In this issue of Blood, Murtadha et al demonstrate the efficacy of a new CD38-targeting immunotherapy for acute myeloid leukemia (AML).1 

Survival rates in newly diagnosed AML have plateaued over the last several decades. Survival in the setting of relapsed disease remains dismal, making AML a disease that would benefit from novel therapies. Numerous immunotherapies have been trialed in patients with AML but with only limited success,2,3 due to a variety of barriers, including on-target/off-tumor toxicity, an immunosuppressive microenvironment, and antigen heterogeneity.4,5 AML is both genotypically and phenotypically heterogeneous compared with other hematologic malignancies, making it challenging to find a universal target. Furthermore, the condition has significant intrapatient heterogeneity, including multiple subpopulations existing within a single patient, which limits the potential of targeting a single antigen.

Experience with the monoclonal CD38-targeting antibody daratumumab has shown that the CD38 antigen has a favorable on-target/off-tumor toxicity profile. Targeting of CD38 has demonstrated encouraging clinical efficacy, such as approval for use in multiple myeloma and exploration in numerous other hematologic malignancies, including T-cell acute lymphoblastic leukemia.6 Although the majority of bulk AML cells express CD38, there is limited expression on leukemic stem cells (LSCs), bringing the viability of targeting CD38 into question, as LSCs are implicated in the propagation and relapse of AML.7 

Modulation of CD38 has been established but has primarily been limited to preclinical modeling.8 In their study, Murtadha et al hypothesize that modulation of CD38 would make it a more compelling target in AML if its expression on LSCs could be successfully induced. Through a series of elegant experiments, they demonstrate that CD38 surface expression in AML blasts is increased in the presence of interferon gamma (IFN-γ), primarily through the upregulation of the transcriptional regulator interferon regulatory factor-1. Because T cells release IFN-γ with activation, there was sufficient mechanistic rationale to explore a novel CD38-directed T-cell engager in AML.

This led to the creation of the single-chain bispecific antibody CD38-CD3 BIONIC (biologics nested inside chains; BN-CD38) by inserting a CD38 nanobody between the light and heavy chains of an anti-CD3 antibody. This compact approach was favored over traditional double-chain approaches because it was hypothesized it would enhance function by mimicking the normal immunologic synapse by decreasing the distance between the T-cell receptor and the major histocompatibility complex. The authors show that coculture of BN-CD38 with T cells and AML cell lines led to activation of T cells (via surface expression of CD69 and CD25), secretion of proinflammatory cytokines (including IFN-γ), and clearance of CD38+ cells. These changes were not demonstrated with their mutated CD38 control and were largely abrogated in the presence of antihuman IFN-γ–neutralizing antibody.

Most importantly, the authors show the successful incorporation of BN-CD38 in multiple patient-derived xenograft models. This includes one model from a patient with relapsed AML with a CD34+/CD38 fraction of >20% and treated with weekly BN-CD38 and human T-cell infusions. In this model, the majority of BN-CD38–treated mice remained disease-free compared with the control mice, which had a median survival of 12 weeks (P = .0007). Beyond inducing the qualitative expression of CD38 on LSCs, it was also noted that BN-CD38 increased CD38 expression on lower expressing bulk cells. This is potentially of equal clinical importance because level of antigen expression has been linked to response with antibody–drug conjugates in AML and other hematologic malignancies.9,10 

An intriguing possibility is that the phenomenon of IFN-γ–induced upregulation of CD38 could be extrapolated to other cell-based therapies, such as chimeric antigen receptor (CAR) T cells, which have shown limited success in the AML space but are also associated with robust IFN-γ expression. Murtadha and colleagues provide further support for the use of CD38-targeting CAR T cells in AML while also raising the possibility of combinatorial strategies, such as adding daratumumab to CAR T cells targeting alternative antigens.

Interestingly, there was no modulation of CD38 on normal immune cells and, paradoxically, there was limited on-target/off-tumor toxicity based on the lack of clearance of normal CD38-expressing cells, including normal hematopoietic stem cells. The lack of targeting of normal hematopoietic cells suggests that the hematologic toxicities of BN-CD38 should be relatively nominal, potentially overcoming a common limitation of other immunotherapeutic AML approaches. However, although reassuring, this finding is equally perplexing as it is distinct from other immunotherapies for hematologic malignancies where on-target/off-tumor toxicity is sometimes used as a surrogate for functionality (eg, CD19-targeting blinatumomab and CAR T-cell therapies eliminate both malignant and normal CD19-expressing B cells). Thus, future studies should aim to confirm this finding in other humanized model systems and seek to identify possible underlying protective mechanisms.

With a rigorous series of in vitro, in vivo, and ex vivo experiments, Murtadha et al demonstrate the viability of targeting CD38 in AML with a T-cell engager, particularly due to the IFN-γ–induced CD38 upregulation and clearance of LSCs (see figure). Their work provides mechanistic clarity into immunotherapeutic treatment barriers and contributes to the growing body of evidence supporting the successful incorporation of immunotherapy for AML.

BN-CD38 eradicates both CD38+ and CD38 AML cells including LSCs. BN-CD38 binding to CD3 on T cells and CD38 on AML blasts results in engagement and activation T cells leading to eradication of CD38+ AML blasts and release of cytokines, including IFN-γ. Binding of released IFN-γ to IFN-γR on the surface of CD38-negative (CD38neg) LSCs induces expression of CD38, allowing for cell killing by engagement of T cells directed by BN-CD38. IFN-γR, interferon gamma receptor. Created with BioRender.com.

BN-CD38 eradicates both CD38+ and CD38 AML cells including LSCs. BN-CD38 binding to CD3 on T cells and CD38 on AML blasts results in engagement and activation T cells leading to eradication of CD38+ AML blasts and release of cytokines, including IFN-γ. Binding of released IFN-γ to IFN-γR on the surface of CD38-negative (CD38neg) LSCs induces expression of CD38, allowing for cell killing by engagement of T cells directed by BN-CD38. IFN-γR, interferon gamma receptor. Created with BioRender.com.

Close modal

Conflict-of-interest disclosure: The authors declare no competing financial interests.

1.
Murtadha
M
,
Park
M
,
Zhu
Y
, et al
.
A CD38-directed, single-chain T-cell engager targets leukemia stem cells through IFN-γ–induced CD38 expression
.
Blood
.
2024
;
143
(
16
):
1599
-
1615
.
2.
Daver
N
,
Garcia-Manero
G
,
Basu
S
, et al
.
Efficacy, safety, and biomarkers of response to azacitidine and nivolumab in relapsed/refractory acute myeloid leukemia: a nonrandomized, open-label, phase II study
.
Cancer Discov
.
2019
;
9
(
3
):
370
-
383
.
3.
Uy
GL
,
Aldoss
I
,
Foster
MC
, et al
.
Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia
.
Blood
.
2021
;
137
(
6
):
751
-
762
.
4.
Vanhooren
J
,
Dobbelaere
R
,
Derpoorter
C
, et al
.
CAR-T in the treatment of acute myeloid leukemia: barriers and how to overcome them
.
Hemasphere
.
2023
;
7
(
9
):
e937
.
5.
Lamble
AJ
,
Tasian
SK
.
Opportunities for immunotherapy in childhood acute myeloid leukemia
.
Blood Adv
.
2019
;
3
(
22
):
3750
-
3758
.
6.
Sonneveld
P
,
Dimopoulos
MA
,
Boccadoro
M
, et al
.
Daratumumab, bortezomib, lenalidomide, and dexamethasone for multiple myeloma
.
N Engl J Med
.
2024;390(4):301-313
.
7.
Jordan
CT
.
The leukemic stem cell
.
Best Pract Res Clin Haematol
.
2007
;
20
(
1
):
13
-
18
.
8.
Costa
F
,
Dalla Palma
B
,
Giuliani
N
.
CD38 expression by myeloma cells and its role in the context of bone marrow microenvironment: modulation by therapeutic agents
.
Cells
.
2019;8(12)
:
1632
.
9.
Biberacher
V
,
Decker
T
,
Oelsner
M
, et al
.
The cytotoxicity of anti-CD22 immunotoxin is enhanced by bryostatin 1 in B-cell lymphomas through CD22 upregulation and PKC-II depletion
.
Haematologica
.
2011
;
97
(
5
):
771
-
779
.
10.
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
.
Sign in via your Institution