• Multiple mechanisms drive CD22 antigen escape, including epitope loss (protein truncation and destabilization) and epitope alteration.

  • Hypermutation caused by error-prone DNA damage repair may serve as a driver of CD22 mutation and escape.

Abstract

Inotuzumab ozogamicin (InO) is an antibody-drug conjugate that delivers calicheamicin to CD22-expressing cells. In a retrospective cohort of InO-treated patients with B-cell acute lymphoblastic leukemia, we sought to understand the genomic determinants of the response and resistance to InO. Pre- and post-InO–treated patient samples were analyzed by whole genome, exome, and/or transcriptome sequencing. Acquired CD22 mutations were observed in 11% (3/27) of post-InO-relapsed tumor samples, but not in refractory samples (0/16). There were multiple CD22 mutations per sample and the mechanisms of CD22 escape included epitope loss (protein truncation and destabilization) and epitope alteration. Two CD22 mutant cases were post-InO hyper-mutators resulting from error-prone DNA damage repair (nonhomologous/alternative end-joining repair, or mismatch repair deficiency), suggesting that hypermutation drove escape from CD22-directed therapy. CD22-mutant relapses occurred after InO and subsequent hematopoietic stem cell transplantation (HSCT), suggesting that InO eliminated the predominant clones, leaving subclones with acquired CD22 mutations that conferred resistance to InO and subsequently expanded. Acquired loss-of-function mutations in TP53, ATM, and CDKN2A were observed, consistent with a compromise of the G1/S DNA damage checkpoint as a mechanism for evading InO-induced apoptosis. Genome-wide CRISPR/Cas9 screening of cell lines identified DNTT (terminal deoxynucleotidyl transferase) loss as a marker of InO resistance. In conclusion, genetic alterations modulating CD22 expression and DNA damage response influence InO efficacy. Our findings highlight the importance of defining the basis of CD22 escape and eradication of residual disease before HSCT. The identified mechanisms of escape from CD22-targeted therapy extend beyond antigen loss and provide opportunities to improve therapeutic approaches and overcome resistance. These trials were registered at www.ClinicalTrials.gov as NCT01134575, NCT01371630, and NCT03441061.

1.
Shor
B
,
Gerber
HP
,
Sapra
P
.
Preclinical and clinical development of inotuzumab-ozogamicin in hematological malignancies
.
Mol Immunol
.
2015
;
67
(
2 Pt A
):
107
-
116
.
2.
Dörken
B
,
Moldenhauer
G
,
Pezzutto
A
, et al
.
HD39 (B3), a B lineage-restricted antigen whose cell surface expression is limited to resting and activated human B lymphocytes
.
J Immunol
.
1986
;
136
(
12
):
4470
-
4479
.
3.
Zein
N
,
Sinha
AM
,
McGahren
WJ
,
Ellestad
GA
.
Calicheamicin gamma 1I: an antitumor antibiotic that cleaves double-stranded DNA site specifically
.
Science
.
1988
;
240
(
4856
):
1198
-
1201
.
4.
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
.
5.
Wynne
J
,
Wright
D
,
Stock
W
.
Inotuzumab: from preclinical development to success in B-cell acute lymphoblastic leukemia
.
Blood Adv
.
2019
;
3
(
1
):
96
-
104
.
6.
Jabbour
EJ
,
Haddad
FG
,
Short
NJ
, et al
.
Phase II study of inotuzumab ozogamicin for measurable residual disease in acute lymphoblastic leukemia in remission
.
Blood
.
2024
;
143
(
5
):
417
-
421
.
7.
Sotillo
E
,
Barrett
DM
,
Black
KL
, et al
.
Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy
.
Cancer Discov
.
2015
;
5
(
12
):
1282
-
1295
.
8.
Orlando
EJ
,
Han
X
,
Tribouley
C
, et al
.
Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia
.
Nat Med
.
2018
;
24
(
10
):
1504
-
1506
.
9.
Zhao
Y
,
Aldoss
I
,
Qu
C
, et al
.
Tumor-intrinsic and -extrinsic determinants of response to blinatumomab in adults with B-ALL
.
Blood
.
2021
;
137
(
4
):
471
-
484
.
10.
O'Brien
MM
,
Ji
L
,
Shah
NN
, et al
.
Phase II trial of inotuzumab ozogamicin in children and adolescents with relapsed or refractory B-cell acute lymphoblastic leukemia: children's oncology group protocol AALL1621
.
J Clin Oncol
.
2022
;
40
(
9
):
956
-
967
.
11.
Zheng
S
,
Gillespie
E
,
Naqvi
AS
, et al
.
Modulation of CD22 protein expression in childhood leukemia by pervasive splicing aberrations: implications for CD22-directed immunotherapies
.
Blood Cancer Discov
.
2022
;
3
(
2
):
103
-
115
.
12.
Pennesi
E
,
Michels
N
,
Brivio
E
, et al
.
Inotuzumab ozogamicin as single agent in pediatric patients with relapsed and refractory acute lymphoblastic leukemia: results from a phase II trial
.
Leukemia
.
2022
;
36
(
6
):
1516
-
1524
.
13.
Chang
HHY
,
Pannunzio
NR
,
Adachi
N
,
Lieber
MR
.
Non-homologous DNA end joining and alternative pathways to double-strand break repair
.
Nat Rev Mol Cell Biol
.
2017
;
18
(
8
):
495
-
506
.
14.
Dahm
K
.
Functions and regulation of human artemis in double strand break repair
.
J Cell Biochem
.
2007
;
100
(
6
):
1346
-
1351
.
15.
Smith
J
,
Tho
LM
,
Xu
N
,
Gillespie
DA
.
The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer
.
Adv Cancer Res
.
2010
;
108
:
73
-
112
.
16.
Kastan
MB
,
Bartek
J
.
Cell-cycle checkpoints and cancer
.
Nature
.
2004
;
432
(
7015
):
316
-
323
.
17.
Tirrò
E
,
Massimino
M
,
Romano
C
, et al
.
Chk1 inhibition restores inotuzumab ozogamicin citotoxicity in CD22-positive cells expressing mutant p53
.
Front Oncol
.
2019
;
9
:
57
.
18.
Dobin
A
,
Davis
CA
,
Schlesinger
F
, et al
.
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
.
2013
;
29
(
1
):
15
-
21
.
19.
Li
H
,
Durbin
R
.
Fast and accurate short read alignment with burrows-wheeler transform
.
Bioinformatics
.
2009
;
25
(
14
):
1754
-
1760
.
20.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
, et al
.
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
.
2013
;
31
(
3
):
213
-
219
.
21.
Wang
K
,
Li
M
,
Hakonarson
H
.
ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data
.
Nucleic Acids Res
.
2010
;
38
(
16
):
e164
.
22.
Zhou
X
,
Edmonson
MN
,
Wilkinson
MR
, et al
.
Exploring genomic alteration in pediatric cancer using protein paint
.
Nat Genet
.
2016
;
48
(
1
):
4
-
6
.
23.
Miller
CA
,
McMichael
J
,
Dang
HX
, et al
.
Visualizing tumor evolution with the fish plot package for R
.
BMC Genomics
.
2016
;
17
(
1
):
880
.
24.
Jumper
J
,
Evans
R
,
Pritzel
A
, et al
.
Highly accurate protein structure prediction with AlphaFold
.
Nature
.
2021
;
596
(
7873
):
583
-
589
.
25.
Schymkowitz
J
,
Borg
J
,
Stricher
F
,
Nys
R
,
Rousseau
F
,
Serrano
L
.
The FoldX web server: an online force field
.
Nucleic Acids Res
.
2005
;
33
(
Web Server issue
):
W382
-
388
.
26.
Jabbour
EJ
,
Sasaki
K
,
Ravandi
F
, et al
.
Inotuzumab ozogamicin in combination with low-intensity chemotherapy (mini-HCVD) with or without blinatumomab versus standard intensive chemotherapy (HCVAD) as frontline therapy for older patients with Philadelphia chromosome-negative acute lymphoblastic leukemia: a propensity score analysis
.
Cancer
.
2019
;
125
(
15
):
2579
-
2586
.
27.
Paietta
E
,
Roberts
KG
,
Wang
V
, et al
.
Molecular classification improves risk assessment in adult BCR-ABL1-negative B-ALL
.
Blood
.
2021
;
138
(
11
):
948
-
958
.
28.
Haddox
CL
,
Mangaonkar
AA
,
Chen
D
, et al
.
Blinatumomab-induced lineage switch of B-ALL with t(4:11)(q21;q23) KMT2A/AFF1 into an aggressive AML: pre- and post-switch phenotypic, cytogenetic and molecular analysis
.
Blood Cancer J
.
2017
;
7
(
9
):
e607
.
29.
Wolfl
M
,
Rasche
M
,
Eyrich
M
,
Schmid
R
,
Reinhardt
D
,
Schlegel
PG
.
Spontaneous reversion of a lineage switch following an initial blinatumomab-induced ALL-to-AML switch in MLL-rearranged infant ALL
.
Blood Adv
.
2018
;
2
(
12
):
1382
-
1385
.
30.
Zoghbi
A
,
Zur Stadt
U
,
Winkler
B
,
Müller
I
,
Escherich
G
.
Lineage switch under blinatumomab treatment of relapsed common acute lymphoblastic leukemia without MLL rearrangement
.
Pediatr Blood Cancer
.
2017
;
64
(
11
).
31.
Ereño-Orbea
J
,
Sicard
T
,
Cui
H
, et al
.
Molecular basis of human CD22 function and therapeutic targeting
.
Nat Commun
.
2017
;
8
(
1
):
764
.
32.
Zhao
B
,
Rothenberg
E
,
Ramsden
DA
,
Lieber
MR
.
The molecular basis and disease relevance of non-homologous DNA end joining
.
Nat Rev Mol Cell Biol
.
2020
;
21
(
12
):
765
-
781
.
33.
Mitui
M
,
Nahas
SA
,
Du
LT
, et al
.
Functional and computational assessment of missense variants in the ataxia-telangiectasia mutated (ATM) gene: mutations with increased cancer risk
.
Hum Mutat
.
2009
;
30
(
1
):
12
-
21
.
34.
Bucher
N
,
Britten
CD
.
G2 checkpoint abrogation and checkpoint kinase-1 targeting in the treatment of cancer
.
Br J Cancer
.
2008
;
98
(
3
):
523
-
528
.
35.
Ray Chaudhuri
A
,
Callen
E
,
Ding
X
, et al
.
Replication fork stability confers chemoresistance in BRCA-deficient cells
.
Nature
.
2016
;
535
(
7612
):
382
-
387
.
36.
Motea
EA
,
Berdis
AJ
.
Terminal deoxynucleotidyl transferase: the story of a misguided DNA polymerase
.
Biochim Biophys Acta
.
2010
;
1804
(
5
):
1151
-
1166
.
37.
Zhang
H
,
Yan
J
,
Lu
Z
, et al
.
Deep sampling of gRNA in the human genome and deep-learning-informed prediction of gRNA activities
.
Cell Discov
.
2023
;
9
(
1
):
48
.
38.
Xiao
H
,
Wang
S
,
Tang
Y
, et al
.
Absence of terminal deoxynucleotidyl transferase expression in T-ALL/LBL accumulates chromosomal abnormalities to induce drug resistance
.
Int J Cancer
.
2023
;
152
(
11
):
2383
-
2395
.
You do not currently have access to this content.
Sign in via your Institution