemia progenitors that initiate malignant hematopoiesis in nonobese diabetic/severe combined immunodeficient mice.
Blood
1999
94
5
1761
1772
24
Rombouts
WJC
Martens
ACM
Ploemacher
RE
Identification of variables determining the engraftment potential of human acute myeloid leukemia in the immunodeficient NOD/SCID human chimera model.
Leukemia
2000
14
5
889
897
25
Rombouts
WJC
Blokland
I
Löwenberg
B
Ploemacher
RE
Biological characteristics and prognosis of adult acute myeloid leukemia with internal tandem duplications in the Flt3 gene.
Leukemia
2000
14
4
675
683
26
Lumkul
R
Gorin
NC
Malehorn
MT
et al
Human AML cells in NOD/SCID mice: engraftment potential and gene expression.
Leukemia
2002
16
9
1818
1826
27
Monaco
G
Konopleva
M
Munsell
M
et al
Engraftment of acute myeloid leukemia in NOD/SCID mice is independent of CXCR4 and predicts poor patient survival.
Stem Cells
2004
22
2
188
201
28
Pearce
DJ
Taussig
D
Zibara
K
et al
AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML.
Blood
2006
107
3
1166
1173
29
Sanchez
PV
Perry
RL
Sarry
JE
et al
A robust xenotransplantation model for acute myeloid leukemia.
Leukemia
2009
23
11
2109
2117
30
Taussig
DC
Pearce
DJ
Simpson
C
et al
Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia.
Blood
2005
106
13
4086
4092
31
Andrews
RG
Torok-Storb
B
Bernstein
ID
Myeloid-associated differentiation antigens on stem cells and their progeny identified by monoclonal antibodies.
Blood
1983
62
1
124
132
32
Griffin
JD
Linch
D
Sabbath
K
Larcom
P
Schlossman
SF
A monoclonal antibody reactive with normal and leukemic human myeloid progenitor cells.
Leuk Res
1984
8
4
521
534
33
Andrews
RG
Takahashi
M
Segal
GM
Powell
JS
Bernstein
ID
Singer
JW
The L4F3 antigen is expressed by unipotent and multipotent colony-forming cells but not by their precursors.
Blood
1986
68
5
1030
1035
34
Andrews
RG
Singer
JW
Bernstein
ID
Precursors of colony-forming cells in humans can be distinguished from colony-forming cells by expression of the CD33 and CD34 antigens and light scatter properties.
J Exp Med
1989
169
5
1721
1731
35
Robertson
MJ
Soiffer
RJ
Freedman
AS
et al
Human bone marrow depleted of CD33-positive cells mediates delayed but durable reconstitution of hematopoiesis: clinical trial of MY9 monoclonal antibody-purged autografts for the treatment of acute myeloid leukemia.
Blood
1992
79
9
2229
2236
36
Bernstein
ID
Andrews
RG
Berenson
R
Bensinger
W
Singer
JW
Buckner
CD
Champlin
RE
Gale
RP
Isolation of human hematopoietic stem cells.
New Strategies in Bone Marrow Transplantation
1991
New York, NY
Wiley-Liss
201
207
37
Cozzio
A
Passegué
E
Ayton
PM
Karsunky
H
Cleary
ML
Weissman
IL
Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors.
Genes Dev
2003
17
24
3029
3035
38
Huntly
BJ
Shigematsu
H
Deguchi
K
et al
MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors.
Cancer Cell
2004
6
6
587
596
39
Krivtsov
AV
Twomey
D
Feng
Z
et al
Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9.
Nature
2006
442
7104
818
822
40
Somervaille
TC
Cleary
ML
Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia.
Cancer Cell
2006
10
4
257
268
41
So
CW
Karsunky
H
Passegue
E
Cozzio
A
Weissman
IL
Cleary
ML
MLL-GAS7 transforms multipotent hematopoietic progenitors and induces mixed lineage leukemias in mice.
Cancer Cell
2003
3
2
161
171
42
Chen
W
Kumar
AR
Hudson
WA
et al
Malignant transformation initiated by Mll-AF9: gene dosage and critical target cells.
Cancer Cell
2008
13
5
432
440
43
Kirstetter
P
Schuster
MB
Bereshchenko
O
et al
Modeling of C/EBPalpha mutant acute myeloid leukemia reveals a common expression signature of committed myeloid leukemia-initiating cells.
Cancer Cell
2008
13
4
299
310
44
Guibal
FC
Alberich-Jorda
M
Hirai
H
et al
Identification of a myeloid committed progenitor as the cancer-initiating cell in acute promyelocytic leukemia.
Blood
2009
114
27
5415
5425
45
Kelly
LM
Gilliland
DG
Genetics of myeloid leukemias.
Annu Rev Genomics Hum Genet
2002
3
179
198
46
Fröhling
S
Scholl
C
Gilliland
DG
Levine
RL
Genetics of myeloid malignancies: pathogenetic and clinical implications.
J Clin Oncol
2005
23
26
6285
6295
47
Vassiliou
GS
Cooper
JL
Rad
R
et al
Mutant nucleophosmin and cooperating pathways drive leukemia initiation and progression in mice.
Nat Genet
2011
43
5
470
475
48
Corral
J
Lavenir
I
Impey
H
et al
An Mll-AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice: a method to create fusion oncogenes.
Cell
1996
85
6
853
861
49
Chen
W
Li
Q
Hudson
WA
Kumar
A
Kirchhof
N
Kersey
JH
A murine Mll-AF4 knock-in model results in lymphoid and myeloid deregulation and hematologic malignancy.
Blood
2006
108
2
669
677
50
Song
WJ
Sullivan
MG
Legare
RD
et al
Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia.
Nat Genet
1999
23
2
166
175
51
Smith
ML
Cavenagh
JD
Lister
TA
Fitzgibbon
J
Mutation of CEBPA in familial acute myeloid leukemia.
N Engl J Med
2004
351
23
2403
2407
52
Nanri
T
Uike
N
Kawakita
T
Iwanaga
E
Mitsuya
H
Asou
N
A family harboring a germ-line N-terminal C/EBPalpha mutation and development of acute myeloid leukemia with an additional somatic C-terminal C/EBPalpha mutation.
Genes Chromosomes Cancer
2010
49
3
237
241
53
Müller
AMS
Duque
J
Shizuru
JA
Lübbert
M
Complementing mutations in core binding factor leukemias: from mouse models to clinical applications.
Oncogene
2008
27
44
5759
5773
54
Wiemels
JL
Xiao
Z
Buffler
PA
et al
In utero origin of t(8;21) AML1-ETO translocations in childhood acute myeloid leukemia.
Blood
2002
99
10
3801
3805
55
Nucifora
G
Larson
RA
Rowley
JD
Persistence of the 8;21 translocation in patients with acute myeloid leukemia type M2 in long-term remission.
Blood
1993
82
3
712
715
56
Jurlander
J
Caligiuri
MA
Ruutu
T
et al
Persistence of the AML1/ETO fusion transcript in patients treated with allogeneic bone marrow transplantation for t(8;21) leukemia.
Blood
1996
88
6
2183
2191
57
Miyamoto
T
Nagafuji
K
Akashi
K
et al
Persistence of multipotent progenitors expressing AML1/ETO transcripts in long-term remission patients with t(8;21) acute myelogenous leukemia.
Blood
1996
87
11
4789
4796
58
Saunders
MJ
Brereton
ML
Adams
JA
Tobal
K
Liu Yin
JA
Expression of AML1/MTG8 transcripts in clonogenic cells grown from bone marrow of patients in remission of acute myeloid leukaemia with t(8;21).
Br J Haematol
1997
99
4
921
924
59
Shih
LY
Liang
DC
Huang
CF
et al
AML patients with CEBPalpha mutations mostly retain identical mutant patterns but frequently change in allelic distribution at relapse: a comparative analysis on paired diagnosis and relapse samples.
Leukemia
2006
20
4
604
609
60
Falini
B
Martelli
MP
Bolli
N
et al
Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity?
Blood
2011
117
4
1109
1120
61
Hou
HA
Kuo
YY
Liu
CY
et al
DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications.
Blood
2012
119
2
559
568
62
Kihara
R
Hoshino
H
Kiyoi
H
Naoe
T
DNMT3A and IDH1/2 mutations are stable during the progression of acute myeloid leukemia [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
637
63
Kottaridis
PD
Gale
RE
Langabeer
SE
Frew
ME
Bowen
DT
Linch
DC
Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors.
Blood
2002
100
7
2393
2398
64
Shih
LY
Huang
CF
Wu
JH
et al
Heterogeneous patterns of FLT3 Asp(835) mutations in relapsed de novo acute myeloid leukemia: a comparative analysis of 120 paired diagnostic and relapse bone marrow samples.
Clin Cancer Res
2004
10
4
1326
1332
65
Bachas
C
Schuurhuis
GJ
Hollink
IH
et al
High-frequency type I/II mutational shifts between diagnosis and relapse are associated with outcome in pediatric AML: implications for personalized medicine.
Blood
2010
116
15
2752
2758
66
Pollard
JA
Meshinchi
S
Harrington
KH
Zeng
R
Bernstein
ID
Walter
RB
Sequential acquisition of somatic mutations in progenitors with differential proliferative potential in human acute myeloid leukemia [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
116
21
332
67
Wang
YY
Zhou
GB
Yin
T
et al
AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia: implication in stepwise leukemogenesis and response to Gleevec.
Proc Natl Acad Sci U S A
2005
102
4
1104
1109
68
Fearon
ER
Burke
PJ
Schiffer
CA
Zehnbauer
BA
Vogelstein
B
Differentiation of leukemia cells to polymorphonuclear leukocytes in patients with acute nonlymphocytic leukemia.
N Engl J Med
1986
315
1
15
24
69
Gale
RE
Wheadon
H
Goldstone
AH
Burnett
AK
Linch
DC
Frequency of clonal remission in acute myeloid leukaemia.
Lancet
1993
341
8838
138
142
70
Jowitt
SN
Liu Yin
JA
Saunders
MJ
Lucas
GS
Clonal remissions in acute myeloid leukaemia are commonly associated with features of trilineage myelodysplasia during remission.
Br J Haematol
1993
85
4
698
705
71
Jinnai
I
Nagai
K
Yoshida
S
et al
Incidence and characteristics of clonal hematopoiesis in remission of acute myeloid leukemia in relation to morphological dysplasia.
Leukemia
1995
9
10
1756
1761
72
Grimwade
D
Enver
T
Acute promyelocytic leukemia: where does it stem from?
Leukemia
2004
18
3
375
384
73
Eppert
K
Takenaka
K
Lechman
ER
et al
Stem cell gene expression programs influence clinical outcome in human leukemia.
Nat Med
2011
17
9
1086
1093
74
Pollard
JA
Alonzo
TA
Gerbing
RB
et al
FLT3 internal tandem duplication in CD34+/CD33− precursors predicts poor outcome in acute myeloid leukemia.
Blood
2006
108
8
2764
2769
75
Dinndorf
PA
Andrews
RG
Benjamin
D
Ridgway
D
Wolff
L
Bernstein
ID
Expression of normal myeloid-associated antigens by acute leukemia cells.
Blood
1986
67
4
1048
1053
76
Scheinberg
DA
Lovett
D
Divgi
CR
et al
A phase I trial of monoclonal antibody M195 in acute myelogenous leukemia: specific bone marrow targeting and internalization of radionuclide.
J Clin Oncol
1991
9
3
478
490
77
Caron
PC
Dumont
L
Scheinberg
DA
Supersaturating infusional humanized anti–CD33 monoclonal antibody HuM195 in myelogenous leukemia.
Clin Cancer Res
1998
4
6
1421
1428
78
Appelbaum
FR
Matthews
DC
Eary
JF
et al
The use of radiolabeled anti–CD33 antibody to augment marrow irradiation prior to marrow transplantation for acute myelogenous leukemia.
Transplantation
1992
54
5
829
833
79
van der Jagt
RHC
Badger
CC
Appelbaum
FR
et al
Localization of radiolabeled antimyeloid antibodies in a human acute leukemia xenograft tumor model.
Cancer Res
1992
52
1
89
94
80
Hamann
PR
Hinman
LM
Beyer
CF
et al
An anti–CD33 antibody-calicheamicin conjugate for treatment of acute myeloid leukemia: choice of linker.
Bioconjug Chem
2002
13
1
40
46
81
Damle
NK
Frost
P
Antibody-targeted chemotherapy with immunoconjugates of calicheamicin.
Curr Opin Pharmacol
2003
3
4
386
390
82
Hamann
PR
Hinman
LM
Hollander
I
et al
Gemtuzumab ozogamicin, a potent and selective anti–CD33 antibody-calicheamicin conjugate for treatment of acute myeloid leukemia.
Bioconjug Chem
2002
13
1
47
58
83
Walter
RB
Fairchild
SE
Flowers
DA
Hong
TC
Bernstein
ID
Priming with myeloid growth factors enhances CD33 expression, decreases P-glycoprotein activity, and improves efficacy of gemtuzumab ozogamicin against acute myeloid leukemia (AML) colony forming cells (CFC) [abstract].
Blood (ASH Annual Meeting Abstracts)
2008
112
11
912
913
84
Linenberger
ML
CD33-directed therapy with gemtuzumab ozogamicin in acute myeloid leukemia: progress in understanding cytotoxicity and potential mechanisms of drug resistance.
Leukemia
2005
19
2
176
182
85
Sullivan
N
Lyne
L
Sensitivity of fibroblasts derived from ataxia-telangiectasia patients to calicheamicin gamma 1I.
Mutat Res
1990
245
3
171
175
86
van Duijn-Goedhart
A
Zdzienicka
MZ
Sankaranarayanan
K
van Buul
PP
Differential responses of Chinese hamster mutagen sensitive cell lines to low and high concentrations of calicheamicin and neocarzinostatin.
Mutat Res
2000
471
1
95
105
87
Goemans
BF
Zwaan
CM
Vijverberg
SJ
et al
Large interindividual differences in cellular sensitivity to calicheamicin may influence gemtuzumab ozogamicin response in acute myeloid leukemia.
Leukemia
2008
22
12
2284
2285
88
Walter
RB
Raden
BW
Hong
TC
Flowers
DA
Bernstein
ID
Linenberger
ML
Multidrug resistance protein attenuates gemtuzumab ozogamicin-induced cytotoxicity in acute myeloid leukemia cells.
Blood
2003
102
4
1466
1473
89
Taksin
AL
Legrand
O
Raffoux
E
et al
High efficacy and safety profile of fractionated doses of Mylotarg as induction therapy in patients with relapsed acute myeloblastic leukemia: a prospective study of the alfa group.
Leukemia
2007
21
1
66
71
90
Walter
RB
Gooley
TA
van der Velden
VH
et al
CD33 expression and P-glycoprotein-mediated drug efflux inversely correlate and predict clinical outcome in patients with acute myeloid leukemia treated with gemtuzumab ozogamicin monotherapy.
Blood
2007
109
10
4168
4170
91
Walter
RB
Raden
BW
Kamikura
DM
Cooper
JA
Bernstein
ID
Influence of CD33 expression levels and ITIM-dependent internalization on gemtuzumab ozogamicin-induced cytotoxicity.
Blood
2005
105
3
1295
1302
92
Jager
E
van der Velden
VHJ
te Marvelde
JG
Walter
RB
Agur
Z
Vainstein
V
Targeted drug delivery by gemtuzumab ozogamicin: mechanism-based mathematical model for treatment strategy improvement and therapy individualization.
PLoS One
2011
6
9
e24265
93
Jedema
I
Barge
RM
van der Velden
VH
et al
Internalization and cell cycle-dependent killing of leukemic cells by gemtuzumab ozogamicin: rationale for efficacy in CD33-negative malignancies with endocytic capacity.
Leukemia
2004
18
2
316
325
94
Sievers
EL
Appelbaum
FR
Spielberger
RT
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
95
Sievers
EL
Larson
RA
Stadtmauer
EA
et al
Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse.
J Clin Oncol
2001
19
13
3244
3254
96
Bross
PF
Beitz
J
Chen
G
et al
Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia.
Clin Cancer Res
2001
7
6
1490
1496
97
Larson
RA
Sievers
EL
Stadtmauer
EA
et al
Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence.
Cancer
2005
104
7
1442
1452
98
Giles
F
Estey
E
O'Brien
S
Gemtuzumab ozogamicin in the treatment of acute myeloid leukemia.
Cancer
2003
98
10
2095
2104
99
Fenton
C
Perry
CM
Gemtuzumab ozogamicin: a review of its use in acute myeloid leukaemia.
Drugs
2005
65
16
2405
2427
100
Tsimberidou
AM
Giles
FJ
Estey
E
O'Brien
S
Keating
MJ
Kantarjian
HM
The role of gemtuzumab ozogamicin in acute leukaemia therapy.
Br J Haematol
2006
132
4
398
409
101
Abutalib
SA
Tallman
MS
Monoclonal antibodies for the treatment of acute myeloid leukemia.
Curr Pharm Biotechnol
2006
7
5
343
369
102
Pagano
L
Fianchi
L
Caira
M
Rutella
S
Leone
G
The role of Gemtuzumab Ozogamicin in the treatment of acute myeloid leukemia patients.
Oncogene
2007
26
25
3679
3690
103
Stasi
R
Evangelista
ML
Buccisano
F
Venditti
A
Amadori
S
Gemtuzumab ozogamicin in the treatment of acute myeloid leukemia.
Cancer Treat Rev
2008
34
1
49
60
104
Caron
PC
Jurcic
JG
Scott
AM
et al
A phase 1B trial of humanized monoclonal antibody M195 (anti–CD33) in myeloid leukemia: specific targeting without immunogenicity.
Blood
1994
83
7
1760
1768
105
van der Velden
VHJ
te Marvelde
JG
Hoogeveen
PG
et al
Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA-676) in acute myeloid leukemia: in vivo and in vitro saturation and internalization by leukemic and normal myeloid cells.
Blood
2001
97
10
3197
3204
106
Farhat
H
Reman
O
Raffoux
E
et al
Fractionated doses of gemtuzumab ozogamicin with escalated doses of daunorubicin and cytarabine as first acute myeloid leukemia salvage in patients aged 50-70-year old: a phase 1/2 study of the Acute Leukemia French Association.
Am J Hematol
2012
87
1
62
65
107
Lo-Coco
F
Cimino
G
Breccia
M
et al
Gemtuzumab ozogamicin (Mylotarg) as a single agent for molecularly relapsed acute promyelocytic leukemia.
Blood
2004
104
7
1995
1999
108
Breccia
M
Cimino
G
Diverio
D
Gentilini
F
Mandelli
F
Lo Coco
F
Sustained molecular remission after low dose gemtuzumab-ozogamicin in elderly patients with advanced acute promyelocytic leukemia.
Haematologica
2007
92
9
1273
1274
109
Takeshita
A
Ono
T
Kojima
Y
et al
Efficacy of gemtuzumab ozogamicin (GO) monotherapy on relapsed/refractory acute promyelocytic leukemia (APL) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
665
666
110
Estey
EH
Giles
FJ
Beran
M
et al
Experience with gemtuzumab ozogamycin (“mylotarg”) and all-trans retinoic acid in untreated acute promyelocytic leukemia.
Blood
2002
99
11
4222
4224
111
Ravandi
F
Estey
E
Jones
D
et al
Effective treatment of acute promyelocytic leukemia with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab ozogamicin.
J Clin Oncol
2009
27
4
504
510
112
Burnett
AK
Hills
RK
Milligan
D
et al
Identification of patients with acute myeloblastic leukaemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial.
J Clin Oncol
2011
29
4
369
377
113
Castaigne
S
Pautas
C
Terre
C
et al
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukemia (ALFA-0701): a randomised, open-label, phase 3 study.
Lancet
2012
379
9825
1508
1516
114
Delaunay
J
Recher
C
Pigneux
A
et al
Addition of gemtuzumab ozogamycin to chemotherapy improves event-free survival but not overall survival of AML patients with intermediate cytogenetics not eligible for allogeneic transplantation: results of the GOELAMS AML 2006 IR study [abstract].
Blood. (ASH Annual Meeting Abstracts)
2011
118
21
37
38
115
Burnett
AK
Hill
RK
Hunter
AE
et al
The addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia.
J Clin Oncol
2012
in press
116
Petersdorf
S
Kopecky
K
Stuart
RK
et al
Preliminary results of Southwest Oncology Group Study S0106: an international intergroup phase 3 randomized trial comparing the addition of gemtuzumab ozogamicin to standard induction therapy versus standard induction therapy followed by a second randomization to post-consolidation gemtuzumab ozogamicin versus no additional therapy for previously untreated acute myeloid leukemia [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
114
22
326
327
119
Lo Coco
F
Ammatuna
E
Noguera
N
Treatment of acute promyelocytic leukemia with gemtuzumab ozogamicin.
Clin Adv Hematol Oncol
2006
4
1
57
62
120
Breccia
M
Lo-Coco
F
Gemtuzumab ozogamicin for the treatment of acute promyelocytic leukemia: mechanisms of action and resistance, safety and efficacy.
Expert Opin Biol Ther
2011
11
2
225
234
121
Jurcic
JG
DeBlasio
T
Dumont
L
Yao
TJ
Scheinberg
DA
Molecular remission induction with retinoic acid and anti–CD33 monoclonal antibody HuM195 in acute promyelocytic leukemia.
Clin Cancer Res
2000
6
2
372
380
122
Jilani
I
Estey
E
Huh
Y
et al
Differences in CD33 intensity between various myeloid neoplasms.
Am J Clin Pathol
2002
118
4
560
566
123
Hauswirth
AW
Florian
S
Printz
D
et al
Expression of the target receptor CD33 in CD34+/CD38-/CD123+ AML stem cells.
Eur J Clin Invest
2007
37
1
73
82
124
Dean
M
Fojo
T
Bates
S
Tumour stem cells and drug resistance.
Nat Rev Cancer
2005
5
4
275
284
125
Jawad
M
Seedhouse
C
Mony
U
Grundy
M
Russell
NH
Pallis
M
Analysis of factors that affect in vitro chemosensitivity of leukaemic stem and progenitor cells to gemtuzumab ozogamicin (Mylotarg) in acute myeloid leukaemia.
Leukemia
2010
24
1
74
80
126
Pollard
JA
Alonzo
TA
Loken
M
et al
Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML.
Blood
2012
119
16
3705
3711
127
Lamba
J
Mortland
LJ
Mitra
A
et al
Clinical significance of CD33 non-synonymous single nucleotide polymorphisms (SNPs) in pediatric patients with acute myeloid leukemia treated with gemtuzumab ozogamicin-containing chemotherapy [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
1489
128
van der Velden
VHJ
Boeckx
N
Jedema
I
et al
High CD33-antigen loads in peripheral blood limit the efficacy of gemtuzumab ozogamicin (Mylotarg) treatment in acute myeloid leukemia patients.
Leukemia
2004
18
5
983
988
129
Jordan
CT
Guzman
ML
Mechanisms controlling pathogenesis and survival of leukemic stem cells.
Oncogene
2004
23
43
7178
7187
130
Wu
AM
Senter
PD
Arming antibodies: prospects and challenges for immunoconjugates.
Nat Biotechnol
2005
23
9
1137
1146
Sign in via your Institution