Revolutionizing Cancer Therapy With Bispecific Antibodies
Whitepaper
Published: December 2, 2024
Credit: iStock
Monoclonal antibodies, in particular immune checkpoint inhibitors (ICIs), have demonstrated remarkable efficacy and versatility in treating a range of cancers. However, they present relatively low overall response rates, a high incidence of immune-related adverse events as well as primary and acquired resistance.
Bispecific antibodies (bsAbs) are emerging as an alternative to overcome these limitations by enhancing immune activation, targeting tumor cells with greater precision and reduced side effects.
This whitepaper discusses the classification of bsAbs targeting immunomodulatory checkpoints and highlights their applications in cancer immunotherapy.
Download this whitepaper to explore:
- The advantages of using bsAbs for immunotherapies
- Key breakthroughs in targeting co-stimulatory and inhibitory checkpoints
- Promising clinical trial outcomes and future potential in oncology
WHITE PAPER
INTRODUCTION
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
In recent decades, one of the most significant
breakthroughs in cancer therapy has been the introduction
of monoclonal antibodies, especially immune checkpoint
inhibitors (ICIs) that target the PD-1/PD-L1 pathway.
These inhibitors have demonstrated remarkable efficacy
and versatility in treating a range of cancers.1 However,
the clinical application of ICIs faces several challenges,
including the relatively low overall response rates, a high
incidence of immune-related adverse events (irAEs), and
the issues of both primary and acquired resistance.1,2
To address these limitations, bispecific antibodies
(bsAbs) have emerged as a promising alternative. to
overcome some of the limitations associated with
monoclonal antibody (mAb) therapy, including ICIs.3
BsAbs offer enhanced therapeutic potential by precisely
linking immune cells with tumor cells, thereby improving
targeted engagement and minimizing off-target effects.
By simultaneously binding two distinct antigens or
epitopes, bsAbs increase therapeutic specificity, reduce
drug resistance, and lower the incidence of severe side
effects.4
Unlike traditional combination therapies that
use multiple monoclonal antibodies and carry and pose
increased risk of toxicity, bsAbs provide a more
controlled and focused treatment , enhancing safety
and efficacy.5
This review discuss the classification of bsAbs targeting
Immune checkpoints are broadly classified into two
primary types: co-stimulatory and co-inhibitory molecules.
Abnormal expression of inhibitory checkpoints on
cancer cells plays a critical role in immune evasion and
drug resistance. BsAbs targeting these checkpoints can
be categorized into three main groups: 1) those that
target dual inhibitory checkpoints, 2) those that target
both co-stimulatory and inhibitory checkpoints, and 3)
those that target immunomodulatory checkpoints along
with non-checkpoint molecules.2
CLASSIFYING BISPECIFIC ANTIBODY
TARGETING IMMUNOMODULATORY
CHECKPOINTS
Targeting dual inhibitory checkpoints
Bispecific Antibodies Targeting Immunomodulatory
Checkpoints for Cancer Therapy
The concept of therapeutically inhibiting two immune
checkpoints has driven the rational design of bsAbs
capable of targeting two inhibitory checkpoints
simultaneously, either on the same cell or on different
cells. This innovation builds upon the success of immune
checkpoint blockade (ICB) therapies, offering enhanced
clinical benefits that have been observed in patients
receiving combination ICB treatments.
Figure 1. BsAb classification2
Fab
A
Fc
VH
VL
CL
CH
B
Knobs-into-holes Duobody DVD-Ig IgG-(scFv2)
C
BiTE DART TandAb
immunomodulatory checkpoints and discusses their
emerging applications in cancer immunotherapy.
PD-1 x CTLA-4 bsAbs
Cadonilimab
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 2 of 9
Combining PD-1 and CTLA-4 targets in a single bsAb has
shown stronger anti-tumor activity compared to individual
mAbs.2
Studies have shown that combining mAbs drugs
targeting CTLA-4 like Ipilimumab with mAbs drugs
targeting PD-1 like Pembrolizumab or Nivolumab improves
therapeutic outcomes in patients with advanced renal cell
carcinoma (RCC), melanoma, and non-small cell lung cancer
(NSCLC).2
However, these mAbs drug combinations are
often associated with an increased risk of severe irAEs.
Notably, PD-1×CTLA-4 bsAbs exhibit enhanced anti-tumor
efficacy while presenting more manageable irAEs.6,7
Cadonilimab (AK104) is the world's first approved PD-1 ×
CTLA-4 bsAb targeting dual inhibitory immune checkpoints.
Its approval for the treatment of relapsed or metastatic
cervical cancer underscores the feasibility and potential of
bsAbs as a novel therapeutic approach.2
In patients with
recurrent or metastatic cervical cancer, frontline therapy
combining Cadonilimab with platinum-based chemotherapy
-with or without Bevacizumab-has demonstrated a
progression-free survival (PFS) advantage, successfully
meeting the primary endpoint of the Phase III AK104-303
trial (NCT04982237).8
Recently, Cadonilimab received approval in China for
treating patients with recurrent or metastatic cervical
cancer who had previously failed platinum-containing
chemotherapy.
Notably, findings from the Phase III COMPASSION-15 trial,
Vudalimab
In addition, another PD-1 × CTLA-4 bsAb drug, Vudalimab
(XmAb20717), exhibits increased affinity for PD-1/CTLA-4
dual-positive cells. Both pre-clinical and clinical findings
have shown manageable side effects.10 Furthermore, phase
II clinical trials of Vudalimab Alone or in combination with
Chemotherapy or targeted therapy for patients with
metastatic castration-resistant prostate cancer (mCRPC)
have been initiated.11
MEDI5752
MEDI5752, unlike Cadonilimab, is a distinct anti-PD-1 × CTLA-4
bsAb fusing an anti–PD-1 mAb and Tremelimumab's variable
binding domains on a DuetMab backbone, and preferentially
binds to PD-1+ T cells in the TME. This design is enhanced
with triple amino acid mutations in the human IgG1 constant
heavy chain to decrease Fc-mediated immune effector
functions.12 A phase II dose-escalation and dose-expansion
study indicated that MEDI5752 has promising antitumor
activity in patients with advanced RCC.2
Figure 2. The bsAbs targeting immunomodulatory checkpoints. The checkpoint-targeted bsAbs are mainly divided into three categories:
targeting dual inhibitory checkpoints (①); targeting co-stimulatory and inhibitory checkpoints (②); and targeting immunomodulatory
checkpoints and non-checkpoint targets (③④).2
PD-1
CTLA-4
TIM-3
LAG-3
1
2
Dual inhibitory
checkpoint bsAbs
Co-stimulatory and
inhibitory
checkpoint bsAbs
T cell
Tumor cell
TAA and
immunomodulatory
checkpoint bsAbs
PD-1
TGF- , VEGF
PD-L1
PD-L1
PD-1
CD137
CTLA-4
4
TAA
GITR
ICOS
CD137
OX40
PD-1
CTLA-4
3 Growth factors/cytokines
and immunomodulatory
checkpoint bsAbs
presented at the AACR Annual Meeting 2024, further
highlight Cadonilimab's potential. The trial demonstrated
that PD-1 × CTLA-4 bsAb, in combination with chemotherapy,
significantly improves both progression-free and overall
survival for patients with untreated, HER2-negative, locally
advanced or metastatic gastric or gastroesophageal
junction (GEJ) cancer. This benefit was observed even in
patients with PD-L1-low tumors, marking an improvement
over chemotherapy alone.9
Tebotelimab
The dual immunomodulator Tebotelimab (MGD013) targets
LAG-3 and PD-1, blocking PD-1 interactions with PD-L1 and
PD-L2 while also preventing LAG-3 from binding to MHC II.
Its DART structure enables synergistic T cell activation.12
Tebotelimab exhibits a more robust capacity for T cell
activation and cytokine secretion compared to individually
blocking PD-1 or LAG-3 signaling.15
A recent clinical study has preliminarily confirmed the
safety and efficacy of Tebotelimab as monotherapy or in
combination with Margetuximab—an anti-HER2 monoclonal
antibody—in patients with HER2-positive recurrent or
refractory advanced solid tumors.16
New progress in nanoantibodies
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 3 of 9
Figure 3. The structure and properties of Z15-0 nanobody13
Figure 4. Clinical trial design and patient flow16
Zeng et al engineered Z15-0-2, a bispecific nanobody
against PD-1 and CTLA-4, demonstrating marked antitumor
efficiacy in mice. They optimized its mRNA sequence to
enhance expression, positioning Z15-0-2 as a promising
candidate for advanced cancer immunotherapy. The
strategy, empolying lipid nanoparticles (LNP) for mRNA
delivery (LNP-mRNA), could surpass current immune
checkpoint inhibitors in efficacy.13
PD-1 x LAG-3 bsAbs
Lymphocyte activation gene-3 (LAG-3) monoclonal
antibodies (mAbs) represent the fifth class of immune
checkpoint inhibitors approved by the FDA for cancer
treatment.14 The co-expression of LAG-3 and PD-1 is closely
associated with exhausted effector T cells in ovarian cancer.
Blocking LAG-3 and PD-1 simultaneously has been shown to
enhance the cytotoxicity of CD8+ T cells, suggesting a
promising therapeutic strategy.2
Clinical investigations are underway for several bsAbs
targeting LAG-3 and PD-1 across various cancer types.
MTD/MAD
EOC (up to 40 pts)
Cervical (up to 16 pts)
TNBC (up to 40 pts)
NSCLC (up to 32 pts total,
16 CPI-naive + 16 post-CPI)
SCCHN (up to 32 pts total,
16 CPI-naive + 16 post-CPI)
SCLC (up to 16 pts)
Cholangio (up to 16 pts)
DLBCL (up to 20 pts, of
which 10 post-CAR-T)
HER2+
locally advanced
or metastatic solid
tumors
HER2+
breast
cancer
(up to 30 pts)
HER2+
GC/GEJ
cancer (4 pts)
Other HER2 +
cancer
(up to 30 pts)
Tebotelimab 300 mg IV
+
margetuximab 15 mg kg –1
IV (both Q3W)
1,200 mg
800 mg
400 mg
120 mg
30 mg
10 mg a
3 mg a
1 mg a
Tebotelimab 600 mg IV
+
margetuximab 15 mg kg –1
IV (both Q3W)
Separate HC C escalation:
120 mg →40 0 mg →600 mg (Q2W)
3+3 design
Flat dosing Q2W:
single-patient cohorts
followed by 3+3 design
3+3 dose escalation
Dose escalation (tebotelimab 1–1,200 mg IV Q2W) Cohort expansion
(tebotelimab 600 mg IV Q2W) Dose escalation
Previously treated
unresectable, locally
advanced or metastatic
solid tumors of any
histology
Tebotelimab monotherapy (Q2W) Combination of tebotelimab and margetuximab (Q3W)
Cohort expansion
(tebotelimab
600 mg IV Q3W +
margetuximab
15 mg kg –1
IV Q3W)
MTD/MAD
Treatment discontinuation ( n = 53)
• PD (n = 39)
• Adverse event ( n = 10)
• Death (n = 2)
• Withdrawal by subject ( n = 0)
• Physician decision ( n = 1)
• Completed ( n = 0)
• Other ( n = 1)
Tebotelimab monotherapy enrollment ( n = 277)
Dose escalation ( n = 54)
Not treated ( n = 1) Not treated ( n = 7)
Cohort expansion ( n = 223)
Evaluable patients
Safety (n = 53)
Response (n = 44)
Evaluable patients
Safety (n = 216)
Response (n = 181)
Treatment ongoing ( n = 0) Treatment ongoing n = 4)
Combination tebotelimab + margetuximab enrollment ( n = 89)
Dose escalation ( n = 20)
Not treated ( n = 0) Not treated ( n = 5)
Cohort expansion ( n = 69)
Evaluable patients
Safety (n = 20)
Response (n = 17)
Evaluable patients
Safety (n = 64)
Response (n = 55)
Treatment ongoing ( n = 1) Treatment ongoing ( n = 8)
Treatment discontinuation ( n = 212)
• PD (n = 153)
• Adverse event (n = 28)
• Death (n = 11)
• Withd rawal by subject ( n = 8)
• Physician decision ( n = 5)
• Completed ( n = 4)
• Other ( n = 3)
Treatment discontinuation ( n = 19)
• PD (n = 14)
• Adverse event (n = 3)
• Death (n = 0)
• Withd rawal by subject ( n = 1)
• Physician decision ( n = 1)
• Completed ( n = 0)
• Other ( n = 0)
Treatment discontinuation ( n = 57)
• PD (n = 46)
• Adverse event (n = 5)
• Death (n = 1)
• Withd rawal by subject ( n = 3)
• Physician decision ( n = 1)
• Completed ( n = 0)
• Other ( n = 1)
FS118
FS118, targeting LAG-3 and PD-L1, exhibits potential in
activating exhausted immune cells and overcoming resistance
to PD-L1 blockade. By simultaneously binding to LAG-3 and
PD-L1, FS118 disrupts key interactions such as PD-1/PD-L1,
CD80/PD-L1, and LAG-3/MHC-II, effectively reversing T cell
suppression and promoting cytokine production in CD4+
and CD8+ T cells.17 In colorectal cancer (CRC) syngeneic
mouse models, FS118 enhances the anti-tumor immune
response and reduces LAG-3 expression on T cells,
indicating its potential as an effective immunotherapy.2
ABL501
ABL501, a bsAb targeting LAG-3 and PD-L1, promotes
dendritic cell maturation by inhibiting PD-L1 signaling,
enhancing CD8+ T cell cytotoxicity.18 In a humanized mouse
model, ABL501 shows a dose-d ependent anti-tumor effect
by rejuvenating immune cells.19 Additionally, bispecific
antibodies targeting LAG-3 × CTLA-4, such as XmAb22841,
are also under clinical investigation.
RG7769
RG7769, a heterodimeric TIM-3 × PD-1 bsAb developed by
Roche, has a high affinity for PD-1 and low affinity for TIM-3.
It selectively targets PD-1+ T cells and PD-1+ TIM-3+ T cells,
enhancing IFN-γ secretion and boosting the anti-tumor
activity of tumor-infiltrating T cells.22 A Phase II study
(NCT04785820) is comparing RG7769 with Nivolumab in
advanced esophageal squamous cell carcinoma. Another
early-phase study on a TIM-3 × PD-L1 bsAb was terminated
due to unexpected drug immunogenicity issues.23
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 4 of 9
TIM-3 × PD-1
TIM-3 (T-cell immunoglobulin and mucin domain 3) is a key
co-inhibitory receptor expressed on various immune cells
Figure 5. The ligands for LAG-3, TIM-3, TIGIT and their interactions14
and frequently co-expressed with PD-1. This co-expression,
notably in tumor-infiltrating lymphocytes (TILs), results in
reduced T cell activity and cytokine production, aiding in
immune dysfunction and tumor immune escape.14 Several
ligands for TIM-3 have been identified, including Galectin-9
(Gal-9), high-mobility group protein B1 (HMGB1),
phosphatidylserine (PtdSer), and carcinoembryonic antigen
cell adhesion molecule 1 (CEACAM-1).14 Galectin-9 is the
primary ligand for TIM-3, promoting cell death in
TIM-3-expressing T cells.20 TIM-3 upregulation on regulatory
T cells contributes to T cell exhaustion, characterized by
functional impairment and altered gene expression.21
IC Ligands Expression Mechanism of action
LAG-3
MHC-II B cells, MON-Mø, DCs, some activated T cells
Tim-3
TIGIT
Negatively regulates T cell responses
CD155 DCs, T cells, B cells, macrophages Increasing the IL-10 secretion
CD113 Liver, testes, lungs, placenta, and kidneys Inhibition of T cell and NK cell activity
CD112 Hematopoietic and non-hematopoietic tissues Inhibiting the activation of T cells and NK cells
Nectin4 Tumor cells Inhibiting NK cell activity
Fap2 Tumor cells Inhibiting NK cell toxicity and T cell activity
FGL-1 FGL-1 protein is primarily secreted from
hepatocytes Inhibiting antitumor immune responses
α-synuclein Neurons, heart, muscle, and other tissues
Gal-3 Inhibiting antitumor T cell responses Tumor cells, macrophages, epithelial cells,
fibroblasts, activated T cells
LSECtin Inhibiting antitumor T cell responses Liver, tumor-associated macrophages,
and other tumor tissues
Gal-9 APC, MDSCs, Naive CD4 T cells, plasma
PtdSer Released from apoptotic cells
LAG-3 can recognize α-synuclein fibrils and
affect its endocytosis and intercellular
transmission, contributing to PD
Gal-9 mainly induces calcium to flow into
the intracellular area of Th1 cells and
induces apoptosis
PtdSer and TIM-3 binding contributes to the
clearance of apoptotic bodies and antigen
cross-presentation by Tim-3+ DCs
CEACAM-1 DCs, monocytes, macrophages, and
activated T cells
CEACAM-1/TIM-3 complex formation has a
crucial role in regulating autoimmunity and
antitumor immunity
HMGB1 Proliferating tissues or estrogen stimulated
cancer cells
Blocking activation and suppresses innate
immune responses to nucleic acids
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 5 of 9
TIGIT × PD-L1
TIGIT (T cell immunoreceptor with Ig and ITIM domains), an
inhibitory IgSF checkpoint, impedes immune cell activation
by competing with essential co-stimulatory receptors like
CD226 and CD69. TIGIT interacts with ligands such as PVR
and PVRL2 on tumor, T, and antigen-presenting cells (APCs).
The approval of Tiragolumab, an anti-TIGIT antibody
combined with Atezolizumab for NSCLC with high PD-L1
expression, highlights TIGIT's potential as a target that can
enhance the effectivenessof other ICIs.24
A bsAb targeting TIGIT and PD-L1 not only elevates IL-2
secretion but also improves survival rates in preclinical
models.25 Additionally, PVRIG, another immune checkpoint
that independently binds to PVRL2, plays a complementary
inhibitory role. A human bsAb targeting both TIGIT and
PVRIG has shown strong anti-tumor activity.26,27
Several bsAbs, including HLX301 and PM1022, which target
TIGIT and PD-L1, are currently in phase I clinical trials.25-28
GITR(L) × PD-1 or GITR × CTLA-4
As a member of the TNFR superfamily (TNFRSF),
glucocorticoid-induced TNF receptor (GITR) acts as a
co-stimulatory checkpoint, promoting T cell activation and
counteracting regulatory T cell (Treg) immunosuppression.2
Research reveals that combining anti-GITR antibodies with
anti-PD-1 mAbs reduces TIGIT expression and regulating
tumor-infiltrating T cells.29 This study paves the way for
exploring bispecific constructs targeting GITR and other
immune checkpoints. To overcome resistance, a multimeric
GITR ligand is combined with an anti-PD-1 mAb to create a
4-1BB × PD-L1
4-1BB, also known as CD137 or TNFRSF9, plays a crucial role
in activating and expanding T cells. Clinical trials have
examed the safety and efficacy of 4-1BB agonistslike
Urelumab and Utolimumab.2
However, the use of these
agonists alone can lead to severe toxicities, limiting the
development of 4-1BB monoclonal antibodies.5
A bispecific construct could potentially mitiage this issue
by selectively regulating the stimulatory effects of 4-1BB
agonists in specific immune cells, offering a way to reduce
toxicity while enhance therapeutic outcomes.2
Simultaneously targeting co-stimulatory and
inhibitory checkpoints bsAbs
ICIs rely on pre-existing immune responses, making the
combination of co-stimulatory agonists and checkpoint
inhibitors pivotal for enhancing therapeutic outcomes.
BsAbs bridge tumor and immune cells, restoring T cell
functions for improved clinical efficacy while minimizing
co-stimulation-indued side effects . This strategy, targeting
checkpoints like 4-1BB, OX40, and ICOS, marks a
transformative advancement in cancer immunotherapy.2
Immune co-stimulation primilary revolves around the
B7-CD28 and TNFR families. CD28 and ICOS are key
co-stimulatory receptors in the B7-CD28 family, while
OX40, CD40, CD27, 4-1BB, GITR, and CD30 are part of the
TNFR family.12
ABL503
By utilizing in trans cell bridging and modifying the Fc
region, bsAb such as ABL503 (PD-L1 × 4-1BB) can enhance
targeting precision while minimizing systemic side effects.
Preclinical studies have shown that ABL503 is well-tolerated,
with low risk of liver toxicity, and demonstrates superior
activity compared to a combination of the corresponding
monoclonal antibodies.32
GEN1046
BNT311/GEN1046 (Acasunlimab) is an investigational PD-L1 ×
4-1BB bsAb that combines Genmab's proprietary DuoBody®
technology and BioNTech’s immunomodulatory antibodies.³³
By conditionally activates 4-1BB on T cells and NK cells, it
triggers an anti-tumor response, inhibiting tumor growth by
enhancing T cell proliferation and the secretion of cytokines
such as IFN-γ, IL-10, and CXCL10.2
In a Phase II trial evaluating Acasunlimab, alone or in
combination with Pembrolizumab in patients with
previously treated metastatic non-small cell lung cancer
(mNSCLC), results indicated that the combination of
GEN1046 with Pembrolizumab demonstrated a manageable
safety profile and promising efficacy. These findings were
presented at the 2024 American Society of Clinical
Oncology (ASCO) Annual Meeting.33
Figure 6. The data of Acasunlimab in ASCO 202433
bispecific construct, enhancing T cell infiltration while
reducing the presence of Treg cells and exhausted T cells.30
Beyond PD-1, CTLA-4 is another inhibitory checkpoint
considered for dual targeting. ATOR114, a GITR × CTLA-4
bsAb, enhances T and NK cell activation, depletes Treg
cells and GITR+ tumor cells. This promising dual-targeting
approach awaits validation of its efficacy in clinical trials.31
Unconfirmed
ORR
Arm A: mono 31%
Arm B: Acasunlimab + low-dose Pembro 25%
30%
Unconfirmed
DCR
50%
65%
75%
Confirmed
ORR
13%
21%
22%
0
18%
33%
mDoR 6-mth PFS
2 mth
6 mth
Arm C: Acasunlimab + high-dose Pembro NR
Note:
Arm A, 100 mg Q3W x 2 cycles then 500 mg Q6W Arm B, 100 mg + pembro 200 mg Q3W Arm C, 100 mg + pembro 400 mg Q6W
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 6 of 9
Moreover, a number of 4-1BB × PD-L1 bispecific antibodies,
such as ATG-101 (NCT04986865), INBRX-105(NCT03809624)
and QLF31907 (NCT05150405), are presently undergoing
phase I clinical trials to assess their effectiveness in
individuals with advanced solid tumors.2
OX40 × PD-L1/CTLA-4
OX40, also known as CD134, is transiently expressed on
CD4+ and CD8+ T cells following T cell receptor activation,
while it is constitutively expressed on intra-tumoral Treg
cells.2
In mouse models of ovarian cancer, combining PD-1
blockade with OX40 stimulation has been shown to
enhance immune cell infiltration and elicit a tumor-specific
T-cell response, highlighting the potential of combining
OX40 with other immune checkpoints.34
KN-052, developed by Jiangsu Alphamab, is an IgG-like
bispecific antibody targeting both OX40 and PD-L1. It
demonstrates robust binding to both targets, inhibits
PD-L1/PD-1 and PD-L1/CD80 interactions, and enhances
immune responses in preclinical models. This leads to
increased tumor growth inhibition in mouse models of
colon cancer, with promising pharmacokinetics observed in
cynomolgus monkeys.35
ICOS × PD-L1
ICOS, a member of the IgSF, provides a stimulatory signal
linked to TCR activation and is consistently expressed in
Treg cells. KY105, a novel ICOS × PD-L1 IgG1 bsAb, activates
ICOS in a PD-L1-dependent manner. In murine models of
colorectal cancer, KY105 has shown promising anti-tumor
efficacy by reducing ICOS+ Treg cells and enhancing IFN-γ
secretion.2
Targeting immune checkpoints and TAAs
Targeting Immune Checkpoints and GFs/Cytokines
The lack of tumor specificity in ICIs monotherapy can lead
to nonspecific targeting of tumor cells and unpredictable
irAEs. Tumor-associated antigens (TAAs) serve as a
"navigation system," directing immune cells within the
tumor microenvironment (TME) while sparing normal
tissues. Combining TAAs with immunomodulatory
checkpoints presents a potential solution to these
challenges.2
By linking immune cells with tumor cells that
express high levels of TAAs, bsAbs can activate immune
cells' anti-tumor responses and induce apoptosis in tumor
cells, effectively combating tumors while minimizing
irAEs.25
Extensively studied TAAs such as EpCAM, EGFR, and HER2
have been customized for specificity in bispecific
molecules.38 EpCAM, present in various normal epithelial
tissues, also serves as a biomarker for cancer stem cells. A
CD40 × EpCAM bsAb has been engineered to activate
dendritic cells (DCs), enhancing the priming of
tumor-specific T cells and extending overall survival in
murine models.39
Receptor tyrosine kinases belonging to the ErbB family
(EGFR, HER2, HER3, HER4) can facilitate tumor initiation and
invasion when aberrantly activated.40 A recently developed
symmetrical bsAb targeting EGFR and PD-1 has shown
promising results. This anti-PD1 × EGFR bispecific antibody
not only inhibits EGFR signaling but also elicits
antibody-dependent cellular cytotoxicity (ADCC) in tumor
cells, leading to tumor regression in xenograft and
syngeneic colorectal cancer models.41
Beyond bsAbs that crosslink TAAs and checkpoints, several
bsAbs targeting growth factors (GFs) and cytokines are
currently undergoing clinical trials.
With a unique design, the bsAb targeting OX40 and CTLA-4
depletes Tregs within the tumor microenvironment while
activating effector and memory T cells.36
ATOR-1015, a bsAb targeting OX40 and CTLA-4, depletes
Tregs in tumors, activates effector T cells, and shows
promise in early-phase trials for advanced solid tumors,
reducing tumor growth while improving survival across
various cancer models.37
The specificity of pro-tumor growth factors and cytokines
can reinforce tumor-killing effects while reducing drug
resistance.2
These factors modulate the immunosuppressive
TME by influencing immune cell differentiation and
function. For instance, TGF-β can drive naive T cells to
become Tregs, independently of IL-6, thus hampering
anti-tumor immunity.2
Numerous GF and cytokine-targeted drugs have been
approved for clinical use, either alone or in combination
with ICIs. For example, combined anti-VEGF and antiPD-1/PD-L1 therapies have demonstrated enhanced patient
responses across various cancers.42
Integrating GFs and cytokines into immune checkpoint
-targeted bsAbs not only enhances immunotherapy
efficacy but also directs these agents precisely to tumor
sites.² Bispecific antibodies targeting checkpoints and
GFs/cytokines, such as YM101 (PD-L1 × TGF-β) and AK112 (PD-1 ×
Figure 6. The structure of KN052 (Source: the website of Jiangsu
Alphamab)
Targeting Immune Checkpoints and
Non-checkpoint Targets BsAb
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 7 of 9
The development of bsAbs targeting immunomodulatory
checkpoints represents a transformative approach in
cancer therapy, offering a promsing alternative to
traditional monclonal antibody treatments. These bsAbs
provide several key advantages, including he ability to
block two distinct inhibitory signals, activate s specific
immune cells within the immunosuppressive TME, and
deliver localized stimulatory or inhibitory effects in
specific tumor tissues. These characteristics have the
potential to yield more effective and safer treatment
options for cancer patients.
One of the central strengths of bsAbs lies in their
capacity to deliver both inhibitory and stimulatory
signals. By targeting two immune checkpoints
simultaneously, bsAbs can enhance T cell activation
while overcoming T cell exhaustion, leading to a more
potent anti-tumor response. This dual blockade not only
VEGF), are in development, offering potential to overcome
immune tolerance in the TME.2
In a Phase I study, AK112 exhibited an overall response rate
(ORR) of 23.5% in patients with platinum-resistant/refractory
epithelial ovarian cancer, with favorable safety profiles.43
The combination of AK112 with chemotherapy yielded
encouraging clinical benefits, including improved
progression-free survival (PFS) and ORR, with manageable
toxicity in NSCLC patients.44
TGF-β initially suppresses tumor growth but can later
contribute to drug resistance and metastasis. Studies
indicate that heightened TGF-β activity can reduce the
effectiveness of anti-PD-1/PD-L1 therapies. Combining
TGF-β blockade with PD-1/PD-L1 inhibition has shown
synergistic effects.45 YM101 promotes antigen presentation,
enhances T cell infiltration, and boosts tumor cell
destruction. It exhibits superior anti-tumor activity
compared to individual anti-TGF-β or anti-PD-L1 treatments
in both in vitro and in vivo models.46
DISCUSSION
restores T cell function but also promotes a more
favorable balance between effector T cells and Tregs,
which is essential for sustaining anti-tumor immunity
over time.
Moreover, bsAbs offer precision by exerting localized
effects in specific tumor tissues, which can minimize
systemic exposure and reduce the risk of adverse
effects commonly associated with immune checkpoint
inhibitors. This targeted approach enables enhanced
therapeutic efficacy while improving safety profiles. For
example, innovative bsAbs like ABL503 and GEN1046,
designed to modulate the activity of 4-1BB and PD-L1,
demonstrate how these agents can selectively enhance
T cell responses while mitigating liver toxicity and other
systemic side effects.
Emerging clinical data underscores the potential of bsAbs
to redefine oncology treatment paradigms. Ongoing
clinical trials suggests bsAbs, when used in combination
with established therapies such as chemotherapy and
targeted therapies, may offer synergistic benefits,
particularly for patients with advanced cancers who
have exhausted standard treatment options. The
combination of bsAbs with other modalities could
significantly enhance overall treatment outcomes.
However, challenges remain in the clinical development
of bsAbs. The complex design , the need for precise
dosing regimens, and potential immunogenicity require
careful evaluation in clinical trials. Additionally,
identifying the optimal patient populations who are
most likely benefit from bsAbs will be critical to
maximizing their therapeutic potential.
In summary, bsAbs targeting immunomodulatory
checkpoints represent a highly promising frontier in
cancer immunotherapy. Their ability to block multiple
inhibitory signals, deliver targeted immune activation,
and minimize systemic side effects positions them as a
potentially safer and more effective alternative to
conventional therapies. Continued research and clinical
trials will be critical in fully elucidating the capabilities of
bsAbs and to optimizing their integration into the
current cancer treatment landscape .
PRODUCT RECOMMENDATIONS
Comprehensive Related
Products for Immuno-Oncology Therapy
Immune Checkpoint
Immune checkpoint proteins Cytokine targets Agonist target proteins
PK assay kits Stable / Reporter cell lines Inhibitor screening kits
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 8 of 9
References
1 Meng L, Wu H, Wu J,et al.Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer
hallmarks.Cell Death Dis.2024;15:3.
2 Zhang T, Lin Y, Gao Q. Bispecific antibodies targeting immunomodulatory checkpoints for cancer therapy. Cancer Biol Med. 2023 Mar24;20
(3):181–95.
3 Hosseini SS, Khalili S, Baradaran B, et al. Bispecific monoclonal antibodies for targeted immunotherapy of solid tumors: Recent advances
and clinical trials. Int J Biol Macromol. 2021 Jan 15;167:1030-1047.
4 Thakur A, Huang M, Lum LG. Bispecific antibody based therapeutics: Strengths and challenges. Blood Rev. 2018 Jul;32(4):339-347.
5 Sun Y, Yu X, Wang X, et al. Bispecific antibodies in cancer therapy: Target selection and regulatory requirements. Acta Pharm Sin B. 2023
Sep;13(9):3583-3597.
6 Shum E, Reilley M, Najjar Y, et al. 523 preliminary clinical experience with XmAb20717, a PD-1 × CTLA-4 bispecific antibody, in patients with
advanced solid tumors. J Immunother Cancer. 2021; 9: A553-A553.
7 Dovedi SJ, Elder MJ, Yang C, et al. Design and efficacy of a monovalent bispecific PD-1/CTLA4 antibody that enhances CTLA4 blockade on
PD-1(+) activated T cells. Cancer Discov. 2021; 11: 1100-1117.
8 Akeso, Inc. "Cadonilimab (PD-1/CTLA-4 Bispecific Antibody) Met Primary Endpoint of Progression-Free Survival in Phase III Trial for
First-Line Treatment of Recurrent/Metastatic Cervical Cancer in All-Comer Patients." News Release. November 27, 2023. Accessed.
9 The ASCO Post Staff. "Bispecific Immune Checkpoint Inhibitor Improves Survival in Patients With Gastric Cancer, Regardless of PD-L1
Status." The ASCO Post, April 9, 2024. Accessed April 22, 2024.
10 Hickingbottom B, Clynes R, Desjarlais J, et al. Preliminary safety and pharmacodynamic (PD) activity of XmAB20717, a PD-1 × CTLA-4
bispecific antibody, in a phase I dose escalation study of patients with selected advanced solid tumors. J Clin Oncol. 2020; 38:
e15001-e15001.
11 UChicago Medicine. "XmAb®20717 (Vudalimab) Alone or in Combination With Chemotherapy or Targeted Therapy in Patients With
Metastatic Castration-Resistant Prostate Cancer." Clinical Trial NCT05005728, UChicago Medicine.
12 Wei J, Yang Y, Wang G, et al. Current landscape and future directions of bispecific antibodies in cancer immunotherapy. Front Immunol.
2022 Oct 28;13:1035276.
13 Zeng J, Fang Y, Zhang Z, et al. Antitumor activity of Z15-0-2, a bispecific nanobody targeting PD-1 and CTLA-4. Oncogene. 2024 Jul;43
(29):2244-2252.
14 Cai L, Li Y, Tan J, et al. Targeting LAG-3, TIM-3, and TIGIT for cancer immunotherapy. J Hematol Oncol. 2023;16:101.
15 LaMotte-Mohs R, Shah K, Smith D, et al. Abstract 3217: MGD013, a bispecific PD-1 × LAG-3 dual-affinity re-targeting (DART®) protein with
T-cell immunomodulatory activity for cancer treatment. Cancer Res. 2016; 76: 3217-3217.
16 Luke JJ, Patel MR, Blumenschein GR, et al. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic
cancers: a phase 1 trial. Nat Med. 2023 Nov;29(11):2814-2824.
17 Kraman M, Faroudi M, Allen NL, et al. FS118, a bispecific antibody targeting LAG-3 and PD-L1, enhances T-cell activation resulting in potent
antitumor activity. Clin Cancer Res. 2020;26(13):3333–3344.
18 Sung E, Ko M, Won JY, Jo Y, Park E, Kim H, et al. LAG-3xPD-L1 bispecific antibody potentiates antitumor responses of T cells through
dendritic cell activation. Mol Ther. 2022; 30: 2800-2816.
19Park E, Kim H, Sung E, et al. Abstract 1633: ABL501, PD-L1 × LAG-3, a bispecific antibody promotes enhanced human T cell activation through
targeting simultaneously two immune checkpoint inhibitors, LAG-3 and PD-L1. Cancer Res. 2021; 81: 1633-1633.
20 Solinas C, De Silva P, Bron D, et al. Significance of TIM3 expression in cancer: from biology to the clinic. Semin Oncol. 2019;46(4-5):372-379.
21 Friedlander A, Addeo A, Banna G. New emerging targets in cancer immunotherapy: the role of TIM3. ESMO Open. 2019;4(suppl 3):e000497.
22 Deak LLC, Seeber S, Perro M, et al. Abstract 2270: RG7769 (PD1-TIM3), a novel heterodimeric avidity-driven T cell specific PD-1/TIM-3
bispecific antibody lacking Fc-mediated effector functions for dual checkpoint inhibition to reactivate dysfunctional T cells. Cancer Res.
2020; 80: 2270-2270.
23Hellmann MD, Bivi N, Calderon B, et al. Safety and immunogenicity of LY3415244, a bispecific antibody against TIM-3 and PD-L1, in patients
with advanced solid tumors. Clin Cancer Res. 2021; 27: 2773-2781.
24 Rodriguez-Abreu D, Johnson ML, Hussein MA, et al. Primary analysis of a randomized, double-blind, phase II study of the anti-TIGIT
antibody tiragolumab (tira) plus atezolizumab (atezo) versus placebo plus atezo as first-line (1L) treatment in patients with PD-L1-selected
NSCLC (CITYSCAPE). J Clin Oncol. 2020; 38: 9503-9503.
25 Mu S, Liang Z, Wang Y, et al. PD-L1/ TIGIT bispecific antibody showed survival advantage in animal model. Clin Transl Med. 2022; 12: e754.
59.
26 Zeng T, Cao Y, Jin T, et al. The CD112R/CD112 axis: a breakthrough in cancer immunotherapy. J Exp Clin Cancer Res. 2021; 40: 285. 60.
27 Dai S, Huang W, Yuan Z, et al. Abstract 5525: An Fc-competent bispecific antibody targeting PD-L1 and TIGIT induces strong immune
responses and potent anti-tumor efficacy. Cancer Res. 2022; 82: 5525-5525.
W���� P����n� �n� I�n�va�i��
A�v�n�� Bi��e�i����
techsupport@acrobiosystems.com
1 Innovation Way, Newark, DE 19711
+1 800-810-0816 (US / Canada)
+86 400-682-2521 (Asia & Pacific)
Bispecific Antibodies Targeting Immunomodulatory Checkpoints for Cancer Therapy Page 9 of 9
About ACROBiosystems
ACROBiosystems Group, founded in 2010 and listed in 2021, is a biotechnology company
aimed at becoming a cornerstone of the global biopharmaceutical industry by providing
life science tools and business model innovation. ACROBiosystems spans across the globe
and maintains over twelve offices, research & development centers, and production bases
across the United States, Europe, and Asia. Numerous partnerships with well-known
enterprises such as Pfizer, Novartis, and other academic institutes have been established.
Through the continuous development of new technologies and tools, ACROBiosystems is
dedicated towards helping their customers overcome challenges from drug discovery to
commercialization through innovative life science tools and solutions.
28 Zhang J, Guo Y. A phase I study to evaluate the tolerance, safety, pharmacokinetic characteristics and preliminary efficacy of PM1022 in
patients with advanced tumors and a phase IIA study to investigate the efficacy of PM1022 in patients with advanced tumors.
29Wang B, Zhang W, Jankovic V, et al. Combination cancer immunotherapy targeting PD-1 and GITR can rescue CD8(+) T cell dysfunction and
maintain memory phenotype. Sci Immunol. 2018; 3: eaat7061.
30 Chan S, Belmar N, Ho S, et al. An anti-PD-1-GITR-L bispecific agonist induces GITR clusteringmediated T cell activation for cancer
immunotherapy. Nat Cancer. 2022; 3: 337-354.
31 Fritzell S, Levin M, Åberg I, et al. ATOR-1144 is a tumor-directed CTLA-4 × GITR bispecific antibody that acts by depleting Tregs and
activating effector T cells and NK cells. Cancer Res. 2019; 79: 4077.
32Herrera M, Pretelli G, Desai J, et al. Bispecific antibodies: advancing precision oncology,Trends in Cancer,2024,ISSN 2405-8033.
33 BioNTech SE. "Investigational AcaSunLimab (Duobody® PD-L1x4-1BB) Combination." BioNTech SE News Release.
34 Guo Z, Wang X, Cheng D, et al. PD-1 blockade and OX40 triggering synergistically protects against tumor growth in a murine model of
ovarian cancer. PLoS One. 2014; 9: e89350.
35Geng Q, Jiao P. Anti-PD-L1-Based Bispecific Antibodies Targeting Co-Inhibitory and Co-Stimulatory Molecules for Cancer Immunotherapy.
Molecules. 2024 Jan 17;29(2):454.
36 Perez-Santos M, Anaya-Ruiz M, Herrera-Camacho I, et al. Bispecific anti-OX40/CTLA-4 antibodies for advanced solid tumors: a patent
evaluation of WO2018202649. Expert Opin Ther Pat. 2019; 29: 921-924.
37 Farhangnia P, Ghomi SM, Akbarpour M, et al. Bispecific antibodies targeting CTLA-4: game-changer troopers in cancer immunotherapy.
Front Immunol. 2023 Jun 27;14:1155778.
38 Munz M, Baeuerle PA, Gires O. The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res. 2009; 69: 5627-5629.
39 Ellmark P, Hägerbrand K, Levin M, et al. 858 a bispecific antibody targeting CD40 and EpCAM induces superior anti-tumor effects
compared to the combination of the monospecific antibodies. J Immunother Cancer. 2020; 8: A511.
40 Kumagai S, Koyama S, Nishikawa H. Antitumour immunity regulated by aberrant expression of PD-L1. Nat Rev Drug Discov. 2021;21:440-462.
41 Li L, Deng L, Meng X, et al. Tumor-targeting anti-EGFR × anti-PD1 bispecific antibody inhibits EGFR overexpressing tumor growth by
combining EGFR blockade and immune activation with direct tumor cell killing. Transl Oncol. 2021; 14: 100916.
42 Petroni G, Buque A, Coussens LM, et al. Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat
Rev Drug Discov. 2022; 21: 440-462.
43Coward J, Mislang ARA, Frentzas S, et al. Safety and efficacy of AK112, an anti-PD-1/VEGF-A bispecific antibody, in patients with advanced
solid tumors in a phase I dose escalation study. J Clin Oncol. 2021; 39: 2515.
44 Zhao Y, Fang W, Yang Y, et al. A phase II study of AK112 (PD-1/VEGF bispecific) in combination with chemotherapy in patients with
advanced non-small cell lung cancer. J Clin Oncol. 2022; 40:9019.
45 Chen X, Wang L, Li P, et al. Dual TGF-beta and PD-1 blockade synergistically enhances MAGE-A3-specific CD8+ T cell response in
esophageal squamous cell carcinoma. Int J Cancer. 2018; 143: 2561-74.
46 Yi M, Zhang J, Li A, et al. The construction, expression, and enhanced anti-tumor activity of YM101: a bispecific antibody simultaneously
targeting TGF-beta and PD-L1. J Hematol Oncol. 2021; 14: 27.
Brought to you by
Download the Whitepaper for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version