ArticlePDF Available

ARTICLE Bispecific antibody simultaneously targeting PD1 and HER2 inhibits tumor growth via direct tumor cell killing in combination with PD1/PDL1 blockade and HER2 inhibition

Authors:

Abstract and Figures

Immune checkpoint blockade has shown significant clinical benefit in multiple cancer indications, but many patients are either refractory or become resistant to the treatment over time. HER2/neu oncogene overexpressed in invasive breast cancer patients associates with more aggressive diseases and poor prognosis. Anti-HER2 mAbs, such as trastuzumab, are currently the standard of care for HER2-overexpressing cancers, but the response rates are below 30% and patients generally suffer relapse within a year. In this study we developed a bispecific antibody (BsAb) simultaneously targeting both PD1 and HER2 in an attempt to combine HER2-targeted therapy with immune checkpoint blockade for treating HER2-positive solid tumors. The BsAb was constructed by fusing scFvs (anti-PD1) with the effector-functional Fc of an IgG (trastuzumab) via a flexible peptide linker. We showed that the BsAb bound to human HER2 and PD1 with high affinities (EC50 values were 0.2 and 0.14 nM, respectively), and exhibited potent antitumor activities in vitro and in vivo. Furthermore, we demonstrated that the BsAb exhibited both HER2 and PD1 blockade activities and was effective in killing HER2-positive tumor cells via antibody-dependent cellular cytotoxicity. In addition, the BsAb could crosslink HER2-positive tumor cells with T cells to form PD1 immunological synapses that directed tumor cell killing without the need of antigen presentation. Thus, the BsAb is a new promising approach for treating late-stage metastatic HER2-positive cancers.
The anti-HER2×PD1 BsAb exhibited synergistic killing effects on cancer cells in a PDL1 expression-independent manner ex vivo. a Human PBMCs were mixed with PDL1-overexpressing N87 tumor cells (N87-PDL1) in the presence of 2, 10, and 50 nM indicated antibodies (the N87-PDL1/PBMC pair). b Activated Jurkat T cells were mixed with parental N87 tumor cells in the presence of 2 and 9 nM indicated antibodies (the N87/T cell pair). With respect to the combination treatment comprising trastuzumab and 609A, equal molar amounts of the two mAbs (2, 10, and 50 nM each) were combined and added to the cells. The number of viable cells was measured as relative luminescence units (RLUs). The different treatment groups were as follows: N87-PDL1 (N87), N87-PDL1 (N87) tumor cells only; N87-PDL1 +PBMCs (N87+T cells), N87-PDL1 cells mixed with PBMCs (N87 cells mixed with T cells) in the absence of antibodies; Anti-PD1 mAb+PBMCs (Anti-PD1 mAb+T cells), N87-PDL1 cells mixed with PBMCs (N87 cells mixed with T cells), and the anti-PD1 mAb, 609A; Trastuzumab+PBMCs (Trastuzumab+T cells), N87-PDL1 cells mixed with PBMCs (N87 cells mixed with T cells) and trastuzumab; Trastuzumab+anti-PD1 mAb +PBMCs (Trastuzumab+anti-PD1 mAb+T cells), combination of trastuzumab and 609A added to N87-PDL1 cells in the presence of PBMCs (N87 cells in the presence of T cells); and Anti-HER2×PD1 BsAb+PBMCs (Anti-HER2×PD1 BsAb+T cells), the BsAb added to N87-PDL1 in the presence of PBMCs (N87 cells in the presence of T cells). *P < 0.05, **P < 0.01, and ****P < 0.0001 by two-way ANOVA. c PD1-overexpressing Jurkat T cells prelabeled with an Alexa Fluor 647-conjugated anti-PD1 mAb (Sinobiological, Cat#MM18) were mixed with N87 cells prelabelled with an Alexa Fluor 546-conjugated anti-HER2 mAb (19H6-Hu) in the presence of the BsAb (left), the mixture of trastuzumab and 609A (center), the BsAb plus 609A (right). The group in red (pseudo-color) (upper right corner) shows cells that have dual emission signals (MFI > 1 × 10 4 for FITC-A and MFI > 3 × 10 4 for APC-A), meaning that the two types of cells were associated together. The group in green (pseudocolor) (upper left corner) shows T cells labeled with the Alexa Fluor 647-conjugated anti-PD1 mAb (MFI > 3 × 10 4 for APC-A). The group in purple (pseudo-color) (lower right corner) shows N87 cells labeled with the Alexa Fluor 546-conjugated anti-HER2 mAb (MFI > 1 × 10 4 for FITC-A). d Immunofluorescence microscopy showing costaining of PD1 (green) on T cells and HER2 (red) on N87 tumor cells in samples treated with the BsAb or a mixture of the anti-PD1 mAb (609A) plus trastuzumab. Note that one tumor cell was ligated with two T cells, while a single T cell was in contact with two tumor cells in the presence of the BsAb. The combination of 609A and trastuzumab failed to induce PD1 synapse formation with T cells. The white arrows denote T cells (DIC channel), for which PD1 staining (green channel) were barely seen owing to the rearrangement of PD1 during synapse formation.
… 
Content may be subject to copyright.
ARTICLE OPEN
Bispecic antibody simultaneously targeting PD1 and HER2
inhibits tumor growth via direct tumor cell killing in
combination with PD1/PDL1 blockade and HER2 inhibition
Chang-ling Gu
1
, Hai-xia Zhu
1
, Lan Deng
1
, Xiao-qing Meng
1
, Kai Li
1
, Wei Xu
1
, Le Zhao
1
, Yue-qin Liu
1
, Zhen-ping Zhu
1
and
Hao-min Huang
1
Immune checkpoint blockade has shown signicant clinical benet in multiple cancer indications, but many patients are either
refractory or become resistant to the treatment over time. HER2/neu oncogene overexpressed in invasive breast cancer patients
associates with more aggressive diseases and poor prognosis. Anti-HER2 mAbs, such as trastuzumab, are currently the standard of
care for HER2-overexpressing cancers, but the response rates are below 30% and patients generally suffer relapse within a year. In
this study we developed a bispecic antibody (BsAb) simultaneously targeting both PD1 and HER2 in an attempt to combine HER2-
targeted therapy with immune checkpoint blockade for treating HER2-positive solid tumors. The BsAb was constructed by fusing
scFvs (anti-PD1) with the effector-functional Fc of an IgG (trastuzumab) via a exible peptide linker. We showed that the BsAb
bound to human HER2 and PD1 with high afnities (EC
50
values were 0.2 and 0.14 nM, respectively), and exhibited potent
antitumor activities in vitro and in vivo. Furthermore, we demonstrated that the BsAb exhibited both HER2 and PD1 blockade
activities and was effective in killing HER2-positive tumor cells via antibody-dependent cellular cytotoxicity. In addition, the BsAb
could crosslink HER2-positive tumor cells with T cells to form PD1 immunological synapses that directed tumor cell killing without
the need of antigen presentation. Thus, the BsAb is a new promising approach for treating late-stage metastatic HER2-positive
cancers.
Keywords: HER2; Trastuzumab; PD1 blockade; bispecic antibody; antibody-dependent cellular cytotoxicity (ADCC); PD1
immunological synapse
Acta Pharmacologica Sinica (2021) 0:19; https://doi.org/10.1038/s41401-021-00683-8
INTRODUCTION
The HER2/neu oncogene encodes an epidermal growth factor
receptor (EGFR)-related receptor, HER2, an orphan receptor whose
ligand has not yet been identied. HER2 dimerizes with other
HER2 family members to activate downstream signaling pathways
and thus plays important roles in cell growth [1,2]. It has been
shown that HER2 is overexpressed in 20%25% of invasive breast
cancer patients and its overexpression is associated with more
aggressive diseases and poor prognosis [3,4].
Trastuzumab (Herceptin), a recombinant humanized monoclonal
antibody (mAb) against HER2 developed by Roche/Genentech, has
become the standard of care for patients with HER2-positive breast
cancer over decades [57]. However, the objective response rates to
trastuzumab monotherapy range between 12% and 34% and the
majority of patients who initially responded to trastuzumab generally
develop resistance or relapse after a year [8]. To this end, alternative
anti-HER2 antibody-based therapeutics including antibody-drug
conjugates, such as T-DM1 and DS8201s, and bispecic antibodies
(BsAbs) are being actively developed and have shown signicant
clinical benets in several cancer indications [912].
Programmed death-1 (PD1) is an inhibitory receptor expressed
in T and B cells as well as myeloid-derived cells [1316]. Binding of
PD1 to its ligand PDL1 (CD274, B7-H1) inhibits T-cell proliferation
and cytokine production and, thus, turns them into a state of
exhaustion [17,18]. Anti-PD1 mAbs capable of blocking PD1 and
PDL1 have shown promising antitumor activity in a broad
spectrum of cancer types in preclinical studies and clinical trials
[1925]. Nevertheless, only a fraction of patents (approximately
20%) achieved durable clinical responses after anti-PD1 antibody
monotherapy, while most patents either had no responses at all or
exhibited transient responses [2630].
BsAbs are antibodies comprising two distinct antigen-
targeting domains and can be used in place of combination of
two mAbs. BsAbs have been extensively studied over years for
therapeutic use, because they can provide therapeutic benets
that mAbs cannot [3135]. A BsAb can bridge its two target
antigens and bring them into close proximity [36]. A BsAb can
retarget the effector cells, e.g., T cells, against tumor cells by
simultaneously binding to cell surface antigens expressed from
both cells and thus signicantly activate the antitumor activities
of effector cells [3739]. A BsAb can also ligate two different
receptors on the same cell and change intracellular downstream
signaling [40]. Using antibody engineering technology to create
a BsAb targeting HER2 and PD1,weaimedtosynergizethe
Received: 19 December 2020 Accepted: 14 April 2021
1
Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., 3SBio Inc. Company, Shanghai 201203, China
Correspondence: Zhen-ping Zhu (zhuzhenping@3sbio.com) or Hao-min Huang (huanghaomin@3s-guojian.com)
www.nature.com/aps
©The Author(s) 2021
1234567890();,:
antitumor activities rendered by HER2 blockade and PD1/PDL1
blockade.
We report the creation of a BsAb by combining an anti-HER2
antibody IgG and a scFv from an anti-PD1 antibody (609A). The
BsAb retained its binding specicity for its original targets and was
able to direct T cells to engage HER2-overexpressing tumor cells,
resulting in PD1 synapse formation. In addition to the ability to
block HER2 signaling and induce antibody-dependent cellular
cytotoxicity (ADCC), the BsAb also activated antitumor immunity
through PD1/PDL1 blockade in vivo. Importantly, the BsAb
exhibited superior tumor cell killing activity in the presence of
peripheral blood mononuclear cells (PBMCs) or activated T cells
relative to combination of the two parental mAbs. Thus, our anti-
HER2×anti-PD1 BsAb represents a new strategy to enhance
antibody therapeutic efcacy through tumor-targeted immune
checkpoint blockade.
MATERIALS AND METHODS
Cell culture
The cell lines used in this study were obtained from American
Type Culture Collection (ATCC; Manassas, VA, USA) unless
otherwise indicated. The cells were cultured at 37 °C with 5%
CO
2
. Cells for protein expression: HEK293E cells were cultured in
FreeStyle 293 medium (Life Technologies, Rockville, MD, USA).
Cells for bioassays: NCI-N87 or BT474 cells were cultured in RPMI-
1640 supplemented with 10% FBS. PD1/CHO-S cells were cultured
in CD Forti-CHO supplemented with 1% Glutamax and 0.4% anti-
clumping agent. PD-L1 aAPC/CHO-K1 cells (Promega, Cat#J1252,
Madison, WI, USA) were cultured in 90% Hams F12 (Gibco
Cat#11765-054) with 10% FBS, 200 μg/mL hygromycin B (Gibco,
Cat#10687-010, Shanghai, China) and 200 μg/mL G418 sulfate
solution (Promega, Cat#V8091). PD1 effector cells (Promega,
Cat#J1250) were cultured in 90% RPMI-1640, supplemented with
10% FBS, 200 μg/mL hygromycin B, 500 μg/mL G418 sulfate
solution, 1% sodium pyruvate (Promega, Cat#11360070), and 1%
MEM nonessential amino acids (Gibco, Cat#11140050). Cells for
ADCC: NK92-CD16 cells (ATCC, Cat#pta8837) were cultured in
αMEM medium without nucleosides and with 2 mM glutamine,
0.1 mM 2-mercaptoethanol, 0.2 mM inositol, 0.02 mM folic acid,
12.5% FBS, 12.5% Horse Serum (ATCC, Cat#302040), and 100 units/
mL human recombinant IL-2. Cells for animal study: MC38 cells
(Jennio Biotech, a mouse colon cancer cell line, Guangzhou, China)
were cultured in DMEM with 10% FBS; JIMT-1 (Cobioer,
Cat#CBP60378, Nanjing, China), a breast cancer cell line, were
cultured in DMEM medium supplemented with 10% FBS. PBMCs
(Cat#SLB-HP050A) and CD3
+
beads-isolated T cells (Cat#SLB-
CD3T-10AN) were purchased from Sailybio (Shanghai, China) and
used when applicable.
Protein expression and purication
Constructs expressing the anti-HER2×PD1 BsAb variants were
generated using the pTT5 vector (NRC Biotechnology Research
Institute, Montreal, QC, Canada). The expression vectors were
transiently transfected into HEK293E cells using 1 μg/mL 25 kDa
linear PEI (Polysciences, Inc.). One day after transfection, valproic
acid (Sigma) was added to the cell culture at a nal concentration
of 3 mM. On day 2 after transfection, medium comprising 10%
GlutaMAX, 10% 400 g/L glucose, and 80% freestyle 293 medium
was added to the cell culture to 10% of the total volume.
Conditioned medium was collected 56 days after transient
transfection.
BsAbs in the culture medium were puried by MabSelect SuRe
(GE) afnity columns using an Akta Avant 25 fast protein liquid
chromatography purication system. After equilibrating the
column with buffer A (25 mM sodium phosphate, 150 mM sodium
chloride, pH 7.4), the culture media containing BsAbs was loaded
into the column, which was then eluted with buffer B (100 mM
sodium citrate, pH 3.5) to collect the desired proteins. The eluted
proteins were neutralized with 1 M Tris-HCl at pH 9.0.
Flow cytometric analysis of BT474 cells and PD1/CHO cells
To measure the binding afnity of the anti-HER2×PD1 BsAb for
HER2-overexpressing cells, BT474 cells (1 × 10
5
/well in 96-well
plate) were incubated with three-fold serial dilutions of the BsAb
ranging from 3.3 pM to 200 nM in 200 μL PBS at 4 °C for 1 h. Cells
were washed three times with PBS and then incubated with FITC-
conjugated goat anti-human IgG (Jackson, Cat.#109-095-003) at
4 °C for 1 h. The cells were washed and resuspended in 200 μL PBS
and were analyzed on FACS (BECKMAN, Cytoex).
To measure the binding afnity for PD1-overexpressing cells,
PD1/CHO cells were used instead.
Bridging ELISA
PD1-ECD proteins (in-house, 200 ng/mL) were coated in 96-well
plates (Thermo Fisher, Cat#439454) at 4 °C overnight. The plates
were washed with PBST (PBS containing 0.05% Tween-20),
blocked for 1 h with PBS containing 2% BSA and incubated with
three-fold serial dilutions of antibodies for another hour at 37 °C.
The plates were then washed three times and incubated with
2 µg/mL His-tagged HER2-ECD for 1 h at 37 °C. After washing, an
HRP-conjugated anti-6×HisTag mAb (Invitrogen, Cat#MA1-21315-
HRP) was added and incubated for 1 h at 37 °C. The plates were
washed and the reaction was developed with TMB substrates. The
plates were then read on a SpectraMax 190 reader (Molecular
Devices) at 450 nm.
Proliferation inhibition assay
BT474 cells, a HER2-overexpressing human breast cancer cell line,
were seeded at 5000 cells/well in a 96-well culture plate (Costar,
Cat#3599) supplemented with RPMI-1640+10% FBS, and incu-
bated overnight at 37 °C with 5% CO
2
. The next day, the cells were
incubated with serially diluted anti-HER2×PD1 BsAb or trastuzu-
mab ranging from 8 pM to 150 nM in a nal volume of 200 μL/well.
The plates were incubated at 37 °C for 67 days and viability was
quantitated with Cell counting Kit-8 (Dojindo, Cat#CK04). The
plates were then read on a SpectraMax 190 reader (Molecular
Devices) at 450 nm.
PD1/PDL1 blockade bioassay
The assay was carried out following the manufacturers instruction
(Promega, Cat#J1250). Briey, PD-L1 aAPC/CHO-K1 cells were
seeded at 4 × 10
4
cells/well at 100 μL in white 96-well plates
followed by cultured overnight in a 37 °C incubator with 5% CO
2
.
The next day, the supernatant was discarded and the PD-L1 aAPC/
CHO-K1 cells were incubated with three-fold serial dilutions of the
BsAb ranging from 0.04 to 300 nM and PD-1 effector cells (5 × 10
4
/
well) in 80 μL assay buffer (99% RPMI-1640 supplemented with L-
glutamine +1% FBS) for 6 h. Bio-GloReagent (80 μL) was added
to each well and the bottom was sealed with opaque lm. The
plate was incubated at ambient temperature for 530 min and
then luminescence was measured using a SpectraMax i3x. The
expression of luciferases under the control of NFAT transcriptional
response elements was measured as a readout in response to
PD1/PDL1 blockade.
ADCC assay
BT474 cells were seeded in 96-well at-bottom plates at a density
of 1 × 10
4
cells/well at 50 μL in RPMI-1640 supplemented with 5%
FBS. NK92a cells (4 × 10
4
/well, Cat#pta8837) were added to each
well in the presence of serially diluted anti-HER2×PD1 BsAbs and
control antibodies for a nal reaction volume of 150 μL, and the
plates were incubated for 3 h at 37 °C and 5% CO
2
. Three hours
later, 100 μL supernatant was transferred to new plates. To
monitor cell lysis, 50 μL LDH substrate was added to each well
containing the supernatant and the plates were incubated at
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
2
Acta Pharmacologica Sinica (2021) 0:1– 9
room temperature for 15 min. The plates were then read at 490
nm on a SpectraMax 190 reader (Molecular Devices). The % lysis
was converted from OD values according to the following formula:
(OD
sample
OD
T
OD
nk
)/OD
lysis
× 100%. The EC
50
was calculated
using GraphPad Prism 7 software (GraphPad Software). In the case
of T cells, pre-activated T cells with anti-CD3 antibody were used
instead of tumor cells.
Tumor cell killing assay
PDL1-overexpressing N87 cells (N87-PDL1) were diluted to 1 × 10
5
cells/mL in RPMI-1640 supplemented with 1% FBS. N87-PDL1 cells
(100 μL/well) were seeded into the wells of 96-well plates and
incubated overnight at 37 °C and 5% CO
2
. The next day, 2, 10, and
50 nM of antibodies [the BsAb, trastuzumab, an anti-PD1 mAb
(609A) and trastuzumab+609A] and controls were added to each
well containing 5000 PBMCs for a reaction volume of 200 μL/well,
and the plates were incubated at 37 °C and 5% CO
2
for 4 days.
Cell-Titer Glo reagent (50 μL/well) was added to the plates
followed by incubation at room temperature for 510 min. The
plates were then read on an MD SpectraMax i3 for luminescence
at 500 ms.
In the case of activated T-cell killing assays, NCI-N87 cells were
seeded in 96-well plates at 1 × 10
4
cells/well. The next day, 2 and 9
nM of antibodies and controls were added to each well containing
5000 activated T cells for a reaction volume of 200 μL/well, and the
plates were incubated at 37 °C and 5% CO
2
for 7 days. The plates
were then read on an MD SpectraMax i3 for luminescence.
Immunouorescence assay
PD1-overexpressing Jurkat T cells were stimulated to enhance PD1
expression. The activated T cells were labeled with 200 nM Alexa
Fluor 488 (488)-conjugated anti-HER2×PD1 BsAb (T1) or 488-
conjugated anti-PD1 mAb (609A) (T2) for 1 h at room temperature
respectively. N87 tumor cells were labeled with 100 nM Alexa
Fluor 546 (546)-conjugated anti-HER2 mAb (19H6-Hu) in the
presence (A1) or absence (A2) of trastuzumab for 1 h at room
temperature, respectively. The 488-labeled T cells were then co-
cultured with 546-labeled N87 cells with the following formats T1
+A2 or T2+A1 in PBS for 30 min at room temperature followed by
transfer into 6-well plates pre-coated with poly-lysine prior to
xation with 4% paraformaldehyde. Images were visualized with
an LCAch N 40×/0.55 PhP objective (IX53, Olympus).
Xenografted and syngeneic tumor models
Animal care and in vivo experiments were approved by the
institutional IACUC of Sunshine Guojian Pharmaceutical (Shang-
hai) Co. Ltd. and performed under approved protocols (approval
code for NCI-N87 xenograft model: AS-2018-058; for
MC38 syngeneic model: AS-2019-006). NCI-N87 xenograft tumor
models were established with 6-week-old female Balb/c nude
mice (purchased from Charles River Laboratories) by subcuta-
neous injection of 8 × 10
6
NCI-N87 cells mixed with 50% Matrigel.
MC38 syngeneic tumor models were established in 1820 g
female PD-1 humanized mice (B6-Pdcd1
em1(hPdcd1)Vst
/Vst, Vital star
Biotech) by subcutaneous injection of 1 × 10
6
MC38 cells. When
tumors reached a volume of approximately 150 mm
3
, the animals
were randomly divided into groups and i.p. injected twice a week.
Tumor volume was measured twice per week and calculated using
the formula V=LW
2
/2 (where V=volume, L=length, and W=
width).
In the case of the JIMT-1 xenograft model, JIMT-1 tumors were
established in M-NSG mice (NOD.Cg-Prkdc
scid
Il2rg
em1
/Smoc)
(Model Organism, Cat# NM-NSG-001) by subcutaneous injection
of 8 × 10
6
JIMT-1 cells mixed with 50% Matrigel. When tumors
reached a volume of approximately 200500 mm
3
,3×10
6
PBMCs
in suspension were intraperitoneally inoculated, and then the
animals were randomly divided into groups and treated as
described above.
Statistical analysis
All numerical data, except for mouse xenograft data, are
presented as the mean ± standard deviation. Mouse xenograft
data are presented as the mean ± standard error. Statistical
analysis was performed with GraphPad Prism 7 software and Excel.
Pvalues were calculated using a two-way ANOVA multiple
comparison test. In all tests, differences with Pvalues < 0.05 (*)
were considered to be statistically signicant.
RESULTS
Construction and production of the anti-HER2×anti-PD1 BsAb
The anti-HER2 antibody, trastuzumab, and the anti-PD1 antibody,
SSGJ-609A (609A), were utilized as the building blocks to construct
the anti-HER2×anti-PD1 BsAb via the IgG-scFv or scFv-IgG fusion
format [4143]. In this format, the scFv of one antibody was fused
via a exible peptide linker [(GGGGS)n,n=05] to the N- or C-
terminus of the heavy chain of the other antibody. Various
constructs were examined for their expression levels in transient
mammalian culture and their bioactivities in terms of binding to
both HER2 and PD1. When the scFv of trastuzumab was fused to
either the N- or C-terminus of the heavy chain of the IgG scaffold
of 609A, it showed a signicantly reduced binding afnity for
BT474 cells (a HER2-overexpressing breast cancer cell line), and
was much less potent in inhibiting proliferation of the tumor cells,
compared to trastuzumab (data not shown). We next used
trastuzumab as the IgG scaffold and fused it with the scFv of
609A. Between the two IgG/scFv fusion orientations examined, the
BsAb constructed with the 609A scFv fused to the N-terminus of
trastuzumab showed ~5-fold lower binding afnity for BT474 cells
than did the BsAb with the 609A scFv fused to the C-terminus
(Supplementary Fig. S1). Thus, the BsAb with the two copies of
609A scFvs fused to the C-terminus of trastuzumab, namely anti-
HER2×PD1 BsAb, was selected for further characterization (Fig. 1a).
As demonstrated by SDS-PAGE, SEC-HPLC, and differential
scanning calorimetry, the anti-HER2×PD1 BsAb exhibited favor-
able chemophysical properties as a drug candidate (Fig. 1bd).
The anti-HER2×PD1 BsAb simultaneously bound to HER2 and PD1
comparable to the parent monoclonal antibodies
The anti-HER2×PD1 BsAb dose-dependently bound to HER2 and
PD1 as shown by ELISA. The EC
50
(the antibody concentration
required for 50% of maximum binding) of the BsAb for HER2 was
0.2 nM. This was comparable to that of trastuzumab, which was
0.22 nM. Similarly, the EC
50
of the BsAb for human PD1 was 0.14
nM, which was comparable to the EC
50
of the parental anti-PD1
mAb, 609A (0.15 nM, Fig. 2a, b). The BsAb also bound efciently to
the receptors on the cell surface as shown by FACS analysis. The
EC
50
of the BsAb binding to BT474 cells was 1.64 nM, comparable
to trastuzumab, which bound to the same cells with an EC
50
of
1.56 nM. The EC
50
of the BsAb binding to PD1-overexpressing CHO
cells was 1.78 nM, which was comparable to the EC
50
of the anti-
PD1 mAb, 609A (1.62 nM, Fig. 2c, d). In addition to binding to PD1-
overexpressing cell lines, we also tested the ability of the BsAb to
bind to primary T cells. As expected, the BsAb was indeed capable
of binding to activated primary T cells (Supplementary Fig. S2). To
conrm that the BsAb can simultaneously bind to its two targets, a
bridging ELISA was performed and the results showed that the
BsAb was capable of crosslinking HER2 and PD1 with an EC
50
of
0.24 nM, whereas the crosslinking activity was not achieved with
the parental mAbs, trastuzumab and 609A (Fig. 2e).
The anti-HER2×PD1 BsAb retained the full biological activities of
the respective parental antibodies in cell-based assays
In tumor cell growth assays, the BsAb effectively inhibited the
proliferation of HER2-overexpressing BT474 cells at an IC
50
(the
antibody concentration inhibiting 50% of cell proliferation) of 0.59
nM on par with that of trastuzumab whose IC
50
was 0.5 nM
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
3
Acta Pharmacologica Sinica (2021) 0:1 9
-4 -2 0 2 4
0
1
2
3
HER2 binding ELISA
log[c on ce ntra tion ](n M )
OD
450
anti-H ER 2 × PD 1 Bs A b
tras tu zu m a b
BT474 binding FACS
log[concentration](nM)
MFI
-4 -2 0 2
0
20000
40000
60000 anti-HER2×PD1 BsAb
trastuzu m ab
-4 -2 0 2 4
0.0
0.5
1.0
1.5
2.0
2.5
PD1 binding ELISA
log[concentration](nM)
OD
450
anti-HER2×PD1 BsAb
609A
log[concentration](nM)
MFI
-2 0 2 4
0
5000
10000
15000 anti-HER2×PD1 BsAb
609A
PD-1/CHO binding FACS
-4 -2 0 2
0.0
0.5
1.0
1.5
2.0
2.5
bridging ELISA
log[concentration](nM)
OD450
anti-HER2×PD1 BsAb
trastuzu m ab
609A
ab c
de
Fig. 2 The anti-HER2×PD1 BsAb simultaneously bound to PD-1 and HER2. a The binding afnity of the BsAb for HER2 was measured by ELISA.
Trastuzumab was used as the positive control. bThe binding afnity of the BsAb for PD1 was measured by ELISA and compared to that of the
parental anti-PD1 mAb, 609A. cThe ability of the BsAb to bind to BT474, a HER2-overexpressing cancer cell line, was measured by FACS and
compared to that of trastuzumab. dThe ability of the BsAb to bind to PD1-overexpressing CHO cells was measured by FACS and compared to that
of the parental anti-PD1 mAb, 609A. eA bridging ELISA was set up such that PD1 proteins were coated on the plates followed by the sequential
addition of the indicated antibodies and His-tagged HER2 proteins. Anti-6×HisTag monoclonal antibody-HRP was added to visualize the positive
binders. The results conrm that the BsAb is capable of simultaneously crosslinking its two targets, HER2 and PD1.
ab
cd
anti-PD1 scFv
Trastuzumab
250
36
28
17
10
55
72
95
130
12M3 4
Fig. 1 The structure and properties of the anti-HER2×PD1 BsAb. a Schematics of the anti-HER2×PD1 BsAb structure. bSDS-PAGE showing
nonreduced and reduced anti-HER2×PD1 BsAb. Lane 1: nonreduced BsAb; Lane 2: reduced BsAb; Lane 3: nonreduced trastuzumab; Lane 4:
reduced trastuzumab; M: Molecular weight markers. cSEC chromatogram showing that the BsAb puried by a single-step protein A afnity
column had over 95% monomeric species. dDifferential scanning calorimetry (DSC) of the anti-HER2×PD1 BsAb showing that the antibody
has a T
onset
(the temperature at onset of melting) of 52.5 °C and T
m
1/2/3 (melting temperatures) of 59.2 °C/68.4 °C/ 83.5 °C, respectively.
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
4
Acta Pharmacologica Sinica (2021) 0:1 9
(Fig. 3a). Similarly, the BsAb potently blocked PD1/PDL1 cell
signaling with an IC
50
of 3.28 nM, which was comparable to that of
609A (0.89 nM, Fig. 3b). Thus, the BsAb retained its ability to inhibit
HER2-overexpressing tumor cell growth via HER2 blockade and to
activate T cells via PD1 blockade.
The anti-HER2×PD1 BsAb exhibited ADCC toward HER2-
overexpressing tumor cells but not T cells
ADCC plays an important role in trastuzumab-mediated tumor cell
killing. Thus, we tested the BsAb for ADCC toward cancer cells and
T cells, as we would like to conrm whether the scFvs (anti-PD1)
fused to the effector-functional Fc of an IgG (trastuzumab) by
linkers still possessed ADCC activity. The BsAb exhibited strong
ADCC toward BT474 tumor cells, comparable to that of
trastuzumab (Fig. 4a). By contrast, no ADCC toward T cells could
be detected, while a control antibody targeting major histocom-
patibility complex (MHC) I of T cells exhibited potent ADCC toward
T cells [44] (Fig. 4b). This result is in line with the previous nding
where a BsAb with anti-PD1 scFvs fused to the N-terminus of
cetuximab showed no ADCC toward T cells [39]. This suggests that
proper spacing between the cell surface receptors and the Fc
region of an IgG molecule or a favorable protein conguration as a
whole, such as an intact IgG format, might be a critical
determinant for initiating ADCC.
The anti-HER2×PD1 BsAb exhibits synergistic killing of cancer cells
in a PDL1 expression-independent manner ex vivo
Given that the BsAb can crosslink PD1 on T cells and HER2 on
tumor cells, we hypothesized that the BsAb may enhance tumor
cell killing by bringing T cells into the proximity of tumor cells and
potentially inducing PD1 synapse formation as seen in our
previous study [39]. We stably transfected NCI-N87 cells, a HER2-
overexpressing gastric cancer cell line that does not express PDL1
when cultured in vitro (data not shown), with a PDL1 construct to
obtain a PDL1/HER2 double-overexpressing cancer cell line (N87-
PDL1) that could potentially respond to trastuzumab and anti-PD1
mAbs. Addition of various concentrations (250 nM) of trastuzu-
mab to N87-PDL1 cells in the presence of human PBMCs dose-
dependently reduced the number of viable cancer cells relative to
antibody-free controls (Fig. 5a). Combinations of trastuzumab and
the anti-PD1 mAb, 609A, showed a killing effect similar to that of
trastuzumab alone. Importantly, treating cancer cells with the
BsAb resulted in a superior reduction of viable cancer cells
compared to that of combination of trastuzumab with 609A (P<
0.0001; Fig. 5a).
Interestingly, 609A alone did not enhance PBMC-mediated N87-
PDL1 cell killing, even though PDL1 was overexpressed by the
tumor cells. To conrm that PDL1 on tumor cells was not
responsible for the lack of killing and that the enhanced killing
ab
-2 -1 0 1 2 3
0
10000
20000
30000
40000
PD1/PDL-1blockadebioassay
log[concentration](nM)
Lum(RLU)
IgG1
anti-HER2×PD1 BsAb
609A
log[concentration](nM)
OD
450
-3-2-10123
0.0
0.5
1.0
1.5 anti-HER 2×PD1 BsAb
trastuz um ab
B T 4 7 4 in h ib itio n
Fig. 3 The anti-HER2×PD1 BsAb inhibited the proliferation of HER2-overexpressing tumor cells and blocked the PD1/PDL1 interaction in
cell-based bioassays. a The BsAb inhibited the proliferation of HER2-overexpressing BT474 cancer cells in a dose-dependent manner similar
to trastuzumab. bThe ability of the BsAb to block PD1/PDL1 signaling was measured and compared to that of the parental anti-PD1 mAb,
609A, using a PD1/PDL1 blockade cell-based assay, in which the expression of luciferases were monitored under the control of nuclear factor
of activated T cells (NFAT ) response elements in response to blockade of PD1/PDL1 signaling (Promega Cat#J1250). A nonspecic IgG1 was
used as the negative control.
-3 -2 -1 0 1 2
-20
0
20
40
60
log [co nc en tratio n]( nM )
Lysis%
trastu zum ab
anti-HER2×PD1 BsAb
ADC C towards BT474 cells
-3 -2 -1 0 1 2
0
20
40
60
log[concentration](nM)
Lysis%
MHCIIgG1
trastu zum ab
anti-HER2×PD1 BsAb
ADCC towards T Cells
ab
Fig. 4 The anti-HER2×PD1 BsAb retained ADCC toward tumor cells but not T cells. a The BsAb exhibited a potency in lysing tumor cells
similar to that of trastuzumab in the ADCC assay. bThe BsAb and trastuzumab failed to mediate ADCC toward T cells, whereas an anti-MHC1
IgG1 antibody showed strong potency in lysing T cells in a dose-dependent manner [44].
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
5
Acta Pharmacologica Sinica (2021) 0:1 9
mediated by the BsAb was not simply due to PD1/PDL1 blockade-
induced T-cell activation, we examined the killing of parental N87
cells that did not express PDL1 with activated Jurkat T cells in the
presence of various antibodies. Consistent with the PBMC results,
the BsAb effectively killed more tumor cells than did either
trastuzumab alone, or the combination of trastuzumab with 609A.
Anti-PD1 mAb alone, again, failed to induce measurable tumor cell
killing in the absence of PDL1 on tumor cells (Fig. 5b). This nding
suggests that the crosslinking of T cells with cancer cells by the
BsAb, in contrast to the effects of the anti-PD1 mAb alone, might
be sufcient to allow T cells to recognize and initiate the killing of
tumors without the need for presentation of tumor-specic
antigens to T cells.
We next demonstrated by FACS that the physical association
between HER2-overexpressing tumor cells and T cells was indeed
established specically by the BsAb (Fig. 5c and Supplementary
Fig. S3). In addition, immunouorescence microscopy showed that
the BsAb bridged T cells with tumor cells and led to formation of
PD1 synapses [45,46]. It is possible that T cell activity could be
signicantly further enhanced by PD1 synapses, since multiple
PD1 synapses could be simultaneously formed on a single T cell.
By contrast, no apparent tumorT cell associations were observed
when the cells were incubated with a mixture of trastuzumab and
609A (Fig. 5d). Taken together, the above ndings suggest that
the physical ligation of cancer cells with T cells by the BsAb might
enhance T cell-mediated antitumor effects in a way that cannot be
achieved by simply mixing two mAbs together.
The anti-HER2×PD1 BsAb exhibits potent antitumor activities in
both xenograft and syngeneic animal models
We next evaluated the antitumor effects of each of the two arms
of the BsAb in vivo. First, we measured the efcacy of the BsAb
for HER2 blockade in an N87 xenograft mouse model. The BsAb
suppressed 75% of N87 tumor growth at a dose of 20 mg/kg on
day 28 after treatment. The antitumor efcacy is comparable to
that of trastuzumab which inhibited 80% of tumor growth at an
equal molar dose of 15 mg/kg (Fig. 6aandSupplementary
Fig. S5d).
Second, we analyzed the antitumor activity of the BsAb for
PD1 blockade in an MC38 tumor cell line-derived human
PD1 transgenic mouse model. The BsAb effectively inhibited
~85% of tumor growth at a dose of 13 mg/kg on day 17 post
abc
d
PD1
HER2
Anti-HER2xPD1 BsAb Trastuzumab+609A BsAb+609A
Dual color cells: 23.31% Dual color cells: 1.26% Dual color cells: 5.00%
DIC PD1 HER2 Merge
Anti-HER2xPD1
BsAb
609A
+
Trastuzumab
50nM
10nM
2nM
600,000
800,000
1,000,000
lum(R LU )
anti-HER2×PD1BsAb+pbmc
trastuzuma b+ anti-PD1 m A b + pb m c
trastuzumab+pbmc
anti-PD1mAb +pbm c
N87-PDL1+pbmc
N87-PDL1
N87-PDL1 inhibition
**** ****
****
9nM
2nM
400,000
600,000
800,000
lum(RLU)
anti-HER2×PD1BsAb + Tcell
trastuzumab+anti-PD1 mAb + T cell
trastuzum ab + T cell
anti-PD1mAb + T cell
N87+ Tcell
N87
N87 inhibition
*
**
Fig. 5 The anti-HER2×PD1 BsAb exhibited synergistic killing effects on cancer cells in a PDL1 expression-independent manner ex vivo. a
Human PBMCs were mixed with PDL1-overexpressing N87 tumor cells (N87-PDL1) in the presence of 2, 10, and 50 nM indicated antibodies
(the N87-PDL1/PBMC pair). bActivated Jurkat T cells were mixed with parental N87 tumor cells in the presence of 2 and 9 nM indicated
antibodies (the N87/T cell pair). With respect to the combination treatment comprising trastuzumab and 609A, equal molar amounts of
the two mAbs (2, 10, and 50 nM each) were combined and added to the cells. The number of viable cells was measured as relative
luminescence units (RLUs). The different treatment groups were as follows: N87-PDL1 (N87), N87-PDL1 (N87) tumor cells only; N87-PDL1
+PBMCs (N87+T cells), N87-PDL1 cells mixed with PBMCs (N87 cells mixed with T cells) in the absence of antibodies; Anti-PD1 mAb+PBMCs
(Anti-PD1 mAb+T cells), N87-PDL1 cells mixed with PBMCs (N87 cells mixed with T cells), and the anti-PD1 mAb, 609A; Trastuzumab+PBMCs
(Trastuzumab+T cells), N87-PDL1 cells mixed with PBMCs (N87 cells mixed with T cells) and trastuzumab; Trastuzumab+anti-PD1 mAb
+PBMCs (Trastuzumab+anti-PD1 mAb+T cells), combination of trastuzumab and 609A added to N87-PDL1 cells in the presence of PBMCs
(N87 cells in the presence of T cells); and Anti-HER2×PD1 BsAb+PBMCs (Anti-HER2×PD1 BsAb+T cells), the BsAb added to N87-PDL1 in the
presence of PBMCs (N87 cells in the presence of T cells). *P< 0.05, **P< 0.01, and ****P< 0.0001 by two-way ANOVA. cPD1-overexpressing
Jurkat T cells prelabeled with an Alexa Fluor 647-conjugated anti-PD1 mAb (Sinobiological, Cat#MM18) were mixed with N87 cells prelabelled
with an Alexa Fluor 546-conjugated anti-HER2 mAb (19H6-Hu) in the presence of the BsAb (left), the mixture of trastuzumab and 609A
(center), the BsAb plus 609A (right). The group in red (pseudo-color) (upper right corner) shows cells that have dual emission signals (MFI >
1×10
4
for FITC-A and MFI > 3 × 10
4
for APC-A), meaning that the two types of cells were associated together. The group in green (pseudo-
color) (upper left corner) shows T cells labeled with the Alexa Fluor 647-conjugated anti-PD1 mAb (MFI > 3 × 10
4
for APC-A). The group in
purple (pseudo-color) (lower right corner) shows N87 cells labeled with the Alexa Fluor 546-conjugated anti-HER2 mAb (MFI > 1 × 10
4
for FITC-
A). dImmunouorescence microscopy showing costaining of PD1 (green) on T cells and HER2 (red) on N87 tumor cells in samples treated with
the BsAb or a mixture of the anti-PD1 mAb (609A) plus trastuzumab. Note that one tumor cell was ligated with two T cells, while a single T cell
was in contact with two tumor cells in the presence of the BsAb. The combination of 609A and trastuzumab failed to induce PD1 synapse
formation with T cells. The white arrows denote T cells (DIC channel), for which PD1 staining (green channel) were barely seen owing to the
rearrangement of PD1 during synapse formation.
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
6
Acta Pharmacologica Sinica (2021) 0:1 9
treatment, on par with that of the anti-PD1 mAb, 609A, where
the parent mAb also inhibited 85% of tumor growth at an equal
molar dose of 10 mg/kg on the same day (Fig. 6band
Supplementary Fig. S5e). Thus, these results indicated that the
anti-HER2×PD1 BsAb retained the antitumor efcacy of each of
its arms to a level comparable to that of their parental mAbs
in vivo. It is worth noting that no apparent toxicity associated
with the BsAb was observed, as the animal body weights kept
climbing over the entire course of the treatment (Supplemen-
tary Fig. S5a, b).
Since the BsAb does not cross-react with murine PD1 and HER2,
we chose a model system in which we introduced human PBMCs
into immune-compromised NOD scid gamma mice in order to test
the synergistic antitumor activity of the BsAb. Human PBMCs were
injected intraperitoneally following the establishment of JIMT-1
tumors in the mice. Although injection of human PBMCs into mice
bearing JIMT-1 tumors slightly reduced the body weights initially,
the body weights of all groups returned to baseline following
treatment (Supplementary Fig. S5c). We then tested the antitumor
efcacy of the BsAb in comparison with that of the combination of
the two parental mAbs. The anti-PD1 mAb, 609A, failed to show
antitumor effects on JIMT-1 tumors relative to the control, whereas
609A in combination with trastuzumab signicantly suppressed
tumor growth compared to the monotherapy. Importantly, the
BsAb also potently inhibited JIMT-1 tumor growth comparable to
that of the combination of the two parent mAbs (Fig. 6cand
Supplementary Fig. S5f), indicating that the BsAb indeed exhibited
dual targeting functions in vivo.
DISCUSSION
The goal of our study was to combine traditional HER2-targeted
therapy with PD1/PDL1 blockade with a novel anti-HER2×PD1
BsAb to treat HER2-overexpressing solid tumors. The BsAb
comprises a full trastuzumab IgG with two copies of anti-PD1
(609A) scFvs symmetrically fused to the C-terminus of the heavy
chains. The orientation of the IgG and the scFvs was selected
based on the nding that the BsAb with scFvs fused to the N-
terminus exhibited ve-fold reduced binding afnity for HER2
receptors on tumor cells relative to that of the BsAb with scFvs
fused to the C-terminus. This is in contrast to an anti-EGFR×PD1
BsAb we previously reported, where fusion of 609A scFvs to the N-
terminus of the cetuximab heavy chains was required to keep the
binding afnity of the BsAb for target cells intact, in comparison
with the parental mAb, cetuximab [39]. Our results emphasize that
the geometrical steric hindrance of a receptor on the surface of
tumor cells should be carefully assessed during the design of
tumor-associated antigen (TAA)-targeting BsAbs.
ADCC plays an important role in trastuzumab-mediated tumor
cell killing [4749]. Ideally, the BsAb is expected to retain full ADCC
toward HER2-overexpressing tumor cells while sparing T cells from
the attack. Indeed, the BsAb comprising the intact trastuzumab
Fig. 6 The anti-HER2×PD1 BsAb exhibited potent antitumor effects in vivo. a A control (black circle), the BsAb (blue square and pink
triangle), and trastuzumab (green triangle) were i.p. injected into mice bearing NCI-N87 tumors at the indicated doses. Tumor volumes (mm
3
)
were measured at the indicated time points. bA control (black circle), the BsAb (green triangle and diamond), and the anti-PD1 mAb, 609A
(red square and triangle) were i.p. injected into human PD1 transgenic mice bearing mouse MC38 tumors at the indicated doses. cAn isotype
control (blue square), 609A (orange triangle), 609A plus trastuzumab (green diamond, 20 mg/kg each), and the BsAb (red circle) were injected
into M-NSG mice bearing JIMT-1 tumors in the presence of human PBMCs at the indicated doses. WT mice (black dot) were used as control.
Tumor volumes (mm
3
) were measured at the indicated time points. Mean ± SEM. *P< 0.05 by two-way ANOVA (the BsAb/PBMC vs Vehicle/
PBMC) and ****P< 0.0001 for the comparison of all indicated groups with the control group.
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
7
Acta Pharmacologica Sinica (2021) 0:1 9
IgG exhibited ADCC toward HER2-overexpressing tumor cells as
effectively as parental trastuzumab, whereas the 609A scFv moiety
failed to induce measurable ADCC toward T cells, even though the
BsAb could effectively bind to PD1 on T cells. Interestingly, we
recently reported that the anti-EGFR×PD1 BsAb with 609A scFvs
fused to the N-terminus of the cetuximab heavy chains also
showed ADCC specic to tumor cells but not to T cells, suggesting
that an intact tertiary conguration of an IgG might be a critical
determinant for induction of ADCC to kill target cells.
Surprisingly, the anti-PD1 mAb, 609A, in the presence of PBMCs
did not enhance the killing of PDL1-overexpressing tumor cells
(N87-PDL1) compared to that of PBMCs alone. This was further
supported by the nding that the combination of trastuzumab
with 609A did not increase the killing of N87-PDL1 cells relative to
that of trastuzumab alone in the presence of PBMCs, suggesting
that blockade of PD1/PDL1 signaling did not play an important
role in T cell-mediated tumor cell killing in this experimental
setting. To conrm this hypothesis, we performed tumor cell
killing assays with NCI-N87 cells, which do not express PDL1, in
the presence of activated T cells. Indeed, the addition of 609A did
not enhance killing compared to the addition of T cells alone, nor
did it enhance trastuzumab-mediated killing when administered
in combination with trastuzumab. In contrast, the BsAb exhibited
superior killing of NCI-N87 tumor cells relative to all other
treatments. These data imply that BsAb-mediated tumor cell
killing may not require presentation of tumor cell antigens to
T cells mediated by the MHC as seen in the case of CD3-targeting
bispecic T-cell engagers [50]. However, T cells activated by PD1
blockade may still rely on, at least to a certain degree, MHC-
dependent antigen presentation for target cell recognition and
killing. It is plausible that in addition to the TCR/CD3 complex,
crosslinking a TAA with a functional regulatory receptor of T cells,
such as PD1, might be sufcient to initiate T cell-dependent
target cell killing without the need for MHC-mediated antigen
presentation.
In addition to direct tumor cell killing, the BsAb-induced
engagement of tumor cells with T cells via HER2 facilitated the
formation of PD1 immunological synapses as seen for the anti-
EGFR×PD1 BsAb [39]. These results suggest that T-cell activation
by PD1 synapse formation and MHC-independent target cell
recognition by T cells might be common advantages of a BsAb
targeting PD1 and a TAA over the combination of two mAbs for
therapeutic use.
Recently, growing evidence has demonstrated that HER2 signaling
might be coupled with PD1/PDL1 signaling in HER2-overexpressing
tumors [51]. Suh et al. demonstrated that PDL1 expression was more
frequent in patients with positive HER2 expression after analysis of
over 250 resected tumor tissues. In support of this clinical nding,
they also showed that cell lines overexpressing HER2 express a higher
level of PDL1 [52]. Interestingly, Fans lab showed that targeting HER2
with trastuzumab upregulated the expression of PDL1 via upregula-
tion of IFNγproduction, which presumably suppressed antitumor
immune responses and led to drug resistance [53]. Thus, a BsAb
simultaneously targeting HER2 and PD1/PDL1 signaling pathways
might be more effective in treating HER2-overexpressing tumors than
the corresponding mAbs. Indeed, our BsAb and a mouse specic
BsAb, BsPDL1 × rErbB2, which targets PDL1 and HER2 [54], showed
enhanced antitumor activities relative to that of monotherapies with
mAbs at equal molar doses in vivo, although the latter is a
monovalent BsAb that did not inhibit tumor cell growth in vitro,
suggesting that combining HER2 and PD1/PDL1 blockades might
improve the treatment of HER2-overexpressing cancers.
In summary, we produced a new anti-HER2×PD1 BsAb. The
BsAb was much more cytotoxic to HER2-overexpressing tumor
cells in the presence of PBMCs or activated T cells than was
trastuzumab either alone or in combination with the parental anti-
PD1 antibody, independent of PDL1 expression. We postulate that,
in addition to direct HER2 inhibition and ADCC, as is the case with
trastuzumab, there are several novel mechanisms that may further
contribute to the enhanced cytotoxicity of the BsAb: (1) the
BsAb redirects T cells to the proximity of tumor cells and induces
direct tumor cell killing most likely in an antigen-presentation-
independent manner; (2) the BsAb can potentially activate T cells
in the tumor microenvironment via PD1/PDL1 blockade, and (3)
the BsAb is capable of further enhancing T cell cytotoxic activity
by the formation of PD1 immunological synapses. Thus, we
provide a new promising option for treating late-stage or
refractory tumors that overexpress HER2.
ACKNOWLEDGEMENTS
We thank Ya Zhang and Xia Dong for their excellent technical assistance. We would
also like to acknowledge expert services provided by the Protein Chemistry and
Analytical Department at Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd.
AUTHOR CONTRIBUTIONS
CLG and HXZ made the constructs, designed and performed in vitro experiments
including cell-based assays and analyzed data; LD and XQM designed and
performed microscopy including acquisition of time-lapse movies; KL performed
ADCC assays; WX puried proteins; LZ and YQL performed in vivo studies; ZPZ
reviewed and edited the manuscript; HMH supervised the study and wrote and
revised the manuscript.
ADDITIONAL INFORMATION
Supplementary information The online version contains supplementary material
available at https://doi.org/10.1038/s41401-021-00683-8.
Competing interests: The authors declare no potential conicts of interest. All
authors are employees of Sunshine Guojian Pharmaceutical (Shanghai) when the
study was carried out.
REFERENCES
1. de Melo Gagliato D, Jardim DL, Marchesi MS, Hortobagyi GN. Mechanisms
of resistance and sensitivity to anti-HER2 therapies in HER2
+
breast cancer.
Oncotarget. 2016;7:6443146.
2. Harbeck N, Pegram MD, Rüschoff J, Möbus V. Targeted therapy in metastatic
breast cancer: The HER2/neu Oncogene. Breast Care. 2010;5:37.
3. Menard S, Casalini P, Campiglio M, Pupa SM, Tagliabue E. Role of HER2/neu in
tumor progression and therapy. Cell Mol Life Sci. 2004;61:296578.
4. Zhang X, Chen J, Weng Z, Li Q, Zhao L, Yu N, et al. A new anti-HER2 antibody that
enhances the anti-tumor efcacy of trastuzumab and pertuzumab with a distinct
mechanism of action. Mol Immunol. 2020;119:4858.
5. Brandão M, Pondé NF, Poggio F, Kotecki N, Salis M, Lambertini M, et al. Combi-
nation therapies for the treatment of HER2-positive breast cancer: current and
future prospects. Expert Rev Anticancer Ther. 2018;18:62949.
6. Yamashita-Kashima Y, Iijima S, Yorozu K, Furugaki K, Kurasawa M, Ohta M, et al.
Pertuzumab in combination with trastuzumab shows signicantly enhanced
antitumor activity in HER2-positive human gastric cancer xenograft models. Clin
Cancer Res. 2011;17:506070.
7. Nahta R, Hung MC, Esteva FJ. The HER-2-targeting antibodies trastuzumab and
pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res.
2004;64:23436.
8. Hurvitz SA, Martin M, Symmans WF, Jung KH, Huang CS, Thompson AM, et al.
Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab
emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRIS-
TINE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2018;19:
11526.
9. Pondé N, Aftimos P, Piccart M. Antibody-drug conjugates in breast cancer: a
comprehensive review. Curr Treat Options Oncol. 2019;20:37.
10. Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, et al. Development and clinical application
of anti-HER2 monoclonal and bispecic antibodies for cancer treatment. Exp
Hematol Oncol. 2017;6:31.
11. Yu S, Zhang J, Yan Y, Yao X, Fang L, Xiong H, et al. A novel asymmetrical anti-
HER2/CD3 bispecic antibody exhibits potent cytotoxicity for HER2-positive
tumor cells. J Exp Clin Cancer Res. 2019;38:355.
12. Lopez-Albaitero A, Xu H, Guo H, Wang L, Wu Z, Tran H, et al. Overcoming
resistance to HER2-targeted therapy with a novel HER2/CD3 bispecic antibody.
Oncoimmunology. 2017;6:e1267891.
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
8
Acta Pharmacologica Sinica (2021) 0:1 9
13. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immu-
notherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med.
2015;21:2433.
14. Riley JL. PD-1 signaling in primary T cells. Immunol Rev. 2009;229:11425.
15. Schütz F, Stefanovic S, Mayer L, von Au A, Domschke C, Sohn C. PD-1/PD-L1
pathway in breast cancer. Oncol Res Treat. 2017;40:2947.
16. Yao H, Wang H, Li C, Fang JY, Xu J. Cancer cell-intrinsic PD-1 and implications in
combinatorial immunotherapy. Front Immunol. 2018;9:1774.
17. Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat
Rev Immunol. 2018;18:15367.
18. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and
immunity. Annu Rev Immunol. 2008;26:677704.
19. Ai L, Xu A, Xu J. Roles of PD-1/PD-L1 pathway: signaling, cancer, and beyond. Adv
Exp Med Biol. 2020;1248:3359.
20. Constantinidou A, Alieris C, Trafalis DT. Targeting programmed cell death -1 (PD-
1) and ligand (PD-L1): a new era in cancer active immunotherapy. Pharmacol
Ther. 2019;194:84106.
21. Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade
in cancer immunotherapy. Comput Struct Biotechnol J. 2019;17:66174.
22. Bardhan K, Anagnostou T, Boussiotis VA. The PD1:PD-L1/2 pathway from dis-
covery to clinical implementation. Front Immunol. 2016;7:550.
23. Balar AV, Weber JS. PD-1 and PD-L1 antibodies in cancer: current status and
future directions. Cancer Immunol Immunother. 2017;66:55164.
24. Carretero-González A, Lora D, Ghanem I, Zugazagoitia J, Castellano D, Sepúlveda
JM, et al. Analysis of response rate with anti PD1/PD-L1 monoclonal antibodies in
advanced solid tumors: a meta-analysis of randomized clinical trials. Oncotarget.
2018;9:870615.
25. Tsai KK, Zarzoso I, Daud AI. PD-1 and PD-L1 antibodies for melanoma. Hum
Vaccin Immunother. 2014;10:31116.
26. Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the
PD-1 signaling pathway. J Biomed Sci. 2017;24:26.
27. Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, et al. PD-1
+
regulatory
T cells amplied by PD-1 blockade promote hyperprogression of cancer. Proc
Natl Acad Sci USA. 2019;116:999910008.
28. Levingston CA, Young MR. Transient immunological and clinical effectiveness of
treating mice bearing premalignant oral lesions with PD-1 antibodies. Int J
Cancer. 2017;140:160919.
29. Noguchi T, Ward JP, Gubin MM, Arthur CD, Lee SH, Hundal J, et al. Temporally
distinct PD-L1 expression by tumor and host cells contributes to immune escape.
Cancer Immunol Res. 2017;5:10617.
30. Patsoukis N, Wang Q, Strauss L, Boussiotis VA. Revisiting the PD-1 pathway. Sci
Adv. 2020;6:113.
31. Bardwell PD, Staron MM, Liu J, Tao Q, Scesney S, Bukofzer G, et al. Potent and
conditional redirected T cell killing of tumor cells using Half DVD-Ig. Protein Cell.
2018;9:1219.
32. Dahlén E, Veitonmäki N, Norlén P. Bispecic antibodies in cancer immunotherapy.
Therapeutic Adv Vaccin Immunother. 2018;6:317.
33. Huang S, van Duijnhoven SMJ, Sijts A, van Elsas A. Bispecic antibodies targeting
dual tumor-associated antigens in cancer therapy. J Cancer Res Clin Oncol.
2020;146:311122.
34. Sedykh SE, Prinz VV, Buneva VN, Nevinsky GA. Bispecic antibodies: design,
therapy, perspectives. Drug Des Dev Ther. 2018;12:195208.
35. Tkachev V, Goodell S, Opipari AW, Hao LY, Franchi L, Glick GD, et al. Programmed
death-1 controls T cell survival by regulating oxidative metabolism. J Immunol.
2015;194:5789800.
36. Koopmans I, Hendriks D, Samplonius DF, van Ginkel RJ, Heskamp S, Wierstra PJ,
et al. A novel bispecic antibody for EGFR-directed blockade of the PD-1/PD-L1
immune checkpoint. Oncoimmunology. 2018;7:e1466016.
37. Benonisson H, AltıntaşI, Sluijter M, Verploegen S, Labrijn AF, Schuurhuis DH, et al.
CD3-bispecic antibody therapy turns solid tumors into inammatory sites but
does not install protective memory. Mol Cancer Ther. 2019;18:31222.
38. Dong J, Sereno A, Aivazian D, Langley E, Miller BR, Snyder WB, et al. A stable IgG-
like bispecic antibody targeting the epidermal growth factor receptor and
the type I insulin-like growth factor receptor demonstrates superior anti-tumor
activity. MAbs. 2011;3:27388.
39. Li L, Deng L, Meng X, Gu C, Meng L, Li K, et al. Tumor-targeting anti-EGFR x anti-
PD1 bispecic antibody inhibits EGFR-overexpressing tumor growth by com-
bining EGFR blockade and immune activation with direct tumor cell killing. Transl
Oncol. 2021;14:100916.
40. Fan G, Wang Z, Hao M, Li J. Bispecic antibodies and their applications. J Hematol
Oncol. 2015;8:130.
41. Brinkmann U, Kontermann RE. The making of bispecic antibodies. MAbs.
2017;9:182212.
42. Gu J, Xu J, Lei C, DSouza P, Moscoso-Castro M, Wang Z, et al. Biology drives
the discovery of bispecic antibodies as innovative therapeutics. Antib Ther.
2020;3:1862.
43. Tiller KE, Tessier PM. Advances in antibody design. Annu Rev Biomed Eng. 2015;
17:191216.
44. Wang C, Thudium KB, Han M, Wang XT, Huang H, Feingersh D, et al. In vitro
characterization of the anti-PD-1 antibody nivolumab, BMS-936558, and in vivo
toxicology in non-human primates. Cancer Immunol Res. 2014;2:84656.
45. Pentcheva-Hoang T, Chen L, Pardoll DM, Allison JP. Programmed death-1 con-
centration at the immunological synapse is determined by ligand afnity and
availability. Proc Natl Acad Sci USA. 2007;104:1776570.
46. Li J, Stagg NJ, Johnston J, Harris MJ, Menzies SA, DiCara D, et al. Membrane-
proximal epitope facilitates efcient T cell synapse formation by anti-
FcRH5/CD3 and is a requirement for myeloma cell killing. Cancer Cell. 2017;31:
38395.
47. van der Haar Àvila I, Marmol P, Cany J, Kiessling R, Pico de Coaña Y. Evaluating
antibody-dependent cell-mediated cytotoxicity by ow cytometry. Methods Mol
Biol. 2019;1913:18194.
48. Arnould L, Gelly M, Penault-Llorca F, Benoit L, Bonnetain F, Migeon C, et al.
Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-
dependent cellular cytotoxicity mechanism? Br J Cancer. 2006;94:25967.
49. Junttila TT, Akita RW, Parsons K, Fields C, Lewis Phillips GD, Friedman LS, et al. Ligand-
independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively
inhibited by the PI3K inhibitor GDC-0941. Cancer Cell. 2009;15:42940.
50. Einsele H, Borghaei H, Orlowski RZ, Subklewe M, Roboz GJ, Zugmaier G, et al.
The BiTE (bispecicTcell engager) platform: development and future potential of
a targeted immunooncology therapy across tumor types. Cancer. 2020;126:
3192201.
51. Padmanabhan R, Kheraldine HS, Meskin N, Vranic S, Al Moustafa AE. Crosstalk
between HER2 and PD-1/PD-L1 in breast cancer: from clinical applications to
mathematical models. Cancers. 2020;12:125.
52. Suh KJ, Sung JH, Kim JW, Han S-H, Lee HS, Min A, et al. EGFR or HER2 inhibition
modulates the tumor microenvironment by suppression of PD-L1 and cytokines
release. Oncotarget. 2017;8:6390110.
53. Chaganty BKR, Qiu S, Gest A, Lu Y, Ivan C, Calin GA, et al. Trastuzumab upre-
gulates PD-L1 as a potential mechanism of trastuzumab resistance through
engagement of immune effector cells and stimulation of IFNγsecretion. Cancer
Lett. 2018;430:4756.
54. Mittal D, Vijayan D, Neijssen J, Kreijtz J, Habraken M, Van Eenennaam H, et al.
Blockade of ErbB2 and PD-L1 using a bispecic antibody to improve targeted
anti-ErbB2 therapy. Oncoimmunology. 2019;8:e1648171.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this license, visit http://creativecommons.
org/licenses/by/4.0/.
© The Author(s) 2021
An anti-HER2xPD1 BsAb exhibited superior antitumor activity
CL Gu et al.
9
Acta Pharmacologica Sinica (2021) 0:1 9
... Radiotherapy and drug therapy are the main treatments for patients with advanced NSCLC in clinical practice [42], among which immunotherapy drugs have achieved good results, but there are also many shortcomings. In recent years, many biological immune agents have been developed and applied in clinical practice, but severe drug resistance limits their use [43][44][45]. In addition, immunosuppressive cells (such as Treg) in the tumor microenvironment can significantly inhibit CTLmediated anti-tumor immunity [46][47][48], which is not conducive to the effect of immunotherapy drugs. ...
Article
Full-text available
Background Immune checkpoint inhibitors, which have attracted much attention in recent years, have achieved good efficacy, but their use is limited by the high incidence of acquired drug resistance. Therefore, there is an urgent need to develop new immunotherapy drugs. Compound taxus chinensis capsule (CTC) is an oral paclitaxel compound drug, clinical results showed it can change the number of regulatory T cells and T helper cell 17 in peripheral blood. Regulating the balance between regulatory T cells and T helper cell 17 is considered to be an effective anticancer strategy. Paclitaxel and ginsenoside metabolite compound K are the main immunomodulatory components, it is not clear that paclitaxel combined with compound K can inhibit tumor development by regulating the balance between regulatory T cell and T helper cell 17. Methods MTT, EdU proliferation and plate colony formation assay were used to determine the concentration of paclitaxel and compound K. AnnexinV-FITC/PI staining, ELISA, Western Blot assay, Flow Cytometry and Immunofluorescence were used to investigate the effect of paclitaxel combined with compound K on Lewis cell cultured alone or co-cultured with splenic lymphocyte. Finally, transplanted tumor C57BL/6 mice model was constructed to investigate the anti-cancer effect in vivo. Results According to the results of MTT, EdU proliferation and plate colony formation assay, paclitaxel (10 nM) and compound K (60 μM) was used to explore the mechanism. The results of Flow Cytometry demonstrated that paclitaxel combined with compound K increased the number of T helper cell 17 and decreased the number of regulatory T cells, which induced pyroptosis of cancer cells. The balance was mediated by the JAK–STAT pathway according to the results of Western Blot and Immunofluorescence. Finally, the in vivo results showed that paclitaxel combined with compound K significantly inhibit the progression of lung cancer. Conclusions In this study, we found that paclitaxel combined with compound K can activate CD8 ⁺ T cells and induce pyroptosis of tumor cells by regulating the balance between regulatory T cells and T helper cell 17. These results demonstrated that this is a feasible treatment strategy for lung cancer.
... These aforementioned studies, along with several other preclinical studies, indicated a potential synergistic effect when PD-1 inhibitors were combined with HER2-targeted therapy. Currently, multiple clinical trials on breast and gastric cancers are underway or have explored the effectiveness of these combined treatment approaches [24][25][26][27]. This review summarizes clinical trials related to the combined treatment of patients with HER2-positive breast cancer, gastric cancer, and non-small cell lung cancer with PD-1 inhibitors. ...
Article
Full-text available
Background Patients with HER2-positive cancers often face a poor prognosis, and treatment regimens containing anti-HER2 have become the first-line treatment options for breast and gastric cancers. However, these approaches are faced with significant challenges in terms of drug resistance. Hence, it is crucial to explore precise treatment strategies aimed at improving survival outcomes. Advancements in treatment Over the past few years, there has been rapid advancement in the realm of tumor therapy, particularly with the swift progress of immune checkpoint inhibitors, including PD-1/PD-L1 inhibitors. They exert anti-tumor effects by disrupting immune-suppressive factors within the tumor microenvironment. However, monotherapy with PD-1/PD-L1 inhibitors has several limitations. Consequently, numerous studies have explored combinatorial immunotherapeutic strategies and demonstrated highly promising avenues of development. Objective This article aims to review the clinical trials investigating PD-1/PD-L1 inhibitor combination therapy for HER2-positive tumors. Additionally, it provides a summary of ongoing trials evaluating the efficacy and safety of these combined treatments, with the intention of furnishing valuable insights for the clinical management of HER2-positive cancer. Conclusion Combinatorial immunotherapeutic strategies involving PD-1/PD-L1 inhibitors hold considerable promise in the treatment of HER2-positive tumors. Continued research efforts and clinical trials are warranted to elucidate optimal treatment regimens that maximize therapeutic benefits while minimizing adverse effects.
... Furthermore, it is not only the molecular geometry that is affecting the bsAb potency but also the relative orientation of the specificities. As an example, significantly reduced HER2 binding was observed when fusing anti-HER2 scFv to an anti-PD1 IgG scaffold compared to reverse orientation, i.e., fusion of the anti-PD1 scFv to the anti-HER2 IgG (Guling et al., 2022). Similar effects from relative orientation have also been shown for fragment-based bsAbs (de Bruin et al., 2018;Andres et al., 2019). ...
Article
Full-text available
Bispecific antibodies (bsAbs) have attracted significant attention due to their dual binding activity, which permits simultaneous targeting of antigens and synergistic binding effects beyond what can be obtained even with combinations of conventional monospecific antibodies. Despite the tremendous therapeutic potential, the design and construction of bsAbs are often hampered by practical issues arising from the increased structural complexity as compared to conventional monospecific antibodies. The issues are diverse in nature, spanning from decreased biophysical stability from fusion of exogenous antigen-binding domains to antibody chain mispairing leading to formation of antibody-related impurities that are very difficult to remove. The added complexity requires judicious design considerations as well as extensive molecular engineering to ensure formation of high quality bsAbs with the intended mode of action and favorable drug-like qualities. In this review, we highlight and summarize some of the key considerations in design of bsAbs as well as state-of-the-art engineering principles that can be applied in efficient construction of bsAbs with diverse molecular formats.
... [73][74][75][76][77][78] Recently, progress has been made in the creation of bispecific antibodies that target both the immune checkpoint molecules PD-1/ PD-L1 and HER2, with antitumor effects proven in preclinical animals. 79,80 In trastuzumab-resistant cancer models, bispecific antibodies targeting immune checkpoint molecules and HER2 may be more successful than individual mAb treatments. The use of H 2 Mabs to create bispecific antibodies is another option for increasing anti-HER2 treatment. ...
Article
Full-text available
Breast cancer patients with high levels of human epidermal growth factor receptor 2 (HER2) expression have worse clinical outcomes. Anti‐HER2 monoclonal antibody (mAb) is the most important therapeutic modality for HER2‐positive breast cancer. We previously immunized mice with the ectodomain of HER2 to create the anti‐HER2 mAb, H 2 Mab‐77 (mouse IgG 1 , kappa). This was then altered to produce H 2 Mab‐77‐mG 2a ‐f, an afucosylated mouse IgG 2a . In the present work, we examined the reactivity of H 2 Mab‐77‐mG 2a ‐f and antitumor effects against breast cancers in vitro and in vivo. BT‐474, an endogenously HER2‐expressing breast cancer cell line, was identified by H 2 Mab‐77‐mG 2a ‐f with a strong binding affinity (a dissociation constant [ K D ]: 5.0 × 10 ⁻⁹ M). H 2 Mab‐77‐mG 2a ‐f could stain HER2 of breast cancer tissues in immunohistochemistry and detect HER2 protein in Western blot analysis. Furthermore, H 2 Mab‐77‐mG 2a ‐f demonstrated strong antibody‐dependent cellular cytotoxicity (ADCC) and complement‐dependent cytotoxicity (CDC) for BT‐474 cells. MDA‐MB‐468, a HER2‐negative breast cancer cell line, was unaffected by H 2 Mab‐77‐mG 2a ‐f. Additionally, in the BT‐474‐bearing tumor xenograft model, H 2 Mab‐77‐mG 2a ‐f substantially suppressed tumor development when compared with the control mouse IgG 2a mAb. In contrast, the HER2‐negative MDA‐MB‐468‐bearing tumor xenograft model showed no response to H 2 Mab‐77‐mG 2a ‐f. These findings point to the possibility of H 2 Mab‐77‐mG 2a ‐f as a treatment regimen by showing that it has antitumor effects on HER2‐positive breast tumors.
Article
Full-text available
Immune cells are pivotal in cancer immunotherapy, yet their therapeutic effectiveness is often hampered by limited tumor infiltration and inhibitory tumor microenvironments. We here introduce an alkaline phosphatase (ALP)‐responsive and transformable supramolecular bis‐specific cell engager (Supra‐BiCE) to harness NK/T cells for effective cancer immunotherapy. The Supra‐BiCE, consisting of both SA‐P (a phosphorylated peptide targeting and blocking PD‐L1) and SA‐T (a phosphorylated peptide targeting and blocking TIGIT) is constructed by a simple co‐assembling strategy. Upon intravenous administration, Supra‐BiCE self‐assembles into nanoribbons and interacts with NK/T cells via TIGIT. Notably, these nanoribbons undergo transformation into long nanofibrils within ALP‐overexpressing tumor regions, resulting in enhanced binding affinities of Supra‐BiCE to both PD‐L1 and TIGIT. Consequently, this leads to the accumulation and retention of NK cells and CD8 ⁺ T cells within tumor regions. Furthermore, the combinatorial blockade of checkpoints by Supra‐BiCE activates infiltrating NK/T cells, as evidenced by upregulated expression of perforin, granzyme B, TNF‐a, and IL‐2. Moreover, the adjustable peptide ratio in Supra‐BiCE enables customization for optimal therapeutic effects against distinct tumor types. Particularly, Supra‐BiCE (T:P = 1:3) achieved 98.27% tumor suppression rate against CT26 cell tumor model. Overall, our study offers a promising tool for engaging NK and T cells for cancer immunotherapy. This article is protected by copyright. All rights reserved
Article
We report here the development of two different sensing strategies based on the use of antigen-conjugated nucleic acid strands for the detection of a bispecific antibody against the tumor-related proteins Mucin1 and epidermal growth factor receptor. Both approaches work well in serum samples (nanomolar sensitivity), show high specificity against the two monospecific antibodies, and are rapid. The results presented here demonstrate the versatility of DNA-based platforms for the detection of bispecific antibodies and could represent a versatile alternative to other more reagent-intensive and time-consuming analytical approaches.
Article
Full-text available
Glioblastoma (GBM) is a highly malignant brain cancer with a poor prognosis despite standard treatments. This investigation aimed to explore the feasibility of PTPN6 to combat GBM with immunotherapy. Our study employed a comprehensive analysis of publicly available datasets and functional experiments to assess PTPN6 gene expression, prognostic value, and related immune characteristics in glioma. We evaluated the influence of PTPN6 expression on CD8+ T cell exhaustion, immune suppression, and tumor growth in human GBM samples and mouse models. Our findings demonstrated that PTPN6 overexpression played an oncogenic role in GBM and was associated with advanced tumor grades and unfavorable clinical outcomes. In human GBM samples, PTPN6 upregulation showed a strong association with immunosuppressive formation and CD8+ T cell dysfunction, whereas, in mice, it hindered CD8+ T cell infiltration. Moreover, PTPN6 facilitated cell cycle progression, inhibited apoptosis, and promoted glioma cell proliferation, tumor growth, and colony formation in mice. The outcomes of our study indicate that PTPN6 is a promising immunotherapeutic target for the treatment of GBM. Inhibition of PTPN6 could enhance CD8+ T cell infiltration and improve antitumor immune response, thus leading to better clinical outcomes for GBM patients.
Article
Full-text available
We developed a strategy to combine conventional targeted therapy with immune checkpoint blockade using a tumor-targeting bispecific antibody (BsAb) to treat solid tumors. The BsAb was designed to simultaneously engage a tumor-associated antigen, epidermal growth factor receptor (EGFR), and programed cell death protein 1 (PD1). In addition to its direct anti-tumor activity via EGFR inhibition, the BsAb mediated efficient antibody-dependent cellular cytotoxicity (ADCC) and activated T cell antitumor immunity through blockade of PD1 from interacting with its counterpart, programed cell death ligand 1 (PDL1). Further, the BsAb exhibited a potent direct tumor cell killing activity in the presence of PBMC, most likely, via activating and, at the same time, physically engaging T cells with tumor cells. Taken together, we here illustrate a new strategy in the design and production of novel BsAbs with enhanced therapeutic efficacy through both direct tumor growth inhibition and T cell activation via tumor-targeted immune checkpoint blockade.
Article
Full-text available
Purpose: Bispecific antibodies (BsAbs) have emerged as a leading drug class for cancer therapy and are becoming increasingly of interest for therapeutic applications. As of April 2020, over 123 BsAbs are under clinical evaluation for use in oncology (including the two marketed BsAbs Blinatumomab and Catumaxomab). The majority (82 of 123) of BsAbs under clinical evaluation can be categorized as bispecific immune cell engager whereas a second less well-discussed subclass of BsAbs targets two tumor-associated antigens (TAAs). In this review, we summarize the clinical development of dual TAAs targeting BsAbs and provide an overview of critical considerations when designing dual TAA targeting BsAbs. Methods: Herein the relevant literature and clinical trials published in English until April 1st 2020 were searched using PubMed and ClinicalTrials.gov database. BsAbs were considered to be active in clinic if their clinical trials were not terminated, withdrawn or completed before 2018 without reporting results. Data missed by searching ClinicalTrials.gov was manually curated. Results: Dual TAAs targeting BsAbs offer several advantages including increased tumor selectivity, potential to concurrently modulate two functional pathways in the tumor cell and may yield improved payload delivery. Conclusions: Dual TAAs targeting BsAbs represent a valuable class of biologics and early stage clinical studies have demonstrated promising anti-tumor efficacy in both hematologic malignancies and solid tumors.
Article
Full-text available
Programmed Death-1 (PD-1; CD279) is an inhibitory receptor induced in activated T cells. PD-1 engagement by its ligands, PD-L1 and PD-L2, maintains peripheral tolerance but also compromises anti-tumor immunity. Blocking antibodies against PD-1 or its ligands have revolutionized cancer immunotherapy. However, only a fraction of patients develop durable antitumor responses. Clinical outcomes have reached a plateau without substantial advances by combinatorial approaches. Thus, great interest has recently emerged to investigate, in depth, the mechanisms by which the PD-1 pathway transmits inhibitory signals with the goal to identify molecular targets for improvement of the therapeutic success. These efforts have revealed unpredictable dimensions of the pathway and uncovered novel mechanisms involved in PD-1 and PD-L1 regulation and function. Here, we provide an overview of the recent advances on the mechanistic aspects of the PD-1 pathway and discuss the implications of these new discoveries and the gaps that remain to be filled.
Article
Full-text available
Immuno‐oncology therapies engage the immune system to treat cancer. BiTE (bispecific T‐cell engager) technology is a targeted immuno‐oncology platform that connects patients' own T cells to malignant cells. The modular nature of BiTE technology facilitates the generation of molecules against tumor‐specific antigens, allowing off‐the‐shelf immuno‐oncotherapy. Blinatumomab was the first approved canonical BiTE molecule and targets CD19 surface antigens on B cells, making blinatumomab largely independent of genetic alterations or intracellular escape mechanisms. Additional BiTE molecules in development target other hematologic malignancies (eg, multiple myeloma, acute myeloid leukemia, and B‐cell non‐Hodgkin lymphoma) and solid tumors (eg, prostate cancer, glioblastoma, gastric cancer, and small‐cell lung cancer). BiTE molecules with an extended half‐life relative to the canonical BiTE molecules are also being developed. Advances in immuno‐oncology made with BiTE technology could substantially improve the treatment of hematologic and solid tumors and offer enhanced activity in combination with other treatments.
Article
Full-text available
Breast cancer is one of the major causes of mortality in women worldwide. The most aggressive breast cancer subtypes are human epidermal growth factor receptor-positive (HER2+) and triple-negative breast cancers. Therapies targeting HER2 receptors have significantly improved HER2+ breast cancer patient outcomes. However, several recent studies have pointed out the deficiency of existing treatment protocols in combatting disease relapse and improving response rates to treatment. Overriding the inherent actions of the immune system to detect and annihilate cancer via the immune checkpoint pathways is one of the important hallmarks of cancer. Thus, restoration of these pathways by various means of immunomodulation has shown beneficial effects in the management of various types of cancers, including breast. We herein review the recent progress in the management of HER2+ breast cancer via HER2-targeted therapies, and its association with the programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) axis. In order to link research in the areas of medicine and mathematics and point out specific opportunities for providing efficient theoretical analysis related to HER2+ breast cancer management, we also review mathematical models pertaining to the dynamics of HER2+ breast cancer and immune checkpoint inhibitors.
Article
Full-text available
A bispecific antibody (bsAb) is able to bind two different targets or two distinct epitopes on the same target. Broadly speaking, bsAbs can include any single molecule entity containing dual specificities with at least one being antigen-binding antibody domain. Besides additive effect or synergistic effect, the most fascinating applications of bsAbs are to enable novel and often therapeutically important concepts otherwise impossible by using monoclonal antibodies (mAbs) alone or their combination [1]. This so-called “obligate bsAbs” could open up completely new avenue for developing novel therapeutics. With evolving understanding of structural architecture of various natural or engineered antigen-binding immunoglobulin domains and the connection of different domains of an immunoglobulin molecule, and with greatly improved understanding of molecular mechanisms of many biological processes, the landscape of therapeutic bsAbs has significantly changed in recent years. As of September 2019, over 110 bsAbs are under active clinical development, and near 180 in preclinical development. In this review article, we introduce a system that classifies bsAb formats into 30 categories based on their antigen-binding domains and the presence or absence of Fc domain. We further review the biology applications of approximately 290 bsAbs currently in preclinical and clinical development, with the attempt to illustrate the principle of selecting a bispecific format to meet biology needs and selecting a bispecific molecule as a clinical development candidate by six critical criteria. Given the novel mechanisms of many bsAbs, the potential unknown safety risk and risk/benefit should be evaluated carefully during preclinical and clinical development stages. Nevertheless we are optimistic that next decade will witness clinical success of bsAbs or multispecific antibodies (msAbs) employing some novel mechanisms of action and deliver the promise as next wave of antibody-based therapeutics.
Article
Full-text available
The majority of patients with metastatic breast cancer who are treated with the anti-HER2 monoclonal antibody, trastuzumab, generally develop resistance to the drug within a year after initiation of the treatment. Here we describe a new anti-HER2 humanized monoclonal antibody, 19H6-Hu, which binds to HER2 extracellular domain (ECD) with high affinity and inhibits proliferation of multiple HER2-overexpressing cancer cell lines as a single agent or in combination with trastuzumab. 19H6-Hu binds to the domain III in proximity to the domain IV of HER2 ECD, which differs from trastuzumab and pertuzumab. 19H6-Hu in combination with trastuzumab was more effective at blocking phosphorylation of ERK1/2, AKT(S473)and HER2 (Y1248) in HER2-positive cancer cells compared to trastuzumab alone or in combination with pertuzumab. Combination of three antibodies, 19H6-Hu, inetetamab (a trastuzumab analog) and pertuzumab exhibited much stronger inhibition of large NCI-N87 tumor xenografts (>400mm3) than the current standard of care, inetetamab (trastuzumab) plus Docetaxel (DTX), as well as the combination of 19H6-Hu, inetetamab and DTX. Our results highlight the functional variability of HER2 domains and provide a new insight into the design of HER2-targeting agents.
Article
Full-text available
A significant proportion of human epidermal growth factor receptor 2 (Her2/ErbB2)-positive metastatic breast cancer patients are refractory to Her2-targeted trastuzumab-like therapy. Some of this resistance has been attributed to the upregulation of immune checkpoints such as programmed cell death-1 (PD-1) and its ligand, PD-L1 in Her2-positive breast cancer patients. Therefore, therapies targeting both the PD-1/PD-L1 interaction and oncogenic Her2 signaling are of significant clinical interest. Here, we constructed a mouse bispecific antibody targeting PD-L1 and rat Her2 (referred to as BsPD-L1xrErbB2) aiming to redirect the anti-PD-L1 response toward Her2-expressing tumor cells. BsPD-L1xrErbB2 demonstrated additive binding to interferon (IFN)-γ treated Her2⁺ TUBO tumor cells, but it did not affect the proliferation of tumor cells in-vitro. BsPD-L1xrErbB2 also blocked the PD-1/PD-L1 interaction. This bispecific antibody was constructed with a mouse IgG2a Fc backbone and interacted with Fcγ receptors and resulted in complement deposition (C3). ADCC and complement action could be potential mechanisms of action of this molecule. BsPD-L1xrErbB2 successfully reduced TUBO tumor growth and increased tumor rejection rate compared to the monovalent anti-PD-L1, monovalent anti-ErbB2 or the combination of anti-PD-L1 and anti-ErbB2 monotherapies. The enhanced anti-tumor effect of BsPD-L1xrErbB2 was dependent on CD8⁺ T lymphocytes and IFN-γ, as depletion of CD8⁺ T lymphocytes and neutralization of IFN-γ completely abolished the antitumor activity of the bispecific antibody. Consistently, BsPD-L1xrErbB2 treatment also increased the frequency of intratumor CD8⁺ T lymphocytes. Taken together, our data support a bispecific antibody approach to enhance the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade in Her2-positive metastatic breast cancers.
Article
Full-text available
Background: Human epidermal growth factor receptor 2 (HER2) is overexpressed in multiple cancers, which is associated with poor prognosis. Herceptin and other agents targeting HER2 have potent antitumor efficacy in patients with HER2-positive cancers. However, the development of drug resistance adversely impacts the efficacy of these treatments. It is therefore urgent to develop new HER2-targeted therapies. Bispecific antibodies (BsAbs) could guide immune cells toward tumor cells, and produced remarkable effects in some cancers. Methods: A BsAb named M802 that targets HER2 and CD3 was produced by introducing a salt bridge and knobs-into-holes (KIHs) packing into the structure. Flow cytometry was performed to determine its binding activity and cytotoxicity. CCK-8, Annexin V/PI staining, western blotting, and ELISA were utilized to study its effect on cell proliferation, apoptosis, the signaling pathways of tumor cells, and the secretion of cytokines by immune cells. Subcutaneous tumor mouse models were used to analyze the in vivo antitumor effects of M802. Results: We generated a new format of BsAb, M802, consisting of a monovalent unit against HER2 and a single chain unit against CD3. Our in vitro and in vivo experiments indicated that M802 recruited CD3-positive immune cells and was more cytotoxic than Herceptin in cells with high expression of HER2, low expression of HER2, and Herceptin resistance. Although M802 showed weaker effects than Herceptin on the PI3K/AKT and MAPK pathways, it was more cytotoxic due to its specific recognition of HER2 and its ability to recruit effector cells via its anti-CD3 moiety. Conclusions: Our results indicated that M802 exhibited potent antitumor efficacy in vitro and in vivo. M802 retained the function of Herceptin in antitumor signaling pathways, and also recruited CD3-positive immune cells to eliminate HER2-positive tumor cells. Therefore, M802 might be a promising HER2 targeted agent.
Chapter
Immunotherapies that target PD-1/PD-L1 axis have shown unprecedented success in a wide variety of human cancers. PD-1 is one of the key coinhibitory receptors expressed on T cells upon T cell activation. After engagement with its ligands, mainly PD-L1, PD-1 is activated and recruits the phosphatase SHP-2 in proximity to T cell receptor (TCR) and CD28 signaling. This event results in dephosphorylation and attenuation of key molecules in TCR and CD28 pathway, leading to inhibition of T cell proliferation, activation, cytokine production, altered metabolism and cytotoxic T lymphocytes (CTLs) killer functions, and eventual death of activated T cells. Bodies evolve coinhibitory pathways controlling T cell response magnitude and duration to limit tissue damage and maintain self-tolerance. However, tumor cells hijack these inhibitory pathways to escape host immune surveillance by overexpression of PD-L1. This provides the scientific rationale for clinical application of immune checkpoint inhibitors in oncology. The aberrantly high expression of PD-L1 in tumor microenvironment (TME) can be attributable to the “primary” activation of multiple oncogenic signaling and the “secondary” induction by inflammatory factors such as IFN-γ. Clinically, antibodies targeting PD-1/PD-L1 reinvigorate the “exhausted” T cells in TME and show remarkable objective response and durable remission with acceptable toxicity profile in large numbers of tumors such as melanoma, lymphoma, and mismatch-repair deficient tumors. Nevertheless, most patients are still refractory to anti-PD-1/PD-L1 therapy. Identifying the predictive biomarkers and design rational PD-1-based combination therapy become the priorities in cancer immunotherapy. PD-L1 expression, cytotoxic T lymphocytes infiltration, and tumor mutation burden (TMB) are generally considered as the most important factors affecting the effectiveness of PD-1/PD-L1 blockade. The revolution in cancer immunotherapy achieved by PD-1/PD-L1 blockade offers the paradigm for scientific translation from bench to bedside. The next decades will without doubt witness the renaissance of immunotherapy.