On 1 September 2017, the US Food and Drug Administration (FDA) approved gemtuzumab ozogamicin (GO) for the treatment of adults with newly diagnosed CD33+ acute myeloid leukemia and for patients aged ≥2 years with CD33+ acute myeloid leukemia who have experienced a relapse or who have not responded to initial treatment. This signals a new chapter in the long and unusual story of GO, which was the first antibody–drug conjugate approved for human use by the FDA.

CD33 (or Siglec 3) is a transmembrane receptor expressed by cells of myeloid origin, but not by normal hematopoietic stem cells.1,2  The idea of using a CD33-targeted drug conjugate in the treatment of acute myeloid leukemia (AML) was initially suggested by the widespread expression of CD33 by AML blasts and the finding that in some cases, AML precursors are mainly or entirely CD33+.2  The latter was first demonstrated using X-linked glucose-6-phosphate dehydrogenase isoenzymes as markers for clonality, showing that in vitro treatment of clonal AML samples from glucose-6-phosphate dehydrogenase heterozygous women with anti-CD33 antibody plus complement could result in outgrowth of nonclonal, presumably normal hematopoiesis.3  Further, we found that following IV infusion, radiolabeled anti-CD33 antibodies selectively bound to AML cells in the peripheral blood and marrow and were rapidly internalized, suggesting their use as a carrier for a potent drug or toxin.4-6  In collaboration with industry, we selected as the toxin N-acetyl γ-calicheamicin dimethyl hydrazide, a derivative of a natural enediyne DNA-damaging antitumor antibiotic.7  The anti-CD33 antibody was humanized and conjugated to the calicheamicin derivative using an acid-labile linker that was stable in circulation but, once internalized, quickly released the toxin under the acidic conditions of the lysosome, leading to DNA binding and cell death.8  Following confirmation of the activity of the drug conjugate in a xenograft model, we conducted a phase 1 dose-escalation study in 40 patients with relapsed/refractory AML.9  Elimination of morphologically detectable AML occurred in 20% of patients, with 12.5% achieving a complete remission (CR) or CR with incomplete platelet recovery (CRp). Dose escalation was stopped at 9 mg/m2, because this dose saturated AML CD33-binding sites, even in patients with substantial numbers of circulating blasts. Because no dose-limiting nonhematological toxicities were identified, this dose (given on days 1 and 14) was chosen for 3 open-label multicenter single arm studies for patients with CD33+ AML in first relapse. An interim analysis of 142 patients (median age, 61 years) found that 16.2% achieved a CR and another 13.4% achieved a CRp, for an overall CR + CRp rate of 29.6%.10,11  Based on these results, in May 2000, gemtuzumab ozogamicin (GO) was given accelerated approval for treatment of patients age >60 years with CD33+ AML who were not candidates for aggressive chemotherapy. As part of their postapproval commitment, the drug sponsor worked with the Southwest Oncology Group to develop study S0106, which randomized 637 adults up to age 60 years with de novo AML to receive up to 2 cycles of standard daunorubicin/cytarabine induction with or without a single dose of GO (6 mg/m2) on day 4.12  In an effort to balance toxicities on the 2 arms, the daunorubicin dose was reduced from 60 mg/m2 in the control arm to 45 mg/m2 in the GO arm. The results of the study showed no overall improvement in survival and increased treatment-related mortality in the experimental arm, leading to GO’s withdrawal from the commercial market in October 2010. The recent reapplication and US Food and Drug Administration (FDA) approval are based on increased understanding of GO dosing, specific results of the pivotal study Acute Leukemia French Association study 701 (ALFA 701), and extensive additional clinical experience with the drug.

In the study leading to the original accelerated approval of GO, the observed toxicities of single-agent GO at 9 mg/m2 included infusion-related toxicities (chills, fever, and mild hypotension), the significant myelosuppression expected when targeting an early myeloid differentiation antigen, and mild, transient bilirubin elevations (seen in 23% of patients).10  Only 1 out of 142 patients had a bilirubin elevation >10× normal, and as a result, GO dosing of 6 to 9 mg/m2 was often chosen for subsequent studies. As the drug began being used in more heavily pretreated patients or in combination with other agents, an increased incidence of toxicities, particularly veno-occlusive disease (VOD) of the liver, began to be observed.13-15  In the briefing document prepared by the FDA for the recent GO review, the sponsors reported, and the FDA confirmed, that a substantially lower maximum serum concentration (Cmax) would be expected with a 3 mg/m2 dose of GO given on days 1, 4, and 7 compared with 9 mg/m2 given on days 1 and 14 (382 vs 1730 ng/mL).16  They further found in an analysis of 358 patients receiving single-agent GO that the incidence of VOD was highly associated with Cmax after the first dose of GO. In a review of studies of GO monotherapy, they observed that the CR rate was equivalent using GO 3 mg/m2 on days 1, 4, and 7 compared with doses of 6 or 9 mg/m2 given on days 1 and 14.16  In addition to the use of lower individual doses resulting in a lower Cmax, ALFA-701 also introduced fractionated dosing in an effort to not only overcome the effects of the initial high levels of peripheral blasts present on day 1 that can act as a sink preventing the drug from binding marrow blasts but also take advantage of rapid reexpression of CD33 molecules on the cell surface of AML blasts after a first exposure to antibody.17-19 

The pivotal trial presented by the sponsor to the FDA was ALFA-0701, a French multicenter, open-label, 1:1 randomized trial of GO (3 mg/m2 on days 1, 4, and 7) plus daunorubicin (60 mg/m2 on days 1 to 3) and cytarabine (200 mg/m2 as continuous infusion on days 1 to 7) (DA) versus DA alone for induction and consolidation involving 271 patients age 50 to 70 years with untreated de novo AML.20  The CR rates in the 2 arms were similar (70.4% vs 69.9% for GO + DA vs DA), but the 3-year event-free survival (the primary end point of the study) was significantly higher in the GO + DA arm (39.8% vs 13.6%, P < .001). Along with S0106 and ALFA-0701, 3 other randomized trials testing the addition of GO in induction therapy have been conducted and reported.21-23  These studies differed somewhat in patient-related variables, induction regimens, and the dose and schedule of GO.24  A meta-analysis that included the 3325 patients entered onto these 5 studies concluded that, although not increasing the proportion of patients achieving a complete remission (odds ratio [OR], 0.91; P = .3), the addition of GO significantly reduced the risk of relapse (OR, 0.81; P = .0001) and improved relapse-free (OR, 0.87; P = .005) and overall survival at 5 years (OR, 0.90; P = .01).24  The survival benefit was most apparent in patients with favorable risk cytogenetics (OR, 0.47; P = .0006) and was also seen in those with intermediate-risk disease (OR, 0.84; P = .005), but not in those with adverse-risk AML (OR, 0.99; P = .9) (Figure 1). GO doses of 3 mg/m2 were as effective as higher doses and had improved safety. The equivalent effectiveness and improved safety of 3 mg/m2 vs 6 mg/m2 was confirmed in a subsequent prospective randomized trial comparing the 2 dose regimens (NCRI AML-17).25  No studies have directly compared the fractionated dosing regimen used in the French ALFA-0701 with any of the single-dose regimens used in other trials, nor are the relative contributions of GO during induction vs consolidation understood.

Figure 1.

Overall survival according to cytogenetic risk category. The 3 sets of curves illustrate the overall survival of patients randomized to combination chemotherapy with or without GO. (A) Overall survival of 246 patients with favorable-risk AML. (B) Overall survival of 1827 patients with intermediate-risk AML. (C) Overall survival of 583 with adverse-risk disease.24  SD, standard deviation.

Figure 1.

Overall survival according to cytogenetic risk category. The 3 sets of curves illustrate the overall survival of patients randomized to combination chemotherapy with or without GO. (A) Overall survival of 246 patients with favorable-risk AML. (B) Overall survival of 1827 patients with intermediate-risk AML. (C) Overall survival of 583 with adverse-risk disease.24  SD, standard deviation.

Close modal

GO has also been studied in pediatric AML, first in a pilot study of 350 patients demonstrating the safety of adding GO at 3 mg/m2 to induction and consolidation and then in a prospective study (AAML0531) that randomized 1022 patients age 0 to 29 years to induction and consolidation chemotherapy with or without GO.26,27  Remission rates were not improved with GO, but relapse risk was significantly reduced, resulting in improved event-free survival (53.1% vs 46.9% at 3 years; P = .04).

GO cytotoxicity requires antibody-conjugate binding to the AML cell surface, followed by internalization, hydrolysis of the linker (allowing calicheamicin activation and release from the lysosome), and calicheamicin binding to DNA (inducing single- and double-strand breaks), followed by activation of downstream pathways leading to apoptotic cell death. Factors affecting each step have been identified and shown to influence GO effectiveness in vitro.28  Critical factors identified in clinical studies include variation in CD33 expression and multidrug-resistance mechanisms.

Clinical studies demonstrate as much as a 2 log-fold variation in CD33 expression by AML blasts with lower levels of CD33 seen in AML cases with adverse karyotype and in patients with core-binding factor (CBF) AML, whereas high expression is seen in AMLs with FLT3-ITD, MLL, and NPM1 mutations.29-33  CD33 expression itself does not appear to have a marked effect on AML treatment outcome using chemotherapy alone, but the addition of GO (at least at 3 mg/m2) appears to preferentially benefit those with higher expression levels.25,31,33  An exception may be patients with CBF AML, who appear to benefit from GO despite relatively low CD33 levels, a finding that might be explained by the inherent sensitivity of CBF blasts to cytotoxic insult. An alternative explanation is that in CBF leukemias overt disease arises from a CD33+ precursor.2  Consistent with this notion is the high activity of GO in the treatment of acute promyelocytic leukemia, which is also thought to arise from a more mature precursor.34,35  In fact, it may be that sensitivity to GO helps define the level of differentiation of the leukemic stem cell.

Although some of the variability in CD33 expression by AML blasts relates to cytogenetic and mutational subtypes, much was unexplained until the recent demonstration that a germline CD33 single-nucleotide polymorphism, rs12459419, is associated with the expression of an alternatively spliced variant of CD33 lacking the GO-binding site (immunoglobulin V domain).36,37  Essentially all available diagnostic antibodies target the same CD33 immunoglobulin V domain, so this single-nucleotide polymorphism defines whether AML blasts will appear to be CD33+ or CD33. In the recently published pediatric AAML0531 study, the rs12459419 genotype was CC (and therefore able to bind GO) in 51% of patients, CT in 39%, and TT in 10%.38  The addition of GO significantly lowered the relapse rate in patients with the CC genotype (26% vs 49%, P ≤ .001) but had no effect on patients with CT or TT genotypes, resulting in a disease-free survival benefit with GO that was restricted to patients with the CC genotype.

In addition to the nature and number of CD33-binding sites, the intracellular accumulation of the toxic moiety is affected by rates of antibody-conjugate internalization and the level of adenosine triphosphate–binding cassette transporter activity such as P-glycoprotein, which exports calicheamicin from the cell.29  Theoretically, because GO is delivered with relative specificity to leukemic marrow, adenosine triphosphate–binding cassette transport inhibition should improve the efficacy of the drug in high P-glycoprotein–expressing AMLs. To exert its cytotoxic effect, calicheamicin binds to the minor groove of DNA in a sequence-specific manner, inducing DNA single- and double-strand breaks that in turn initiate downstream pro- and antisurvival signaling events. Less is known about how these responses are modulated, but it is sobering to realize that there are many-fold differences in the sensitivity of AML clinical samples exposed to free calicheamicin using a traditional 4-day 3-(4,5-dimethylthiazol-2-yl)-2,5-dimethyltetrazolium bromide assay.29 

The acute infusion-related toxicities seen with GO are transient and usually respond to standard interventions. As expected with an agent that eliminates early myeloid precursors, bone marrow suppression is seen when GO is used as a single agent. When added to combination chemotherapy in the ALFA-701 trial, myeloid recovery following induction was not delayed, whereas platelet recovery was prolonged by 4 days. The toxicity of greatest concern is the development of VOD. The risk of VOD appears to be relatively low when individual doses of no greater than 3 mg/m2 are used in combination with conventional therapy as part of initial therapy of AML, but the risks increase with higher doses or use of the drug in more heavily pretreated patients.39  The recent observation that VOD is also seen with inotuzumab ozogamicin, an anti-CD22 calicheamicin conjugate, suggests that a component of GO-associated VOD is CD33 independent,40  whereas the appearance of liver toxicity associated with SGN-CD33A, which uses a pyrrolobenzodiazepine dimer instead of calicheamicin, argues that targeting CD33 may also contribute.41 

The results of ALFA-701 and the meta-analysis of randomized trials support the safety and efficacy of the addition of GO to induction therapy for patients with newly diagnosed CD33+ AML, establishing this as a new standard of care. The outstanding results reported from several albeit nonrandomized trials with the use of GO in recurrent APL and the combination of GO, all-trans-retinoic acid, and arsenic trioxide in the treatment of patients with newly diagnosed high-risk APL make these attractive treatment approaches as well.34,42,43  Going forward, the most fruitful studies of GO itself may be those investigating the implications of CD33 splice variation and those that attempt to improve our understanding of the cellular basis for susceptibility to GO, focusing on GO uptake and trafficking, sensitivity to DNA damage, and expression of CD33 by the AML stem cell. The recent approval of midostaurin for FLT3-mutated AML and CPX-351 for therapy-related AML or AML with myelodysplasia-related changes presents the additional challenge of defining the relative benefits of these 3 drugs in specific subsets of AML and whether (and how) they can be effectively and safely combined.

Contribution: F.R.A. and I.D.B. wrote the paper.

Conflict-of-interest disclosure: F.R.A. is on the advisory board of Adaptive Biotechnologies. I.D.B. declares no competing financial interests.

Correspondence: Frederick R. Appelbaum, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave North, D5-310, PO Box 19024, Seattle, WA 98109-1024; e-mail: fappelba@fhcrc.org.

1.
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
.
2.
Walter
RB
,
Appelbaum
FR
,
Estey
EH
,
Bernstein
ID
.
Acute myeloid leukemia stem cells and CD33-targeted immunotherapy
.
Blood
.
2012
;
119
(
26
):
6198
-
6208
.
3.
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
.
4.
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
.
5.
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
.
6.
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
.
7.
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
.
8.
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
.
9.
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
.
10.
Sievers
EL
,
Larson
RA
,
Stadtmauer
EA
, et al
;
Mylotarg Study Group
.
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
.
11.
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
.
12.
Petersdorf
SH
,
Kopecky
KJ
,
Slovak
M
, et al
.
A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia
.
Blood
.
2013
;
121
(
24
):
4854
-
4860
.
13.
McKoy
JM
,
Angelotta
C
,
Bennett
CL
, et al
.
Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project
.
Leuk Res
.
2007
;
31
(
5
):
599
-
604
.
14.
Magwood-Golston
JS
,
Kessler
S
,
Bennett
CL
.
Evaluation of gemtuzumab ozogamycin associated sinusoidal obstructive syndrome: findings from an academic pharmacovigilance program review and a pharmaceutical sponsored registry
.
Leuk Res
.
2016
;
44
:
61
-
64
.
15.
Wadleigh
M
,
Richardson
PG
,
Zahrieh
D
, et al
.
Prior gemtuzumab ozogamicin exposure significantly increases the risk of veno-occlusive disease in patients who undergo myeloablative allogeneic stem cell transplantation
.
Blood
.
2003
;
102
(
5
):
1578
-
1582
.
16.
Oncologic Drugs Advisory Committee. FDA Briefing Document: BLA 761060, Myelotarg (gemtuzumab ozogamicin). Applicant: Wyeth Pharmaceuticals Inc., a subsidiary of Pfizer Inc. https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/OncologicDrugsAdvisoryCommittee/UCM566013.pdf. Accessed 15 August 2017
.
17.
van der Velden
VH
,
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
.
18.
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
.
19.
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
.
20.
Castaigne
S
,
Pautas
C
,
Terré
C
, et al
;
Acute Leukemia French Association
.
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
.
2012
;
379
(
9825
):
1508
-
1516
.
21.
Burnett
AK
,
Hills
RK
,
Milligan
D
, et al
.
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial
.
J Clin Oncol
.
2011
;
29
(
4
):
369
-
377
.
22.
Burnett
AK
,
Russell
NH
,
Hills
RK
, et al
.
Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia
.
J Clin Oncol
.
2012
;
30
(
32
):
3924
-
3931
.
23.
Delaunay
J
,
Recher
C
,
Pigneux
A
, et al
. Addition of gemtuzumab ozogamicin 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.
2011
;118(21). Abstract 79.
24.
Hills
RK
,
Castaigne
S
,
Appelbaum
FR
, et al
.
Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials
.
Lancet Oncol
.
2014
;
15
(
9
):
986
-
996
.
25.
Burnett
A
,
Cavenagh
J
,
Russell
N
, et al
;
UK NCRI AML Study Group
.
Defining the dose of gemtuzumab ozogamicin in combination with induction chemotherapy in acute myeloid leukemia: a comparison of 3 mg/m2 with 6 mg/m2 in the NCRI AML17 Trial
.
Haematologica
.
2016
;
101
(
6
):
724
-
731
.
26.
Cooper
TM
,
Franklin
J
,
Gerbing
RB
, et al
.
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
.
2012
;
118
(
3
):
761
-
769
.
27.
Gamis
AS
,
Alonzo
TA
,
Meshinchi
S
, et al
.
Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531
.
J Clin Oncol
.
2014
;
32
(
27
):
3021
-
3032
.
28.
Godwin
CD
,
Gale
RP
,
Walter
RB
.
Gemtuzumab ozogamicin in acute myeloid leukemia
.
Leukemia
.
2017
;
31
(
9
):
1855
-
1868
.
29.
Walter
RB
,
Gooley
TA
,
van der Velden
VHJ
, 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
.
30.
Olombel
G
,
Guerin
E
,
Guy
J
, et al
.
The level of blast CD33 expression positively impacts the effect of gemtuzumab ozogamicin in patients with acute myeloid leukemia
.
Blood
.
2016
;
127
(
17
):
2157
-
2160
.
31.
Khan
N
,
Hills
RK
,
Virgo
P
, et al
.
Expression of CD33 is a predictive factor for effect of gemtuzumab ozogamicin at different doses in adult acute myeloid leukaemia
.
Leukemia
.
2017
;
31
(
5
):
1059
-
1068
.
32.
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
.
33.
Pollard
JA
,
Loken
M
,
Gerbing
RB
, et al
.
CD33 Expression and its association with gemtuzumab ozogamicin response: results from the randomized phase III Children’s Oncology Group trial AAML0531
.
J Clin Oncol
.
2016
;
34
(
7
):
747
-
755
.
34.
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
.
35.
Thomas
D
,
Majeti
R
.
Biology and relevance of human acute myeloid leukemia stem cells
.
Blood
.
2017
;
129
(
12
):
1577
-
1585
.
36.
Malik
M
,
Chiles
J
III
,
Xi
HS
, et al
.
Genetics of CD33 in Alzheimer’s disease and acute myeloid leukemia
.
Hum Mol Genet
.
2015
;
24
(
12
):
3557
-
3570
.
37.
Laszlo
GS
,
Harrington
KH
,
Gudgeon
CJ
, et al
.
Expression and functional characterization of CD33 transcript variants in human acute myeloid leukemia
.
Oncotarget
.
2016
;
7
(
28
):
43281
-
43294
.
38.
Lamba
JK
,
Chauhan
L
,
Shin
M
, et al
.
CD33 splicing polymorphism determines gemtuzumab ozogamicin response in de novo acute myeloid leukemia: report from randomized phase III Children’s Oncology Group trial AAML0531
.
J Clin Oncol
.
2017
;
35
(
23
):
2674
-
2682
.
39.
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
.
40.
Kantarjian
HM
,
DeAngelo
DJ
,
Stelljes
M
, et al
.
Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia
.
N Engl J Med
.
2016
;
375
(
8
):
740
-
753
.
41.
Godwin
CD
,
McDonald
GB
,
Walter
RB
.
Sinusoidal obstruction syndrome following CD33-targeted therapy in acute myeloid leukemia
.
Blood
.
2017
;
129
(
16
):
2330
-
2332
.
42.
Burnett
AK
,
Russell
NH
,
Hills
RK
, et al
;
UK National Cancer Research Institute Acute Myeloid Leukaemia Working Group
.
Arsenic trioxide and all-trans retinoic acid treatment for acute promyelocytic leukaemia in all risk groups (AML17): results of a randomised, controlled, phase 3 trial
.
Lancet Oncol
.
2015
;
16
(
13
):
1295
-
1305
.
43.
Abaza
Y
,
Kantarjian
H
,
Garcia-Manero
G
, et al
.
Long-term outcome of acute promyelocytic leukemia treated with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab
.
Blood
.
2017
;
129
(
10
):
1275
-
1283
.
Sign in via your Institution