In this issue of Blood, Kung Sutherland et al report on the preclinical activity of SGN-CD33A, a humanized anti-CD33 antibody conjugated to a pyrrolobenzodiazepine (PBD) dimer via a protease-cleavable linker, against acute myeloid leukemia (AML) cells in vitro and in vivo.1 

At this time, there is a void in the field of antibody–drug conjugates (ADC) targeting CD33 as a therapeutic option for treatment of AML after the voluntary withdrawal of gemtuzumab ozogamicin (GO) from the market.

Even though CD33 is an antigen that is both considered to be expressed in more committed myeloid precursors2,3  and is not present in AML stem cells, the clinical relevance of CD33 as a target is validated by survival benefit with GO in subgroups of patients with AML in randomized clinical trials.4-6  The reliable efficacy of drug conjugation with antibody and the greater stability of conjugate to avoid the exposure of non-target-expressing tissue to drugs can improve CD33 ADCs as therapeutic agents. The PBD dimer released after protease cleavage of SGN-CD33A causes DNA cross-linking and is capable of inducing cell cycle arrest and apoptosis. Engineered cysteine moieties at linker attachment sites allow more precise PBD dimer loading of antibody, thus improving predictability of payload delivery. The activity of SGN-CD33A requires CD33 expression, but its activity does not correlate with levels of CD33 surface expression. In vitro studies with SGN-CD33A showed approximately 3-fold more potency than GO shows against primary AML cells. What makes SGN-CD33A potentially interesting is that this ADC appears to have activity irrespective of the multidrug resistance phenotype and p53 status of AML cells. Additional in vivo studies using immune-competent isogeneic mouse models of disseminated AML with relevant translocations/mutations and variable p53 backgrounds are needed to add valuable information to the current report. In addition, data regarding levels of “naked” PBD dimer in plasma after infusions are important, as this may have relevance with regard to nonspecific toxicity.

There is a distinct need in the field for CD33-based ADCs to quickly fill the void left by the withdrawal of GO from the market, and the rapid preclinical and clinical development of agents such as SGN-CD33A is needed.

Conflict-of-interest disclosure:The author declares no competing financial interests.

1
Kung Sutherland
 
MS
Walter
 
RB
Jeffrey
 
SC
, et al. 
SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML.
Blood
2013
, vol. 
122
 
8
(pg. 
1455
-
1463
)
2
Andrews
 
RG
Torok-Storb
 
B
Bernstein
 
ID
Myeloid-associated differentiation antigens on stem cells and their progeny identified by monoclonal antibodies.
Blood
1983
, vol. 
62
 
1
(pg. 
124
-
132
)
3
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
, vol. 
68
 
5
(pg. 
1030
-
1035
)
4
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
5
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
, vol. 
30
 
32
(pg. 
3924
-
3931
)
6
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. 2011;118(21):37-38
Sign in via your Institution