ArticlePDF Available

Preparation and characterization of a novel nanobody against T-cell immunoglobulin and mucin-3 (TIM-3)

Authors:

Abstract

Objective(s) As T-cell immunoglobulin and mucin domain 3 (TIM-3) is an immune regulatory molecule; its blocking or stimulating could alter the pattern of immune response towards a desired condition. Based on the unique features of nanobodies, we aimed to construct an anti-TIM-3 nanobody as an appropriate tool for manipulating immune responses for future therapeutic purposes. Materials and Methods We immunized a camel with TIM-3 antigen and then, synthesized a VHH phage: mid library from its B cell’s transcriptome using nested PCR. Library selection against TIM-3antigen was performed in three rounds of panning. Using phage-ELISA, the most reactive colonies were selected for sub-cloning in soluble protein expression vectors. The Nanobody was purified and confirmed with a nickel-nitrilotriacetic acid (Ni-NTA) column, SDS-PAGE and Western blotting. A flowcytometric analysis was performed to analyze the binding and biologic activities of theTIM-3 specific nanobody with TIM-3 expressing HL-60 and HEK cell lines. Results Specific 15kD band representing for nanobody was observed on the gel and confirmed with Western blotting. The nanobody showed significant specific immune-reactivity against TIM-3 with a relatively high binding affinity. The nanobody significantly suppressed the proliferation of TIM-3 expressing HL-60 cell line. Conclusion Finally, we successfully prepared a functional anti-humanTIM-3 specific nanobody with a high affinity and an anti-proliferative activity on an AML cell line in vitro.
Iranian Journal of Basic Medical Sciences
ijbms.mums.ac.ir
Preparation and characterization of a novel nanobody against
T-cell immunoglobulin and mucin-3 (TIM-3)
Vida Homayouni 1, Mazdak Ganjalikhani-hakemi 1*, Abbas Rezaei 1, Hossein Khanahmad 2,
Mahdi Behdani 3, Fatemeh Kazemi Lomedasht 3
1 Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
2 Genetic Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
3 Biotechnology Research Center, Biotechnology Department, Venom & Bio-therapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
A R T I C L E I N F O
A B S T R A C T
Article type:
Original article
Objective(s): As T-cell immunoglobulin and mucin domain 3 (TIM-3) is an immune regulatory
molecule; its blocking or stimulating could alter the pattern of immune response towards a
desired condition. Based on the unique features of nanobodies, we aimed to construct an anti-TIM-
3 nanobody as an appropriate tool for manipulating immune responses for future therapeutic
purposes.
Materials and Methods: We immunized a camel with TIM-3 antigen and then, synthesized a VHH
phagemid library from its B cell’s transcriptome using nested PCR. Library selection against TIM-
3antigen was performed in three rounds of panning. Using phage-ELISA, the most reactive
colonies were selected for sub-cloning in soluble protein expression vectors. The Nanobody was
purified and confirmed with a nickel-nitrilotriacetic acid (Ni-NTA) column, SDS-PAGE and Western
blotting. A flowcytometric analysis was performed to analyze the binding and biologic activities of
theTIM-3 specific nanobody with TIM-3 expressing HL-60 and HEK cell lines.
Results: Specific 15kD band representing for nanobody was observed on the gel and confirmed
with Western blotting. The nanobody showed significant specific immune-reactivity against TIM-3
with a relatively high binding affinity. The nanobody significantly suppressed the proliferation of
TIM-3 expressing HL-60 cell line.
Conclusion: Finally, we successfully prepared a functional anti-humanTIM-3 specific nanobody
with a high affinity and an anti-proliferative activity on an AML cell line in vitro.
Article history:
Received: May 15, 2016
Accepted: Jun 30, 2016
Keywords:
Antibody
Heavy chain antibody
Nanobody
Phage display
T-cell immunoglobulin and -
mucin domain 3
Please cite this article as:
Homayouni V, Ganjalikhani-hakemi M, Rezaei A, Khanahmad H, Behdani M, Kazemi Lomedasht F. Preparation and characterization of a
novel nanobody against T-cell immunoglobulin and Mucin-3 (TIM-3).
Iran J Basic Med Sci 2016; 19:1201-1208.
Introduction
The human TIM family members (TIM-1, TIM-3,
TIM-4) are commonly expressed as surface
glycoproteins containing an IgV domain, a mucin stalk,
a transmembrane domain, and a cytoplasmic tail (1, 2).
The gene encoding TIM-3 is known as HAVCR2, which
denotes hepatitis A virus (HAV) cellular receptor 2 (3).
TIM-3 has important and complex roles in the
regulation of immune responses. TIM-3 was originally
identified as surface molecule on CD4 effector Th1 cells
(4, 5). Now, it is known that TIM-3 is also expressed on
dendritic cells, microglia, macrophages, mast cells; NK
cells, activated and exhausted CD8 T cells (4, 6, 7).
The exhausted CD8T cells are generated as a
consequence of prolonged immune responses to
chronic infections and tumors (8). These cells are
characterized by a failure to proliferate and to exert
their effector functions such as cytotoxicity and
cytokine secretion in response to antigen stimulation
(9). Stimulation of TIM-3 by its ligand, galectin-9,
results in Th1 cell death, implicating a role for TIM-3 in
negatively regulating Th1 responses. Blocking of TIM-3
has been shown to increase IFN-γ secreting T cells,
mediating the pathophysiology of Th1-driven
autoimmune diseases. TIM-3 expressed on
macrophages and monocytes is implicated in
phagocytosis of apoptotic cells and cross-presentation
(10). In tumors, TIM-3 expression is induced on innate
immune cells, leading to a suppression of innate
responses to nucleic acids released from apoptotic
tumor cells by interacting with HMGB1. As discussed,
TIM-3 interacts with galectin-9 (Gal 9) and causes
exhaustion and apoptosis of antigen-specific Th1 cells
and CTLs, which correlates with impaired antitumor
immune response (11). Blocking of TIM-3 interaction
with its ligand would restore effector functions of
exhausted CD8 T cells and its anti-tumor activity (12-
14). Therefore, TIM-3 blockade and Treg depletion
could have a synergistic effect on tumor growth
inhibition (15).
*Corresponding author: Mazdak Ganjalikhani-hakemi. Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences. Isfahan, Iran.
Tel: +98-31-37929082; Fax: +98-31-3792903 1; email: mghakemi@med.mui.ac.ir
Homayouni et al Novel Nanobody prepared against TIM-3
Iran J Basic Med Sci, Vol. 19, No.11, Nov 2016
1202
TIM
-3 is also expressed on cancer stem cells in patients
with acute myeloid leukemia (AML) and has a role in
promoting myeloid-derived suppressor cells (MDSC).
Furthermore, in AML cell lines (HL-60), TIM-3 moderately
activates the mammalian target of rapamycin (mTOR, a
master regulator of myeloid cell translational pathways).
This results in activation of the hypoxia-inducible factor 1
(HIF-1) transcription complex
, which upregulates
glycolysis and expression/secretion of the pro-
angiogenic vascular endothelial growth factor (VEGF)
(16). In a study, anti-human TIM-3 antibody blocked
AML engraftment in a xenotransplant model (17).
The role of TIM-3 in modulation of autoimmune
diseases has been studied in experimental autoimmune
encephalomyelitis (EAE), as a mouse model of MS. Low-
level expression of TIM-3 on EAE Th1 cells may explain
their Th1 resistance to inhibition induced by TIM-3/
galectin-9 pathway (18).
Now a day, antibodies are widely used as diagnostic
and therapeutic tools in clinic. The large size of
monoclonal antibodies (mAb) in some clinical
situations is a drawback, which limits their penetration
and distribution deep into tissues (19). In addition, the
Fc portion of mAbs is more likely to activate
complement system or phagocytosis process in vivo
rather than having solely a blocking or stimulating
activity. To overcome these limitations, fragments of
antibodies have been generated. Among antibody
fragments, nanobodies (Nbs) have unique features,
which make them appropriate tools for manipulating
immune responses for therapeutic purposes (20, 21).
This unique type of antibody is found in sera of camels
(dromedaries) and llamas. These camelid antibodies
lack light chains and are called heavy-chain antibodies
(HcAbs). The variable domain of HcAbs is named VHH,
Nanobody or single domain antibody, which is
responsible only for antigen binding. Nbs are the
smallest fully functional antigen-binding fragment of
these antibodies. It appears that Nbs have very low
immunogenicity for human because of a high degree of
sequence similarity between VHH and human VH
sequences (22, 23). Nbs are more efficient in
immunotherapy, because they have small size, which
enable them to access inaccessible positions in the
body. They also have high affinity and specificity for
their cognate antigen. Based on the importance of
blocking and/or stimulating of TIM-3 signaling in
different pathologic conditions, this study was aimed to
design and produce a monoclonal Nb with a high
affinity against the TIM-3 protein for further in vitro
and in vivo studies.
Materials and Methods
Preparation of antigen for immunization
The HEK 293 cell line that expressed recombinant
human TIM-3 was prepared for immunization (in
press). A 6 months Camelus dromedaries was
intramuscularly immunized with 50 μg purified Human
TIM-3 Protein (Sinobiological, inc) plus complete
Freund’s adjuvant (CFA, CMG company) followed by the
adjuvant free lysates prepared from 5×107 human TIM-
3 expressing cells for 4 times every 2 weeks. Whole
blood was collected before the first injection and 7 days
after each injection, and the sera were isolated for
measurement of the antibody titer.
Library construction
Ten days after the last immunization, 400 ml blood
was collected and peripheral blood mononuclear cells
(PBMC) were separated using Lymphoprep (Greiner
Bio-one). Total RNA was extracted from isolated PBMCs
with the RNX Plus reagent (Cinnagen, Iran) and cDNA
was synthetized with a RevertAid First Strand cDNA
Synthesis Kit (Fermentas, Germany) using OligodT
primer. Nested PCR was done for VHH amplification
using specific primers. The leader-specific primer
CALL001 (5′-GTC CTG GCT GCT CTT CTA CAA GG-3′)
and CH2-specific primer CALL002 (5′ -GGT ACG TGC
TGT TGA ACT GTT CC-3′) were used for VH and VHH
amplification.
PCR products were electrophoresed on a 1%
agarose gel and the 600-700 bp fragments (VHH-CH2
without CH1 exon) were purified from the gel with
AccuPrep Gel Purification Kit (Bioneer, Korea). Purified
bands were re-amplified using nested primers A6E
(5′-GAT GTG CAG CTG CAG GAG TCT GGR GGA GG-3′)
and primer 38 (5′-GGA CTA GTG CGG CCG CTG GAG
ACG GTG ACC TGG GT-3′) for framework 1 and
framework 4 regions. The pHEN4 phagemid vector and
the amplified PCR product were digested with PstI and
NotI restriction enzymes and then, ligated with T4 DNA
ligase enzyme. Recombinant vector was transformed
into electro-competent Escherichia coli TG1 cells.
Colony PCR was done to confirm the successful cloning.
Enrichment of the VHH library
The VHH libraries were displayed on phages after
their infection with VCSM13 helper phages. This library
was grown in 330 ml 2xTY media containing 100 μg/ml
ampicillin and 4% of glucose. Bacteria at mid-log phase
(OD=0.5 at 600 nm) were infected with1012 CFU
of VCSM13 helper phage. Infected bacteria were
incubated for 30 min at 37 °C. After a centrifugation, the
resulted pellet was cultured in 2×TY supplied with
50 mg/ml Kanamycin and incubated at 37 °C for 16 hr
while shaking at 250 rpm. The culture medium was
centrifuged for 20 min at 9000 rpm at 4 °C and the
supernatant was mixed with polyethylene glycol (PEG)-
NaCl. This was incubated for 60 min on ice. After
centrifugation for 15 min at 4000 rpm and 4 °C, the
pellet was isolated and re-suspended in 1 ml of PBS.
Enrichment of the specific phage was done with three
rounds of in vitro selection on microtiter plates coated
with 10 μg recombinant TIM-3 protein. After each
selection round, binders were eluted with 100 mM
triethylamine (pH 10) and immediately neutralized
Novel nanobody prepared against TIM-3 Homayouni et al
Iran J Basic Med Sci, Vol. 19, No. 11, Nov 2016
1203
with 1M TrisHCl, pH 8. Phage particles were finally
used to infect exponentially growing E. coli TG1
bacteria.
Polyclonal phage ELISA, applying the extracted
phages, was used for evaluation of the panning process
after each round. After three rounds of panning,
individual colonies of third round were randomly
picked for periplasmic extract ELISA (PE-ELISA) in
order to detect positive clones. In PE-ELISA expression
of soluble periplasmic VHHs was induced with 1 mM
isopropyl-d-1-thiogalactopyranoside (IPTG). The
periplasmic proteins were extracted using osmotic
shock with TES buffer and extracted material
containing recombinant VHH was tested for antigen
recognition.
Expression and characterization of VHH Nanobody
After PE-ELISA, samples with the highest optical
density (OD) were selected for further experiments.
The VHH genes of the selected colonies were sub-
cloned into the pHEN6C expression vector with BstEII
and PstI restriction sites. This vector includes a C-
terminal His-tag. The recombinant pHEN6C was
transformed into E. coli WK6 (Pasteur Institute, Iran)
and expressed Nb was obtained. Briefly, the
periplasmic crude extract proteins were prepared using
osmotic shock and loaded on a His-Select column
(Sigma-Aldrich, Germany). After washing with PBS, the
bound proteins were eluted with 500 mM imidazole.
SDS-PAGE and Western blotting
The purity and identity of the protein was evaluated
on a15% gel with SDSPAGE method. The gel was
stained with Coomassie brilliant blue. Western blotting
was done with anti-His tag antibody (Abcam, UK). For
Western blotting, the gel was run at a constant voltage
of 100 V for 45 min using a Mini Protein Tetra System
(Bio-Rad, USA), in order to transfer the protein bands to
the nitrocellulose membrane (CMG Company). The
nitrocellulose membrane was then blocked with 5%
skimmed milk in PBS-T for 16 hr at 4°C. The membrane
was washed and detection was done with 1/5000
dilution of HRP conjugated anti-His Tag antibody
(Abcam, UK) and diaminobenzidine (DAB) (CMG
Company) as substrate.
Measurement of TIM-3 Nanobody affinity
Affinity of the purified Nanobodies (Nb94 and
Nb60) was determined using ELISA method. TIM-3
antigen (HAVCR2 Protein /Sino Biological Inc.) was
coated in a 96 well microplate wells (Nunc, Denmark)
in different concentrations (5 and 10 μg/ml). After
washing and blocking, the Nb was added at 0.001, 0.01,
0.1, 1, 10 nM concentrations. HRP-conjugated anti-His
antibody (Abcam, UK) was added and the immune
reactivity was assessed with TMB substrate. The
reaction was stopped with a stop solution and OD was
measured at 450 nm using a microplate reader
(Hyperion).
Flowcytometric analysis
The TIM-3 expressing HL-60 cells (an AML cell line),
induced with PMA, and TIM-3 expressing HEK 293
cells, stably transfected, as well as TIM-3 negative
HEK293 cells (as negative control) were used
for flowcytometric analysis of anti-TIM-3 Nb
immunoreactivity. About 5×105 cells were washed
three times and re-suspended in a total volume of 100
μl PBS. One microgram of Nb was added, and cells were
incubated for 45 min on ice. After washing with PBS,
cells were incubated with 1 μg mouse anti-His-tag
antibody-PE (Biolegend, UK) for 1 hr on ice. Anti-TIM-
3-PE monoclonal antibody (mAb) (Biolegend, UK) was
applied as positive control for another group of cells.
Cells were washed with PBS and analyzed with Cell
Quest Pro software in a FACS Callibour (BD Biosciences,
USA) instrument.
CFSE Proliferation assay
In the assay, HL-60 cells were grown in RPMI
medium, induced with PMA and labeled with
10 mM carboxyfluorescein-succinimidyl ester (CFSE)
(Biolegend, UK) at 37 °C for 20 min in the dark and
washed twice with PBS containing 10% FBS to remove
excessive CFSE. Cells were seeded at 2.5×105 cells/well
in 24-well plates and incubated at 37 °C with 5% CO2. At
least 24 hr before analysis, 2 μl (1 μM) galectin-9
(Biolegend, UK) was added into each well. One group
was treated with our anti-TIM-3 Nanobody (10 µl) as
the test group and a standard anti-TIM-3 mAb (10 µl)
was added to another group as the positive control. One
group of the seeded cells was not treated with galectin-
9 as the negative control. At the appropriate point in
time, cells were washed twice, re-suspended in PBS
buffer, and analyzed immediately using a FACS Calibur
flowcytometer (Becton Dickinson, USA).
Statistical analysis
Paired-T-Test and one way ANOVA were applied to
compare the results before and after interventions
between two and more than two groups, respectively.
P<0.05 was considered as the statistical significance. All
the analyses were done using SPSS 20 (SPSS Inc.,
Chicago, IL, USA).
Table 1. Camel anti-TIM-3 antibody level after each round of
immunization
Test
Control
(Non specific binding)
0.149±0.01
0.19±0.05
1.7±0.2
0.141±0.04
Homayouni et al Novel Nanobody prepared against TIM-3
Iran J Basic Med Sci, Vol. 19, No.11, Nov 2016
1204
Figure 1. A. Library construction. Analysis of the first PCR product
with agarose gel electrophoresis (1%); the PCR product with different
sizes 900, 700, and 600 bp, M shows: molecular weight marker (1 kb).
B. Analysis of the second PCR product by gel electrophoresis (1%); the
VHH fragment with 400 bp size is showed. M, molecular weight
marker (1 kb). The a band shows 1000 bp.The b band shows 700 bp.
The c band shows 400 bp
Figure 2. Colony PCR analysis of colonies from the library. The most of
the colonies had an inserted fragment of VHH gene (600 bp). M,
molecular weight marker (100 bp)
Results
Immunization and construction of Nanobody library
Anti-human TIM-3 antibody level was increased
significantly in the serum of immunized camel
(P=0.0001) 10 days after the last injection (Table 1).
Very low optical density in controls in compare with
tests (P=0.0001) confirmed no non-specific binding
(NSB) in the experiment. Then, lymphocytes were
isolated from anti-coagulated blood of the immunized
camel. Total RNAs were purified and cDNA was
synthesized. The variable domains of the heavy-chain
antibodies were amplified with PCR and bands related
to both classical antibody (900 bp) and heavy chain
antibody (600-700 bp) were obtained (Figure 1A).
In nested PCR, a 400 bp region between the
framework 1 and framework 4 (VHH) was amplified
(Figure 1B). The digested PCR fragments were ligated
into pHEN4 phagemid vector and transformed into E.
coli TG1. The obtained library of transformant has
2.1×107 members. Colony PCR analysis of 25 randomly
picked colonies from the library showed that the most
of the colonies had a phagemid containing an inserted
fragment of VHH gene (Figure 2).
We identified TIM-3 Nbs by bio-panning in three
rounds to validate the quality of the library using phage
display technology. In ELISA enrichment, we obtained a
relatively enriched phage eluted from wells coated with
TIM-3 protein. The third round of panning showed the
highest immuno-reactivity in the polyclonal phage
ELISA (Figure 3). 48 clones were screened from the
third round of panning that bound to TIM-3
recombinant protein using PE-ELISA. The Nb49and
Nb60 clones showed the highest color intensity at 450
nm by ELISA reader and selected for expression (Figure
4).
Figure 3. The panning process with phage ELISA. The absorbance and
enrichment against TIM-3 was increased and the highest enrichment
was obtained in the third panning round. The absorbance against skim
milk (negative control) remained constant
Figure 4. Periplasmic extract (PE)-ELISA for selecting the most
secretory anti-TIM-3 VHH expressing colonies. A selected nanobody
clone showing the highest optical density at 450 nm in compare with
negative control. Presented data are mean±SD of three identical
repeats of the same experiment. Asterisks (*) shows statistical
significance compared with control
Novel nanobody prepared against TIM-3 Homayouni et al
Iran J Basic Med Sci, Vol. 19, No. 11, Nov 2016
1205
Expression of soluble VHH Nanobody
For expression of the binder Nbs, Nb49 and Nb60
were sub-cloned into a pHEN6C expression vector. Nb
was expressed in fusion with N-terminal His-tag and
purified by NTA-affinity chromatography, with a 0.005
μg concentration.
Nanobody Characterization
Using SDS-PAGE analysis, a single 15 kDa band
representing for the presence of the purified Nb (VHH)
was observed on polyacrylamide gel. No contamination
or degraded product was detected. Western blotting
confirmed the identity of the 15 kDa band (Figure 5).
Then, using ELISA method and Beaty equation, the
affinity of the Nb was calculated as 6×10-8 M.
A flowcytometric analysis was performed to access
the specific binding activity of the Nanobody to human
TIM-3 expressed on cell surface, compared with a
standard anti-TIM-3 antibody. Based on the obtained
results, the frequency of TIM-3 positive HEK293 cells
detected with our Nb was almost similar with the
standard anti-TIM-3 antibody (80% and 85%,
respectively). The results for HL-60 cells were also
similar (53% detected with the Nb and 65%with the
standard antibody) (Figure 6).
CFSE proliferation Assay
Engagement of TIM-3 on HL-60 cells with galectin-9
leads to the proliferation of the HL-60 cells. Here, using
PMA, HL-60 cells were induced to express TIM-3 and
then were treated with galectin-9 as a specific TIM-3
ligand. The cells proliferation was significantly
increased 24 hr after treatment with galectin-9
compared with un-treated (negative) control
(P=0.0001). Meanwhile, the proliferation of TIM-3
expressing HL-60 cells was significantly inhibited
(P=0.0001) using both the Nb and the commercial
antibody versus control (17.2% and16.3% versus
100%, respectively) (Figure7).
Figure 5. SDS-PAGE and Western blotting. A) SDS-PAGE analysis
of Nanobody purified by immobilized metal affinity
chromatography (IMAC); lane M: molecular weight marker; lane 1:
Nb49 and 2: Nb60 (TIM-3- Nanobody) eluted by 500 mM
imidazole buffer. B) Lanes 1 and 2: the specific reaction of the
HRP-conjugated anti-His Tag antibody with Nb49, Nb60,
respectively. Lane M: molecular weight marker
Figure 6. Flowcytometry analysis. A) HEK293 stably transfected
cell line, express in 85% TIM-3 stained with anti-TIM-3PE .B)
HEK293 stably transfected cell line, express in 80% TIM-3 stained
with anti-TIM-3 nanobody, mouse anti-His PE-conjugated
antibody. C) HL-60 cell line that induce for TIM-3 expression and
stained with anti-TIM-3 PE. About 65% of cells showed expressed
TIM-3.D) HL-60 cell line that induced for TIM-3 expression and
stained with anti-TIM-3 Nanobody and anti-His PE conjugated
antibody. About 53% of cells showed expressed TIM-3
Figure 7. Nanobody TIM-3 induced inhibition of HL-60 cell line
proliferation. TIM-3 expressing HL60 cells were treated with
Galectin-9 to be induced for proliferation. A) Proliferation
inhibition of HL-60 cells using anti-TIM-3 antibody. B)
Proliferation inhibition of HL-60 cells using anti-TIM-3 nanobody.
In the histograms, (1) represents for HL-60 proliferation; (2)
represents for HL60 cells were treated with Galectin-9; and (3)
represents for Proliferation inhibition. C) More than 80%
inhibition of proliferation was observed using both the antibody
and the Nanobody. No meaningful difference was observed
between the antibody and the Nanobody inhibitory effect
(P˃0.05). Asterisks (*) shows statistical significance compared
with control
Homayouni et al Novel Nanobody prepared against TIM-3
Iran J Basic Med Sci, Vol. 19, No.11, Nov 2016
1206
Discussion
TIM-3 exerts various important roles in the
regulation of T cell responses and innate immune
system. It has three known ligands including Galectin-9,
Phosphatidyl Serin (PS), and high-mobility group
protein 1 (HMGB-1). The interaction of Galectin-9-TIM-
3 in TIM-3-expressing Th1 cells leads to cell death. PS
binding by TIM-3 can mediate uptake of apoptotic cells
by phagocytes. Engagement of TIM-3 by HMGB-1 has
been shown to inhibit activation of resident DCs in the
tumor micro-environment (24, 25). More recently, it
has been shown that TIM-3 is present at the immune
synapse and can be colocalized with phosphatases that
suppress TCR signaling. TIM-3 has also been shown to
inhibit innate immune cells functions like IL-12
secretion and TLR-mediated activation (26, 27). Based
on these important functions in immune responses,
TIM-3 is now considered as a promising therapeutic
target for different pathologic conditions.
In the context of infectious diseases, one study has
shown that the blockade of TIM-3 engagement with its
ligands in mouse models of chronic infections can
augment T cell responses, which helps to eradicate the
infection (28). Independent reports have shown that
specific blocking of TIM-3 and PD-1 signaling pathways
improves T cell responses leading to control of viral
infection in chronically infected patients (14, 24). In a
recent study, it has been reported that, in the presence
of TIM-3 blocking antibodies, the production of IL-6,
IFN-γ and TNF-α is increased in Mycobacterium
tuberculosis and in viral infections, which was very
useful in inducing macrophage activation and
restricting microbial growth (28).
Several blocking mAbs have been generated to
target mouse and human TIM-3, so far. These include
RMT3-23, 8B.2C12, 2E2, AF2365, ATIK2a, 344823 and
344801 which differentially affect immune functions
(14, 19, 29, 30). RMT323 is an anti-mouse TIM-3
blocking mAb and has been widely used and tested in
various murine tumor models including melanoma,
colon adenocarcinoma, sarcoma, prostate carcinoma,
and ovarian cancer. Immunotherapy with RMT323
mAb alone was highly effective to increase tumor-
infiltrating IFN-γ-producing CD4 and CD8 T cells and to
suppress T regulatory (Treg) functions in sarcoma and
colon adenocarcinoma models (30-32).
TIM-3 also is being considered as a target antigen
for anti-leukemia therapy including T-cell leukemia and
AML as well as for reversing T cell exhaustion and
restoring anti-tumor immunity. Therefore, the success
in TIM-3 blockade could be a major step forward to the
development of immunotherapy for the treatment of
different diseases such as cancers (33).
Inaccessible anatomical points are a bottleneck for
obtaining acceptable clinical outcome after intervention
with mAbs, as some therapeutic monoclonal antibodies
need to penetrate deep into such tissues to reach their
targets. In order to facilitate Ab accession to
inaccessible points, Ab fragments such as Scfv, Fv, Fab
and HcAbs have been introduced, so far.
HcAbs can overcome this bottleneck as they include
only a single variable domain (called VHH from camel
Ab), which generates high affinities towards a large
spectrum of antigens. These small domains (15 kDa)
can be easily produced in bacteria or yeasts and are
then called domain antibodies (dAbs), or Nbs (34). Nbs
are clearly privileged compared with conventional
antibody fragments. Some reports of the use of Nbs
against tumor associated cell-surface markers have
shown that Nbs can act like normal antibodies (35-37).
Heavy chain antibodies (VHH) have provided new
opportunities in clinical applications of mAbs. These
unique antibodies interact with the antigen by virtue of
only one single variable domain. The recombinant Nbs
selected from phage display libraries are well
expressed, highly soluble in aqueous environments,
very robust and have high sequence homology with the
human variable region gene family (23).
The affinity of different antibodies has been
previously reported in the range of 10-7 to 10-10M (38).
In the present work, the equilibrium dissociation
constant (Kd) of the anti-TIM-3 Nb was determined as
6×10 -8 M which is a suitable affinity. Several studies
shown that very high affinities can be suboptimal for
therapeutic antibodies that target solid tumors. As high
affinity antibodies tightly bind their specific antigen
upon the first encounter, at the periphery of the tumor,
they do not penetrate deeper inside the tumor until all
antigen molecules are saturated at the periphery. By
contrast, moderate binders are released from these first
encountered antigens and penetrate deeper into the
tumor, ultimately leading to uniform intratumoral
distribution and higher tumor uptake (34). VHHs,
unlike conventional antibodies, can recognize epitopes
in the inaccessible sites such as caves and clefts; hence,
penetrate more deeply in different sites (34).
Roovers et al have characterized a Nb against
epidermal growth factor receptor (EGFR) and showed
in vitro inhibition of EGF binding to EGFR and in vivo
therapeutic effect on tumor in a mouse model (39).
Behdani et al have shown that the VEGFR2-specific Nb
can recognize antigen on the HUVEC cell surface and
can inhibit in vitro endothelial tube formation and could
be considered as a cancer therapy agent (36).
Although many VHH phage libraries have been
constructed so far, based on our surveys, no VHH
against TIM-3 has been reported in literature or in
industry. In the current study, we successfully prepared
a novel anti-human TIM-3 (CD366) Nb from a camel
immune library using phage display method. We
showed its high binding capacity to TIM-3 comparable
with a commercial antibody. Interestingly, this Nb
showed a high anti-proliferation effect on HL-60, an
AML cell line, which was comparable with, even more
than, the inhibitory effect of a standard anti-TIM-3
antibody.
Novel nanobody prepared against TIM-3 Homayouni et al
Iran J Basic Med Sci, Vol. 19, No. 11, Nov 2016
1207
Conclusion
Altogether, we have been described the successful
generation of an anti-TIM-3-specific Nb from an
immune camel library, followed by soluble expression
of VHH protein and its binding capacity to TIM-3
(CD366). We also showed its high ability to block
Gal9/TIM-3 stimulated proliferation of a leukemic cell
line in vitro.
Acknowledgement
This work was financially supported by Iran
National Science Foundation (Grant No. 92028659) and
Isfahan University of Medical Sciences (Grant No.
392532).
Conflict of interest
The authors declare that there is no conflict of
interests regarding the publication of this paper.
References
1. Kane LP. T cell Ig and mucin domain proteins and
immunity. J Immunol 2010; 184:2743-2749.
2. Umetsu DT, Umetsu SE, Freeman GJ, DeKruyff RH.TIM
gene family and their role in atopic diseases. Curr Top
Microbiol Immunol 2008; 321:201-215.
3. Tong D, Zhou Y, Chen W, Deng Y, Li L, Jia Z, et al. T cell
immunoglobulin- and mucin-domain-containing
molecule 3 gene polymorphisms and susceptibility to
pancreatic cancer. Mol Biol Rep 2012; 39:9941-9946.
4. Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H,
Chernova T, et al. Th1-specific cell surface protein Tim-3
regulates macrophage activation and severity of an
autoimmune disease. Nature 2002; 415:536-541.
5. Tang ZH, Liang S, Potter J, Jiang X, Mao HQ, Li Z. Tim-
3/galectin-9 regulate the homeostasis of hepatic NKT
cells in a murine model of nonalcoholic fatty liver
disease. J Immunol 2013; 190:1788-1796.
6. Anderson AC, Anderson DE, Bregoli L, Hastings WD,
Kassam N, Lei C, et al. Promotion of tissue inflammation
by the immune receptor Tim-3 expressed on innate
immune cells. Science 2007; 318:1141-1143.
7. Nagahara K, Arikawa T, Oomizu S, Kontani K,
Nobumoto A, Tateno H, et al. Galectin-9 increases Tim-
3+ dendritic cells and CD8+ T cells and enhances
antitumor immunity via galectin-9-Tim-3 interactions. J
Immunol 2008; 181:7660-7669.
8. Wherry EJ. T cell exhaustion. Nat Immunol 2011;
12:492-499.
9. Le Mercier I, Lines JL, Noelle RJ. Beyond CTLA-4 and
PD-1, the Generation Z of Negative Checkpoint
Regulators. Front Immunol 2015; 6:418.
10. Ngiow SF, von Scheidt B, Akiba H, Yagita H, Teng
MW, Smyth MJ. Anti-TIM3 antibody promotes T Cell
IFN-g-mediated antitumor immunity and suppresses
established tumors. Cancer Res 2011; 71:3540-3551.
11. Baghdadi M, Takeuchi S, Wada H, Seino K. Blocking
monoclonal antibodies of TIM proteins as orchestrators
of anti-tumor immune response. MAbs 2014; 6:1124-
1132.
12. Jin HT, Anderson AC, Tan WG, West EE, Ha SJ, Araki
K, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell
exhaustion during chronic viral infection. Proc Natl
Acad of Sci U S A 2010; 107:14733-1478.
13. Dietze KK, Zelinskyy G, Liu J, Kretzmer F, Schimmer
S, Dittmer U. Combining regulatory T cell depletion and
inhibitory receptor blockade improves reactivation of
exhausted virus-specific CD8 (+) T cells and efficiently
reduces chronic retroviral loads. PLoS Pathog 2013;
9:e1003798.
14. Sakuishi K, , Apetoh L, Sullivan JM, Blazar BR,
Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1
pathways to reverse T cell exhaustion and restore anti-
tumor immunity. J Exp Med 2010; 207:2187- 2194.
15. Sakuishi K, Ngiow SF, Sullivan JM, Teng MW,
Kuchroo VK, Smyth MJ, Anderson AC. TIM3FOXP3
regulatory T cells are tissue-specific promoters of T-cell
dysfunction in cancer. Oncoimmunology 2013;
2:e23849.
16. Gonçalves Silva I, Gibbs BF, Bardelli M, Varani L,
Sumbayev VV. Differential expression and biochemical
activity of the immune receptor Tim-3 in healthy and
malignant human myeloid cells. Oncotarget 2015;
6:33823-33833.
17. Anderson AC. Tim-3, a negative regulator of anti-
tumor immunity. Curr Opin Immunol 2012; 24:213-216.
18. Freeman GJ, Casasnovas JM, Umetsu DT, DeKruyff
RH. TIM genes:a family of cell surface
phosphatidylserine receptors that regulate innate and
adaptive immunity. Immunol Rev 2010; 235:172-189.
19. Siontorou CG. Nanobodies as novel agents for
disease diagnosis and therapy. Int J Nanomed 2013;
8:42154227. .
20. Bell A, Wang ZJ, Arbabi-Ghahroudi M, Chang TA,
Durocher Y, Trojahn U, et al. Differential tumor-targeting
abilities of three single-domain antibody formats.
Cancer Lett 2010; 289:8190.
21. Hamers-Casterman C, Atarhouch T, Muyldermans S,
Robinson G, Hamers C, Songa EB, et al. Naturally
occuring antibodies devoid of light chains. Nature 1993;
363:446-448.
22. Yardehnavi N, Behdani M, Bagheri KP,
Mahmoodzadeh A, Khanahmad H, Shahbazzadeh D, et al.
A camelid antibody candidate for development of a
therapeutic agent against Hemiscorpius lepturus
envenomation. FASEB J 2014; 28:4004-4014.
23. Muyldermans S. Single domain camel antibodies:
current status. J Biotechnol 2001; 74:277-302.
24. DeKruyff RH, Bu X, Ballesteros A, Santiago C, Chim
YL, Lee HH, et al. T cell/transmembrane, Ig, and mucin-3
allelic variants differentially recognize
phosphatidylserine and mediate phagocytosis of
apoptotic cells. J Immunol 2010; 184:1918-1930.
25. Cao E, Zang X, Ramagopal UA, Mukhopadhaya A,
Fedorov A, Fedorov E, et al. T cell immunoglobulin
mucin-3 crystal structure reveals a galectin-9-
independent ligand-binding surface. Immunity 2007;
26:311-321.
26. Chiba S, Chiba S, Baghdadi M, Akiba H, Yoshiyama H,
Kinoshita I, Dosaka-Akita H, et al. Tumor-infiltrating DCs
suppress nucleic acid-mediated innate immune
responses through interactions between the receptor
TIM-3 and the alarmin HMGB1. Nat Immunol 2012;
13:832-842.
27. Clayton KL, Haaland MS, Douglas-Vail MB, Mujib S,
Chew GM, Ndhlovu LC, et al. T cell Ig and mucin domain-
containing protein 3 is recruited to the immune synapse,
Homayouni et al Novel Nanobody prepared against TIM-3
Iran J Basic Med Sci, Vol. 19, No.11, Nov 2016
1208
disrupts stable synapse formation, and associates with
receptor phosphatases. . J Immunol 2014; 192:782-791.
28. Sada-Ovalle I, Ocaña-Guzman R, Pérez-Patrigeón S,
Chávez-Galán L, Sierra-Madero J, Torre-Bouscoulet L,
et al. Tim-3 blocking rescue macrophage and T cell
function against Mycobacterium tuberculosis infection
in HIV+ patients. J Int AIDS Soc 2015; 19:20078.
29. Sakuishi K, Ngiow SF, Sullivan JM, Teng MW,
Kuchroo VK, Smyth MJ, et al. TIM3FOXP3 regulatory T
cells are tissue-specific promoters of T-cell dysfunction
in cancer. Oncoimmunology 2013; 2:e23849.
30. Baghdadi M, Takeuchi S, Wada H, Seino K.Blocking
monoclonal antibodies of TIM proteins as orchestrators
of anti-tumor immune response.
mAbs 2014; 6:1124-1132.
31. Chiba S, Baghdadi M, Akiba H, Yoshiyama H,
Kinoshita I, Dosaka-Akita H, et al. Tumor-infiltrating DCs
suppress nucleic acid-mediated innate immune
responses through interactions between the receptor
TIM-3 and the alarmin HMGB1. Nat Immunol 2012;
13:832-842.
32. Ngiow SF, von Scheidt B,Akiba H, Yagita H, Teng
MWL, Smyth MJ. Anti-TIM3 antibody promotes T cell
IFN-g-mediated antitumor immunity and sup- presses
established tumors. Cancer Res 2011; 71:3540- 3551.
33. Leone RD, Lo YC, Powell JD. A2aR antagonists:
Next generation checkpoint blockade for cancer
immunotherapy. Comput Struct Biotechnol J 2015;
13:265-272.
34. Chames P, Van Regenmortel M, Weiss E, Baty
D.Therapeutic antibodies: successes, limitations and
hopes for the future. Br J Pharmacol 2009; 157:220-233.
35. Omidfar K, Rasaee MJ, Modjtahedi H, Forouzandeh
M, Taghikhani M, Bakhtiari A, et al. Production and
characterization of a new antibody specific for the
mutant EGF receptor,EGFRvIII, in Camelus Bacterianus.
Tumor Biol 2004; 25:179-187.
36. Behdani M, Zeinali S, Khanahmad H, Karimipour M,
Asadzadeh N, Azadmanesh K,et al. Generation and
characterization of a functional Nanobody against the
vascular endothelial growth factor receptor-2;
angiogenesis cell receptor. Mol Immunol 2012; 50:35-
41.
37. Ahmadvand D, Rasaee MJ,Rahbarizadeh F,
Kontermann RE, Sheikholislami F. Cell selection and
characterization of a novel human endothelial cell
specific nanobody. Mol Immunol 2009; 46:1814-1823.
38. Abul Abbas AHL, Shiv Pillai. Cellular and Molecular
Immunology. 2015; 8th edition.
39. Roovers RC, Laeremans T, Huang L, De Taeye S,
Verkleij AJ, Revets H, et al. Efficient inhibition of EGFR
signaling and of tumour growth by antagonistic anti-
EFGR Nanobodies. Cancer Immunol Immunother 2007;
56:303-317.
... Fooladinezhad et al. and Mohammad-Ganji et al. have both shown that the TIM-3-Gal-9 interaction is a key player in the domains of infection, inflammation, tumor immunity, and autoimmunity control, as the downstream effects work to suppress the immune cell responses [143,145]. Further work showed that the TIM-3-Gal-9 signaling pathway causes cellular exhaustion and subsequent apoptosis of tumor-antigen-specific Th1 cells, further impairing the antitumor immune response [140,142,146]. Importantly, blocking of the TIM-3-Gal-9 signaling pathways has been shown to reactivate the T-cellmediated antitumor immune response, as evidenced by increased cytokine production and T-cell proliferation within the HCC TME [137,147]. ...
... These findings above show that an effective blockade of TIM-3 can help limit, or even reverse, some of the immunosuppression caused by the receptor. Multiple studies have shown that inhibition of TIM-3 reduces the number of suppressor immune cells, increases the production of pro-inflammatory cytokines such as IFN-γ, IL-6, and TNF-α, and restores the effector and antitumor activity of exhausted CD8 T-cells [146,148,150]. Additionally, blockage of the TIM-3-Gal-9 pathway causes a significant decrease in the immunosuppressive activity of Tregs in vitro [151], and this blockade has wholly reversed Treg-mediated immune suppression in some types of cancer and significantly decreased the development of cancer [140]. ...
Article
Full-text available
Simple Summary Liver cancer continues to exhibit increasing incidence and mortality worldwide. Subsequently, there has been a concomitant effort to development novel therapeutics that effectively target and treat these malignancies. This paper aims to provide a thorough review of the available immunotherapy treatments that target liver cancer, with a specific focus on the role and potential therapeutic strategies available to the interventional radiologist. The underlying biochemical and molecular mechanisms underlying these diseases are discussed to highlight the specific targets for immunotherapy, the clinical efficacy and safety of available pharmaceutical treatments are analyzed, and future advancements in the field, such as combination therapies and personalized medicine, are explored as they pertain to the immunotherapy of liver cancer. Abstract Liver cancer, including hepatocellular carcinoma and intrahepatic cholangiocarcinoma, is increasing in incidence and mortality across the globe. An improved understanding of the complex tumor microenvironment has opened many therapeutic doors and led to the development of novel pharmaceuticals targeting cellular signaling pathways or immune checkpoints. These interventions have significantly improved tumor control rates and patient outcomes, both in clinical trials and in real-world practice. Interventional radiologists play an important role in the multidisciplinary team given their expertise in minimally invasive locoregional therapy, as the bulk of these tumors are usually in the liver. The aim of this review is to highlight the immunological therapeutic targets for primary liver cancers, the available immune-based approaches, and the contributions that interventional radiology can provide in the care of these patients.
... TIM-3 protein is involved in the regulation of the immune response and immune tolerance. It is expressed on Th1, Th17, monocytes, dendritic cells, and macrophages and is involved in CD8( +) T-cell depletion [6][7][8][9]. TIM-3 protein also plays a role in efferocytosis [5,10,11]. A key feature is its ability to bind specific ligands, which activate biochemical pathways. ...
Article
Full-text available
T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) expression has been a trending topic in recent years due to its differential expression in a wide range of neoplasms. TIM-3 is one of the key immune checkpoint receptors that interact with GAL-9, PtdSer, HMGB1 and CEACAM1. Initially identified on the surface of T helper 1 (Th1) lymphocytes and later on cytotoxic lymphocytes (CTLs), monocytes, macrophages, natural killer cells (NKs), and dendritic cells (DCs), TIM-3 plays a key role in immunoregulation. Recently, a growing body of evidence has shown that its differential expression in various tumor types indicates a specific prognosis for cancer patients. Here, we discuss which types of cancer TIM-3 can serve as a prognostic factor and the influence of coexpressed immune checkpoint inhibitors, such as LAG-3, PD-1, and CTLA-4 on patients' outcomes. Currently, experimental medicine involving TIM-3 has significantly enhanced the anti-tumor effect and improved patient survival. In this work, we summarized clinical trials incorporating TIM-3 targeting monoclonal and bispecific antibodies in monotherapy and combination therapy and highlighted the emerging role of cell-based therapies.
... Some advantages of nanobodies include high physical and thermal stability, high solubility, proteolytic resistance, simple genetic engineering, high tissue penetration, and low immunogenicity in humans (Maali et al., 2023). We and others have developed nanobodybased ICIs targeting PD-1 (Silva-Pilipich et al., 2020;Wen et al., 2020;Zhang et al., 2022), PD-L1 (Silva-Pilipich et al., 2023;Zhang et al., 2017), CTLA-4 (Sotoudeh et al., 2021;Wan et al., 2017), and TIM-3 (Homayouni et al., 2016;Xia et al., 2022) from phage display libraries. These nanobodies exhibit high specificity and affinity for their targets, as well as effective antitumor activity. ...
Chapter
Immune checkpoint inhibitors (ICIs) based on monoclonal antibodies represent a breakthrough for the treatment of cancer. However, their efficacy varies among tumor types and patients, and they can lead to adverse effects due to on-target/off-tumor activity, since they are administered systemically at high doses. An alternative and attractive approach for the delivery of ICIs is the use of gene therapy vectors able to express them in vivo. This review focuses on the most recent studies using viral vectors able to express ICIs locally or systemically in preclinical models of cancer. These vectors include non-replicating viruses, oncolytic viruses able to propagate specifically in tumor cells and destroy them, and self-amplifying RNA vectors, armed with different formats of antibodies against immune checkpoints. Non-replicating vectors usually lead to long-term ICI expression, potentially eliminating the need for repeated administration. Vectors with replication capacity, although they have a shorter window of expression, can induce inflammation which enhances the antitumor effect. Finally, these engineered vectors can be used in combination with other immunostimulatory molecules or with CAR-T cells, further boosting the antitumor immune responses.
... The high affinity and specificity properties make it a candidate tool for cancer treatment. The small size, high stability and solubility, low immunogenicity, resistance to heat-induced denaturation, and higher tissue penetration rate makes it a preferable choice for cancer therapy than conventional antibodies [16,33,36]. Because a recombinant immunotoxin that targets EpCAM can increase the specific anti-tumor activity against EpCAM-expressing cells; in this study, we used a nanobody targeting EpCAM as specific targeting and the T and catalytic domain of the diphtheria toxin. ...
Article
Full-text available
Epithelial Cell Adhesion Molecule (EpCAM) is overexpressed in a variety of cancers such as colon, stomach, pancreas, and prostate adenocarcinomas. Inhibition of EpCAM is considered as a potential target for cancer therapy. In current study, anti-EpCAM immunotoxin (α-EpCAM IT) was developed using genetic fusion of α-EpCAM single domain antibody (nanobody) (α-EpCAM Nb) to truncated form of diphtheria toxin. The expression of recombinant α-EpCAM IT was induced by Isopropyl β-d-1-thiogalactopyranoside (IPTG) and confirmed by SDS-PAGE and western blot. Recombinant α-EpCAM IT was purified from the inclusion bodies and refolded using urea gradient procedure. The cytotoxicity and apoptosis activity of α-EpCAM IT on EpCAM over-expressing (MCF7), low-expressing (HEK293), and no-expressing (HUVEC) cells were evaluated by 3–4,5-Dimethylthiazol-2-yl (MTT) assay and annexin V-FITC-PI assay as well. In addition, anti-tumor activity of α-EpCAM IT was evaluated on nude mice bearing MCF7 tumor cells. Results showed success expression and purification of α-EpCAM IT. The α-EpCAM IT showed time and dose-dependent anti-proliferative activity on MCF-7 cells. However, α-EpCAM IT did not show any anti-proliferative activity on HEK293 and HUVEC cells as well. In addition, the annexin V-FITC-PI assay results showed that α-EpCAM IT significantly increased apoptotic rate in MCF-7 cells with no effect on HEK293 and HUVEC as well. Moreover, α-EpCAM IT significantly reduced tumor size in vivo study. The achieved results indicate the potential of designing α-EpCAM IT as a novel therapeutic for cancer therapy.
... It takes a lot of time to prepare antibodies so that they can be used as biological elements in biosensors. Thus, researchers came up with the idea that smaller parts of antibodies could be utilized instead of a whole antibody [44][45][46][47][48][49][50]. ...
Article
Full-text available
Fast and precise diagnostic techniques are required for the treatment of many disorders. Biosensors are one of the diagnostic devices that are applicable in biological and medical sciences. Biosensors could be utilized to recognize biological molecules with high sensitivity. Biosensors are consisted of different components and have different types. Each type of biosensor is used in a particular field according to its specific features. Nanobodies are a novel class of antibodies with small size, high affinity, and specificity to their target. The unique properties of nanobodies make them appropriate tools for diagnostic applications. In this paper, we review biosensors, and their features and roles in medicine. Antibody/nanobody-based biosensors are also specifically discussed.
... The affinity of different antibodies has been reported in the 10-7 to 10-10 m range [31]. Some studies have shown that very high affinity is suboptimal for therapeutic antibodies targeting solid tumors [19]. Since high-affinity antibodies tightly bind their specific antigens at the first contact, they will not go Nb10. ...
Article
Full-text available
Monoclonal antibodies and antibody-derived biologics are essential tools for cancer research and therapy. The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. A nanobody constructed by molecular engineering of heavy-chain-only antibody, which is unique in camel or alpaca, is a burgeoning tools of diagnostic and therapeutic in clinic. In this study, we immunized a 4-year-old female alpaca with TIM-3 antigen. Then, a VHH phage was synthesized from the transcriptome of its B cells by nested PCR as an intermediate library; the library selection for Tim-3 antigen is carried out in three rounds of translation. The most reactive colonies were selected by periplasmic extract monoclonal ELISA. The nanobody was immobilized by metal affinity chromatography (IMAC) purification with the use of a Ni-NTA column, SDS-PAGE, and Western blotting. Finally, the affinity of TIM3-specific nanobody was determined by ELISA. As results, specific 15 kD bands representing nanomaterials were observed on the gel and confirmed by Western blotting. The nanobody showed obvious specific immune response to Tim-3 and had high binding affinity. We have successfully prepared a functional anti-human Tim-3 nanobody with high affinity in vitro.
... We developed Nbs that are able to bind mouse or human LAG3, yet their therapy efficacy remains to be determined (Lecocq et al., , 2021a. Homayouni (2016) developed anti-TIM3 Nbs and showed that they inhibit the proliferation of an AML cell line. ...
Chapter
Cancer is a heterogeneous disease, requiring treatment tailored to the unique phenotype of the patient's tumor. Monoclonal antibodies (mAbs) and variants thereof have enabled targeted therapies to selectively target cancer cells. Cancer cell-specific mAbs have been used for image-guided surgery and targeted delivery of radionuclides or toxic agents, improving classical treatment strategies. Cancer cell-specific mAbs can further inhibit tumor cell growth or can stimulate immune-mediated destruction of cancer cells, a feature that has also been achieved through mAb-mediated manipulation of immune cells and pathways. Drawbacks of mAbs and their variants, together with the discovery of camelid heavy chain-only antibodies and the many advantageous features of their variable domains, referred to as VHHs, single domain antibodies or nanobodies (Nbs), resulted in the exploration of Nbs as an alternative targeting moiety. We therefore review the state-of-the-art as well as novel exploitation strategies of Nbs for targeted cancer therapy.
Article
Immune checkpoints inhibition is a privileged approach to combat cancers and other human diseases. The TIM-3 (T cell immunoglobulin and mucin-domain containing-3) inhibitory checkpoint expressed on different types of immune cells is actively investigated as an anticancer target, with a dozen of monoclonal antibodies in (pre)clinical development. A soluble form sTIM-3 can be found in the plasma of patients with cancer and other diseases. This active circulating protein originates from the proteolytic cleavage by two ADAM metalloproteases of the membrane receptor shared by tumor and non-tumor cells, and extracellular vesicles. In most cancers but not all, overexpression of mTIM-3 at the cell surface leads to high level of sTIM-3. Similarly, elevated levels of sTIM-3 have been reported in chronic autoimmune diseases, inflammatory gastro-intestinal diseases, certain viral and parasitic diseases, but also in cases of organ transplantation and in pregnancy-related pathologies. We have analyzed the origin of sTIM-3, its methods of dosage in blood or plasma, its presence in multiple diseases and its potential role as a biomarker to follow disease progression and/or the treatment response. In contrast to sPD-L1 generated by different classes of proteases and by alternative splicing, sTIM-3 is uniquely produced upon ADAM-dependent shedding, providing a more homogenous molecular entity and a possibly more reliable molecular marker. However, the biological functionality of sTIM-3 remains insufficiently characterized. The review shed light on pathologies associated with an altered expression of sTIM-3 in human plasma and the possibility to use sTIM-3 as a diagnostic or therapeutic marker.
Article
Full-text available
The immune system is essential in recognizing and eliminating tumor cells. The unique characteristics of the tumor microenvironment (TME), such as heterogeneity, reduced blood flow, hypoxia, and acidity, can reduce the efficacy of cell-mediated immunity. The primary goal of cancer immunotherapy is to modify the immune cells or the TME to enable the immune system to eliminate malignancies successfully. Nanobodies, known as single-domain antibodies, are light chain-free antibody fragments produced from Camelidae antibodies. The unique properties of nanobodies, including high stability, reduced immunogenicity, enhanced infiltration into the TME of solid tumors and facile genetic engineering have led to their promising application in cell-mediated immunotherapy. They can promote the cancer therapy either directly by bridging between tumor cells and immune cells and by targeting cancer cells using immune cell-bound nanobodies or indirectly by blocking the inhibitory ligands/receptors. The T-cell activation can be engaged through anti-CD3 and anti-4-1BB nanobodies in the bispecific (bispecific T-cell engagers (BiTEs)) and trispecific (trispecific T-cell engager (TriTEs)) manners. Also, nanobodies can be used as natural killer (NK) cell engagers (BiKEs, TriKEs, and TetraKEs) to create an immune synapse between the tumor and NK cells. Nanobodies can redirect immune cells to attack tumor cells through a chimeric antigen receptor (CAR) incorporating a nanobody against the target antigen. Various cancer antigens have been targeted by nanobody-based CAR-T and CAR-NK cells for treating both hematological and solid malignancies. They can also cause the continuation of immune surveillance against tumor cells by stopping inappropriate inhibition of immune checkpoints. Other roles of nanobodies in cell-mediated cancer immunotherapy include reprogramming macrophages to reduce metastasis and angiogenesis, as well as preventing the severe side effects occurring in cell-mediated immunotherapy. Here, we highlight the critical functions of various immune cells, including T cells, NK cells, and macrophages in the TME, and discuss newly developed immunotherapy methods based on the targeted manipulation of immune cells and TME with nanobodies.
Article
Full-text available
Introduction: T cell immunoglobulin and mucin domain (Tim) 3 and programmed death 1 (PD-1) are co-inhibitory receptors involved in the so-called T cell exhaustion, and in vivo blockade of these molecules restores T cell dysfunction. High expression of Tim-3 and PD-1 is induced after chronic antigen-specific stimulation of T cells during HIV infection. We have previously demonstrated that the interaction of Tim-3 with its ligand galectin-9 induces macrophage activation and killing of Mycobacterium tuberculosis. Our aim in this study was to analyze the Tim-3 expression profile before and after six months of antiretroviral therapy and the impact of Tim-3 and PD-1 blocking on immunity against M. tuberculosis. Materials and methods: HIV+ patients naïve to anti-retroviral therapy (ART) were followed up for six months. Peripheral immune-cell phenotype (CD38/HLA-DR/galectin-9/Tim-3 and PD-1) was assessed by flow cytometry. Supernatants were analyzed with a multiplex cytokine detection system (human Th1/Th2 cytokine Cytometric Bead Array) by flow cytometry. Control of bacterial growth was evaluated by using an in vitro experimental model in which virulent M. tuberculosis-infected macrophages were cultured with T cells in the presence or absence of Tim-3 and PD-1 blocking antibodies. Interleukin-1 beta treatment of infected macrophages was evaluated by enumerating colony-forming units. Results: We showed that HIV+ patients had an increased expression of Tim-3 in T cells and were able to control bacterial growth before ART administration. By blocking Tim-3 and PD-1, macrophages and T cells recovered their functionality and had a higher ability to control bacterial growth; this result was partially dependent on the restitution of cytokine production. Conclusions: In this study, we demonstrated that increased Tim-3 expression can limit the ability of the immune system to control the infection of intracellular bacteria such as M. tuberculosis. The use of ART and the in vitro manipulation of the Tim-3 and PD-1 molecules restored the functionality of T cells and macrophages to restrict bacterial growth. Our results provide a novel immune strategy that may be implemented in the near future in order to improve the immune responses in HIV+ patients.
Article
Full-text available
INTRODUCTION: T cell immunoglobulin and mucin domain (Tim) 3 and programmed death 1 (PD-1) are co-inhibitory receptors involved in the so-called T cell exhaustion, and in vivo blockade of these molecules restores T cell dysfunction. High expression of Tim-3 and PD-1 is induced after chronic antigen-specific stimulation of T cells during HIV infection. We have previously demonstrated that the interaction of Tim-3 with its ligand galectin-9 induces macrophage activation and killing of Mycobacterium tuberculosis. Our aim in this study was to analyze the Tim-3 expression profile before and after six months of antiretroviral therapy and the impact of Tim-3 and PD-1 blocking on immunity against M. tuberculosis. MATERIALS AND METHODS: HIV+ patients naïve to anti-retroviral therapy (ART) were followed up for six months. Peripheral immune-cell phenotype (CD38/HLA-DR/galectin-9/Tim-3 and PD-1) was assessed by flow cytometry. Supernatants were analyzed with a multiplex cytokine detection system (human Th1/Th2 cytokine Cytometric Bead Array) by flow cytometry. Control of bacterial growth was evaluated by using an in vitro experimental model in which virulent M. tuberculosis-infected macrophages were cultured with T cells in the presence or absence of Tim-3 and PD-1 blocking antibodies. Interleukin-1 beta treatment of infected macrophages was evaluated by enumerating colony-forming units. RESULTS: We showed that HIV+ patients had an increased expression of Tim-3 in T cells and were able to control bacterial growth before ART administration. By blocking Tim-3 and PD-1, macrophages and T cells recovered their functionality and had a higher ability to control bacterial growth; this result was partially dependent on the restitution of cytokine production. CONCLUSIONS: In this study, we demonstrated that increased Tim-3 expression can limit the ability of the immune system to control the infection of intracellular bacteria such as M. tuberculosis. The use of ART and the in vitro manipulation of the Tim-3 and PD-1 molecules restored the functionality of T cells and macrophages to restrict bacterial growth. Our results provide a novel immune strategy that may be implemented in the near future in order to improve the immune responses in HIV+ patients.
Article
Full-text available
The T cell immunoglobulin and mucin domain 3 (Tim-3) is a plasma membrane-associated receptor which is involved in a variety of biological responses in human immune cells. It is highly expressed in most acute myeloid leukaemia (AML) cells and therefore may serve as a possible target for AML therapy. However, its biochemical activities in primary human AML cells remain unclear. We therefore analysed the total expression and surface presence of the Tim-3 receptor in primary human AML blasts and healthy primary human leukocytes isolated from human blood. We found that Tim-3 expression was significantly higher in primary AML cells compared to primary healthy leukocytes. Tim-3 receptor molecules were distributed largely on the surface of primary AML cells, whereas in healthy leukocytes Tim-3 protein was mainly expressed intracellularly. In primary human AML blasts, both Tim-3 agonistic antibody and galectin-9 (a Tim-3 natural ligand) significantly upregulated mTOR pathway activity. This was in line with increased accumulation of hypoxia-inducible factor 1 alpha (HIF-1`) and secretion of VEGF and TNF-`. Similar results were obtained in primary human healthy leukocytes. Importantly, in both types of primary cells, Tim-3-mediated effects were compared with those induced by lipopolysaccharide (LPS) and stem cell factor (SCF). Tim-3 induced comparatively moderate responses in both AML cells and healthy leukocytes. However, Tim-3, like LPS, mediated the release of both TNF-` and VEGF, while SCF induced mostly VEGF secretion and did not upregulate TNF-` release.
Article
Full-text available
In the last two years, clinical trials with blocking antibodies to the negative checkpoint regulators CTLA-4 and PD-1 have rekindled the hope for cancer immunotherapy. Multiple negative checkpoint regulators protect the host against autoimmune reactions but also restrict the ability of T cells to effectively attack tumors. Releasing these brakes has emerged as an exciting strategy for cancer treatment. Conversely, these pathways can be manipulated to achieve durable tolerance for treatment of autoimmune diseases and transplantation. In the future, treatment may involve combination therapy to target multiple cell types and stages of the adaptive immune responses. In this review, we describe the current knowledge on the recently discovered negative checkpoint regulators, future targets for immunotherapy.
Article
Full-text available
The last several years have witnessed exciting progress in the development of immunotherapy for the treatment of cancer. This has been due in great part to the development of so-called checkpoint blockade. That is, antibodies that block inhibitory receptors such as CTLA-4 and PD-1 and thus unleash antigen-specific immune responses against tumors. It is clear that tumors evade the immune response by usurping pathways that play a role in negatively regulating normal immune responses. In this regard, adenosine in the immune microenvironment leading to the activation of the A2a receptor has been shown to represent one such negative feedback loop. Indeed, the tumor microenvironment has relatively high concentrations of adenosine. To this end, blocking A2a receptor activation has the potential to markedly enhance anti-tumor immunity in mouse models. This review will present data demonstrating the ability of A2a receptor blockade to enhance tumor vaccines, checkpoint blockade and adoptive T cell therapy. Also, as several recent studies have demonstrated that under certain conditions A2a receptor blockade can enhance tumor progression, we will also explore the complexities of adenosine signaling in the immune response. Despite important nuances to the A2a receptor pathway that require further elucidation, studies to date strongly support the development of A2a receptor antagonists (some of which have already been tested in phase 3 clinical trials for Parkinson disease) as novel modalities in the immunotherapy armamentarium.
Article
Full-text available
Monoclonal antibody (mAb)-based treatment of cancer has a significant effect on current practice in medical oncology, and is considered now as one of the most successful therapeutic strategies for cancer treatment. MAbs are designed to initiate or enhance anti-tumor immune responses, which can be achieved by either blocking inhibitory immune checkpoint molecules or triggering activating receptors. TIM gene family members are type-I surface molecules expressed in immune cells, and play important roles in the regulation of both innate and adaptive arms of the immune system. Therapeutic strategies based on anti-TIMs mAbs have shown promising results in experimental tumor models, and synergistic combinations of anti-TIMs mAbs with cancer vaccines, adoptive T-cell therapy, radiotherapy and chemotherapy will have great impact on cancer treatment in future clinical development.
Article
Full-text available
Chronic infections with human viruses, such as HIV and HCV, or mouse viruses, such as LCMV or Friend Virus (FV), result in functional exhaustion of CD8(+) T cells. Two main mechanisms have been described that mediate this exhaustion: expression of inhibitory receptors on CD8(+) T cells and expansion of regulatory T cells (Tregs) that suppress CD8(+) T cell activity. Several studies show that blockage of one of these pathways results in reactivation of CD8(+) T cells and partial reduction in chronic viral loads. Using blocking antibodies against PD-1 ligand and Tim-3 and transgenic mice in which Tregs can be selectively ablated, we compared these two treatment strategies and combined them for the first time in a model of chronic retrovirus infection. Blocking inhibitory receptors was more efficient than transient depletion of Tregs in reactivating exhausted CD8(+) T cells and reducing viral set points. However, a combination therapy was superior to any single treatment and further augmented CD8(+) T cell responses and resulted in a sustained reduction in chronic viral loads. These results demonstrate that Tregs and inhibitory receptors are non-overlapping factors in the maintenance of chronic viral infections and that immunotherapies targeting both pathways may be a promising strategy to treat chronic infectious diseases.
Article
Hemiscorpius lepturus scorpionism poses one of the most dangerous health problems in many parts of the world. The common therapy consists of using antivenom antibody fragments derived from a polyclonal immune response raised in horses. However, this immunotherapy creates serious side effects, including anaphylactic shock sometimes even leading to death. Thus, many efforts have been made to introduce new replacement therapeutics that cause less adverse reactions. One of the most attractive approaches to replacing the available therapy is offered by single-domain antibody fragments, or nanobodies (Nbs). We immunized dromedaries with H. lepturus toxin and identified a functional recombinant Nb (referred to as F7Nb) against heminecrolysin (HNc), the major known hemolytic and dermonecrotic fraction of H. lepturus venom. This Nb was retrieved from the immune library by phage display selection. The in vitro neutralization tests indicated that 17.5 nmol of the F7Nb can inhibit 45% of the hemolytic activity of 1 EC100 (7.5 μg/ml) of HNc. The in vivo neutralization tests demonstrated that F7Nb had good antihemolytic and antidermonecrotic effects against HNc in all tested mice. Surprisingly, F7Nb (8.75 nmol) neutralized 1 LD100 of HNc (10 μg) via an intracerebroventricular route or 1 LD100 (80 μg) via a subcutaneous route. All of the control mice died. Hence, this Nb is a potential leading novel candidate for treating H. lepturus scorpionism in the near future.-Yardehnavi, N., Behdani, M., Pooshang Bagheri, K., Mahmoodzadeh, A., Khanahmad, H., Shahbazzadeh, D., Habibi-Anbouhi, M., Hassanzadeh Ghassabeh, G., Muyldermans, S. A camelid antibody candidate for development of a therapeutic agent against Hemiscorpius lepturus envenomation.
Article
CD8(+) CTLs are adept at killing virally infected cells and cancer cells and releasing cytokines (e.g., IFN-γ) to aid this response. However, during cancer and chronic viral infections, such as with HIV, this CTL response is progressively impaired due to a process called T cell exhaustion. Previous work has shown that the glycoprotein T cell Ig and mucin domain-containing protein 3 (Tim-3) plays a functional role in establishing T cell exhaustion. Tim-3 is highly upregulated on virus and tumor Ag-specific CD8(+) T cells, and antagonizing Tim-3 helps restore function of CD8(+) T cells. However, very little is known of how Tim-3 signals in CTLs. In this study, we assessed the role of Tim-3 at the immunological synapse as well as its interaction with proximal TCR signaling molecules in primary human CD8(+) T cells. Tim-3 was found within CD8(+) T cell lipid rafts at the immunological synapse. Blocking Tim-3 resulted in a significantly greater number of stable synapses being formed between Tim-3(hi)CD8(+) T cells and target cells, suggesting that Tim-3 plays a functional role in synapse formation. Further, we confirmed that Tim-3 interacts with Lck, but not the phospho-active form of Lck. Finally, Tim-3 colocalizes with receptor phosphatases CD45 and CD148, an interaction that is enhanced in the presence of the Tim-3 ligand, galectin-9. Thus, Tim-3 interacts with multiple signaling molecules at the immunological synapse, and characterizing these interactions could aid in the development of therapeutics to restore Tim-3-mediated immune dysfunction.