Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Neurobiology of Disease 134 (2020) 104636

Contents lists available at ScienceDirect

Neurobiology of Disease
journal homepage: www.elsevier.com/locate/ynbdi

Review

Active immunization with tau epitope in a mouse model of tauopathy T


induced strong antibody response together with improvement in short
memory and pSer396-tau pathology
A. Joly-Amadoa, ,1, H. Davtyanb,c,1, K. Serraneaua, P. Julesa, A. Zitnyara, E. Pressmana,

K. Zagorskib, T. Antonyanb, A. Hovakimyanb, H.J. Paeke, M.N. Gordona,2, D.H. Cribbsc,


N. Petrovskyd, M.G. Agadjanyanb, A. Ghochikyanb,3, D. Morgana,2,3
a
USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
b
The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
c
Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
d
Flinders Med. Ctr., Bedford Park, Adelaide 5042, Australia
e
Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, USA

ABSTRACT

Abnormal tau hyperphosphorylation and its aggregation into neurofibrillary tangles are a hallmark of tauopathies, neurodegenerative disorders that include Alzheimer's
disease (AD). Active and passive Tau-immunotherapy has been proposed as a therapeutic approach to AD with mixed results. One of the limitations of active im-
munotherapy may be associated with the mediocre immunogenicity of vaccines that are not inducing therapeutically potent titers of antibodies. The aim of this study was to
test the efficacy of an anti-tau vaccine, AV-1980R/A composed of N terminal peptide of this molecule fused with an immunogenic MultiTEP platform and formulated in a
strong adjuvant, AdvaxCpG in a Tg4510 mouse model of tauopathy. Experimental mice were immunized with AV-1980R/A and a control group of mice were injected with
adjuvant only. Nontransgenic and tetracycline transactivator (tTA) transgenic littermates were included as baseline controls to contrast with the tau phenotype. Active
immunization with AV-1980R/A induced very strong anti-tau humoral immune responses in both nontransgenic and transgenic mice with evidence of IgG in brains of AV-
1980R/A vaccinated mice. These experimental animals displayed an improvement in short-term memory during a novel object recognition test. However, impairments in
other behavioral tasks were not prevented by AV-1980R/A vaccinations. At the same time, high titers of anti-tau antibodies reduced hyperphosphorylated pSer396 tau but
did not lower the level of other phosphorylated tau species in the brains of AV-1980R/A vaccinated mice. These data indicate that active immunotherapy with an N-
terminal Tau epitope was only partially effective in improving cognition and reducing pathology in the stringent Tg4510 mouse model of tauopathy.

1. Introduction individuals developed a detrimental T cell- mediated inflammatory re-


sponse (aseptic meningoencephalitis) leading to early termination of the
Alzheimer's disease is the most common form of dementia, in which trial (Orgogozo et al., 2003; Ferrer et al., 2004) indicating that activation
progressive accumulation of amyloid plaques and hyperphosphorylated of autoreactive T cells may infiltrate the brain and cause serious adverse
tau aggregated into neurofibrillary tangles (NFTs) results in progressive events. In addition, we now understand that even though amyloid de-
cognitive impairments. Active and passive immunotherapy targeting Aβ position occurs early in the disease, it doesn't account for clinical
has been successful in multiple AD animal models (reviewed in (Morgan symptoms by itself (Jack et al., 2009). Brain atrophy, neuronal and sy-
et al., 2005, Wisniewski and Boutajangout, 2010, Agadjanyan et al., naptic losses appear to be the key components of cognitive impairments
2015) and has been tested in an increasing number of clinical trials in AD (DeKosky and Scheff, 1990; Terry et al., 1991; Fox et al., 1999),
without much success (Lobello et al., 2012; Lannfelt et al., 2014; Winblad and are more likely to be caused by tau pathology.
et al., 2014; Wisniewski and Goni, 2014; Agadjanyan et al., 2015; Indeed, NFTs are observed early in the pathogenesis of AD and in-
Schilling et al., 2018; Selkoe, 2018; Bachmann et al., 2019; Panza et al., crease during aging (Braak and Braak, 1991). NFTs progression is
2019). In the first active immunization trial with full-length fAβ42 some correlated with cognitive deficits (Duyckaerts et al., 1997), supporting


Corresponding author.
E-mail address: ajoly@usf.edu (A. Joly-Amado).
1
A.J-A. and H.D. contributed equally to this study
2
Present address: 400 Monroe Ave NW, Michigan State University, Grand Rapids MI 49503
3
These senior authors contributed equally to this work

https://doi.org/10.1016/j.nbd.2019.104636
Received 28 May 2019; Received in revised form 30 August 2019; Accepted 7 October 2019
Available online 17 October 2019
0969-9961/ © 2019 Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Fig. 1. Design of the Vaccine Study Conducted in Tg4510 (tetO-MAPT*P301L) Transgenic Mice. Starting at 3 months of age, 3 groups of Tg4510 mice received a total of
7 immunizations with either adjuvant AdvaxCpG (n = 12) or tau vaccine, AV1980R/A (n = 12). Intramuscular injections 1,2,3 were performed every other week at age
3 months, 3.5 months and 4 months. Injection 4,5,6 were performed every other month at 5 months, 6 months and 7 months of age. A final dose was given at 8 months,
one week before euthanization. Due to high immune response, vaccine dose was decreased from 40 micrograms to 20 micrograms after injection 4. Additional groups of
non-transgenic and tTA littermates were used as control for behavior and vaccine titers. Non-transgenic and tTA littermates were not injected with either adjuvant or
antigen. A battery of behavior tests was performed 15 days after injection 6 (end timepoint, at 7 months of age). To assess the efficacy of the vaccine while stopping
transcription of tau, doxycycline was given to the mice in the water during 8 days at a concentration of 200 mg/kg at the end of the last battery of behavior tests. An
additional reversal testing was performed after treatment with doxycycline. Mice were euthanized at 8.5 months of age, 2 weeks after the seventh injection.

a pivotal role for tau pathology and spreading in AD-related memory the first time on immunogenicity of this vaccine in a mouse model of
impairments (Dujardin et al., 2015). As such, there is a possibility that tauopathy and the efficacy of AV-1980R/A on pathology and cognitive
targeting tau may represent a more effective method of treating AD impairments in Tg4510 mice.
than removing Aβ if a patient is already exhibiting clear signs of cog-
nitive impairments. Therefore, development of safe and efficient im-
munotherapy targeting pathological tau could not only benefit AD pa- 2. Materials and methods
tients, but also may become a useful tool against tauopathies in general.
However, as with Aβ immunotherapy, some studies reported in- 2.1. Animals
creased neuroinflammation and encephalopathy following active im-
munization with full length tau (Rosenmann et al., 2006) or phospho- All animal testing procedures were approved by the Institutional
tau epitopes (Rozenstein-Tsalkovich et al., 2013). Furthermore, phos- Animal Care and Use Committee of the University of South Florida and
phorylation is essential for the regulation of tau's normal physiological were performed in accordance with the eighth edition “Guide for the
role in microtubule spatial organization. Therefore, one significant Care and Use of Laboratory Animals,” published by the National
concern that is associated with active tau immunotherapy is that Academy of Science, the National Academies Press, Washington, DC
phospho-tau peptides may induce an immune response against phy- (2011).
siological tau species (Kontsekova et al., 2014). Targeting non-phos- Parental mutant tau and tetracycline-controlled transactivator (tTA;
phorylated epitopes or toxic tau conformation may be necessary. 129S6 background) protein transgenic mouse lines were maintained
Changes in the conformations of tau are particularly important because separately and bred to produce Tg4510 mice, tTA only mice, and
they can directly affect the function of the protein and its toxic role in nontransgenic littermates as described (Santacruz et al., 2005; Dickey
disease. et al., 2009). Mice were housed individually and maintained on a
Recently, the phosphatase-activating domain (PAD) motif, a non- twelve-hour light/dark cycle. Food (Envigo 2018–18% protein- diet)
phosphorylated epitope, was identified in the extreme N-terminus of and water were given ad libitum.
tau (Kanaan et al., 2011). This domain is normally hidden in a paper-
clip-like conformation of the native protein, but becomes exposed in
aggregated, pathological tau (Jeganathan et al., 2006). Abnormal ex- 2.2. Epitope vaccines and purification of proteins
posure of this motif has been linked to dysregulation of axonal transport
and neuronal function (LaPointe et al., 2009; Kanaan et al., 2011; Ward Recombinant protein was purified from E. coli BL21 (DE3) cells
et al., 2012). Immunohistochemical studies of human postmortem tis- transformed with previously constructed pET24a/3Tau2-18-MultiTEP
sues in AD patients demonstrated that exposure of the N-terminal re- plasmids as described (Davtyan et al., 2016). The final recombinant
gion of tau is an early event in AD that increases with progression of the protein was analyzed in 10% Bis-Tris gel electrophoresis (NuPAGE
disease (Kanaan et al., 2012; Ward et al., 2012). Therefore, this PAD Novex Gel, Invitrogen, CA). Protein bands were visualized by Coo-
motif is a reasonable target epitope for active immunotherapy. In fact, massie dye and specificity of the bands was confirmed by Western Blot
intracranial administration of monoclonal antibody targeting PAD (WB) with anti-tau2-18 1C9 monoclonal antibodies (Davtyan et al.,
motif reduced total tau and phosphorylated forms of tau in brains of 2016). The level of endotoxin was measured using E-TOXATE kits, as
Thy-Tau22 tau transgenic mice (Agadjanyan et al., 2017). In addition, recommended by the manufacturer (Sigma, St Louis, MO). For vaccine
we previously reported that a MultiTEP-based recombinant protein preparation the AV-1980R protein was mixed with AdvaxCpG adjuvant.
vaccine, AV-1980R/A targeting PAD was highly immunogenic in Mice were immunized with 40 μg or 20 μg protein (per injection) for-
wildtype C57BL6 mice (Davtyan et al., 2016). Here we are reporting for mulated in 1 mg adjuvant (Fig. 1).

2
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

2.3. Experimental design tone) and exposed to the conditioned stimulus for 3 min (cued tone).
Learning was assessed by measuring freezing behavior (i.e. motionless
Tg4510 mice aged 3 months old received a total of 7 immunizations position) every second.
with either AdvaxCpG adjuvant (n = 12) or the tau vaccine, AV-1980R/
A (n = 12). Mice were injected with 50 μl of indicated antigen in- 2.5. Tissue collection
tramuscularly (into right hind legs tibialis anterior muscle) using in-
sulin syringes (3/10 ml/cc) with the BD Ultra-Fine needle (30G). To assess antibody titers over the course of the experiments, blood
Injection 1, 2, and 3 were performed at biweekly intervals starting at 3 samples were collected from the submandibular vein before (pre-bleed)
mo. of age (Fig. 1). Injections 4, 5, and 6 were performed at 5 mo., 6 and after immunizations 2, 3, 4 and by intracardiac puncture at eu-
mo. and 7 mo. of age. A final dose was given after 8 mo, one week prior thanasia. Blood was collected without anti-coagulant, incubated one
to tissue collection. Due to the strong antibody response (see below), hour at room temperature and then kept overnight at 4 °C. The next day
the vaccine dose was decreased from 40 micrograms to 20 micrograms samples were centrifuged for 10 min at 4000 rpm at room temperature
after injection 4. Additional groups of non-transgenic and tTA litter- (RT), in a bench-top centrifuge. The serum (upper phase) was carefully
mates were used as controls for behavior and vaccine titers. A battery of collected and centrifuged for 10 min at 7000 rpm. The supernatant was
behavioral tests was performed 15 days after injection 6 (at 7 mo. of collected and kept at -80 °C for antibody titer measurements.
age). To assess the efficacy of the vaccine while stopping transcription One week after the last injection (injection 7), mice were euthanized
of tau, doxycycline was given to the Tg4510 mice via drinking water for at 8.5 months of age with a solution containing pentobarbital and
8 days at a concentration of 200 mg/kg at the end of the second battery phenytoin, then transcardially perfused with 25 ml of 0.9% normal
of behavior tests. An additional reversal testing in the radial arm water saline solution. Before perfusion, spleens were collected and single cell
maze was performed after treatment with doxycycline. Mice of both suspension was prepared as described in (Davtyan et al., 2014a, 2014b)
sexes were included in each treatment condition. using RBC (Red Blood Cells) Lysing Buffer (Sigma-Aldrich).
Brains were collected immediately following perfusion. The brain
2.4. Behavioral testing was trimmed to exclude olfactory bulbs or spinal cord and weighed.
One hemisphere was dissected and frozen for western blot analysis and
All behavioral testing was performed in groups of mice balanced for the second hemisphere was immersion fixed in freshly-prepared 4%
gender by an observer unaware of the treatment/genotype of the mice. phosphate-buffered paraformaldehyde for 24 h. The fixed hemispheres
The open field was used as a standard test of general activity. Animals were cryoprotected in successive incubations of 10%, 20% and 30%
were monitored for 15 min in a 40 cm square open field with video sucrose solutions for 24 h each. Subsequently, brains were frozen on a
tracking software (ANY-Maze, Stoelting, IL), under moderate lighting. cold stage and sectioned in the horizontal plane (25 μm thickness) on a
General activity levels were evaluated by measurements of horizontal sliding microtome and stored in Dulbecco's phosphate buffered saline
and vertical activity. with 10 mM sodium azide solution at 4 °C.
Each animal was placed in a walled Y-maze for a single 5 min trial.
The sequence of arm entries and total number of arm choices were 2.6. Detection of splenocytes producing cytokines
recorded. Spontaneous alternation (entering all three arms sequentially
without repetition) was expressed as a percentage, as calculated ac- Cytokine CBA (Cytometric bead array) assay. Concentrations (pg/
cording to the method of (Anisman, 1975). ml) of Th1 [IFN-γ, IL-2 and THF-α] and Th2 [IL-4 and IL-5] cytokines
Short term memory was evaluated by the novel object recognition were detected using Mouse Th1/Th2 Cytokine CBA kit by flow cyto-
test (NOR) which consists of a 40 × 40 cm arena monitored and metry according to manufacturer's instructions. Splenocytes were re-
quantified by video tracking (ANY-Maze, Stoelting, IL). Two objects stimulated in vitro with a cocktail of 12 Th epitope peptides (2 μg/ml
similar in scale to the mouse were placed along the center line of the each) for 24 h and supernatant was collected for assay.
arena approximately 3–5 cm from the outside wall. Each animal was ELISpot assay. Analysis of IFN-γ producing T cells was performed in
given three acclimation trials of 5 min each with a 5 min inter-trial splenocyte cultures from immunized mice by ELISpot assay (BD
interval. After each trial, the arena and object cues were cleaned with Biosciences, CA), as previously described (Cribbs et al., 2003,
70% ethanol to minimize olfactory cues. After the acclimation trials, Petrushina et al., 2007, Davtyan et al., 2013, Davtyan et al., 2014a,
one of the acclimated objects was replaced with a novel object. Animals 2014b). Cultures of splenocytes were re-stimulated in vitro with a
were given a 5 min exploratory trial during which object exploration cocktail of 12 peptides representing the Th epitopes in the MultiTEP
was monitored by video recording. Working memory was evaluated by vaccine (Davtyan et al., 2014a, 2014b) (2 μg/ml of each peptide), and
the discrimination ratio, which is defined as the time exploring the several individual peptides [PADRE, P23, P30, P17, P28] incorporated
novel object minus the time spent with familiar and divided by com- in MultiTEP platform, as well as Tau2-18 peptides at concentration of
bined time spent exploring both novel and familiar objects. 10 μg/ml for 20 h. The numbers of IFN-γ spot-forming cells (SFC) per
A detailed description of radial arm water maze (RAWM) has been 106 splenocytes stimulated by different peptides were then counted.
published previously (Alamed et al., 2006). For fear conditioning, a
0.5 mA mild foot shock was paired with an auditory conditioned sti- 2.7. Detection of B cells producing tau-specific antibodies
mulus within a novel environment. Freezing on the training day in
response to the foot shock was used as an estimate of learning during Antibody-secreting B cells specific to tau were detected in spleno-
the acquisition trial. Animals were placed in the fear conditioning ap- cytes by ELISpot (Mabtech Inc., Cincinnati, OH) as described in
paratus for 3 min, and then a 30 s noise at 70 dB was delivered with a Davtyan et al. (Davtyan et al., 2016). Briefly, splenocytes from ex-
0.5-mA shock applied to the floor grid during the last 2 s of the con- perimental and control mice were incubated for 24 h in 96-well plates
ditioned stimulus. Training consisted of two mild shocks paired with coated with tau2-18 peptides. After incubation the assay was performed
two conditioned stimuli with a 2 min interval between each shock. For as recommended by the manufacturer.
contextual memory, the mice were placed in the acquisition chamber
and monitored for freezing to the context 24 h after training (no shocks 2.8. Detection of tau-specific antibodies
or auditory cue given) and tested for 3 min. Immediately after the
contextual test, mice were placed in a novel environment, consisting of The concentrations of anti-tau antibodies in serum was determined
a chamber with different shape, floor and olfactory cues from the by ELISA, as described previously (Davtyan et al., 2016). To measure
training chamber. Mice were allowed to explore it for 3 min (cued no anti-tau antibody concentration plates were coated with 1 μg/per well

3
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

with tau2-18 peptide (GenScript, NJ). Anti-tau antibody concentration ThermoFisher Scientific, MA), according to the manufacturer's instruc-
was calculated using a calibration curve generated with 1C9 mAb tions.
(generated at The Institute for Molecular Medicine, Huntington Beach,
CA). HRP-conjugated anti-mouse IgG (Jackson ImmunoResearch La- 2.11. Statistical methods
boratories, ME) was used as secondary antibody.
Data were analyzed by ANOVA or repeated measures ANOVA fol-
2.9. Histopathology lowed by Fisher's protected least significant difference post hoc tests
(statview) or ANCOVA. In all tests, a P-value < 0.05 was considered
Stereological principles were employed to select evenly spaced significant. All data are presented as mean values ± standard error of
sections to be stained for each marker. Immunohistochemical proce- the mean (SEM) unless specified otherwise.
dural methods were as described previously (Gordon et al., 2002).
Sections from all animals were placed in a multi-sample staining tray 3. Results
and endogenous peroxidase was blocked (10% methanol, 3% H2O2 in
phosphate buffered saline, 10 mM NaPO4, 154 NaCl, pH 7.4, 3.1. Immunogenic efficacy of AD vaccine in Tg4510 mice
PBS;30 min). Tissue samples were permeabilized with 0.2% lysine,
1%Triton X-100 in PBS solution and incubated overnight in anti-tau Recently we demonstrated that MultiTEP-based recombinant pro-
phosphorylated at Ser396 (rabbit polyclonal, Anaspec), total tau H150 tein vaccine AV-1980R/A (tau vaccine) is highly immunogenic in
(rabbit polyclonal, SantaCruz Biotechnology), anti-tau phosphorylated wildtype C57BL6 mice (Davtyan et al., 2016). To test the immunogenic
at AT8 (Thermo scientific, Waltham, MA), Iba-1 (Wako), GFAP (Dako), and therapeutic efficacy of tau vaccination in tau transgenic mice, 3-
CD86 (Novus biologicals, Littleton CO). Additionally, we examined the month-old Tg4510 mice were immunized with AV-1980R/A, while
staining for mouse IgG with biotinylated horse anti-mouse IgG specific control groups injected with adjuvant (AdvaxCpG). Mice received seven
secondary antibody (Vector Laboratories, Burlingame, CA, USA) to injections at the time points shown in Fig. 1.
detect whether the systemically injected antibodies may have bound to Cellular immune responses were measured in splenocytes of vacci-
brain tissue. Sections were washed in PBS, and then incubated in bio- nated mice by detection of production of Th1/Th2 cytokines (Fig. 2). As
tinylated secondary antibody (Vector Laboratories, Burlingame, CA). expected, AV-1980R/A vaccine generated a strong cellular immune
The tissue was again washed after 2 h and incubated with Vectastain® response specific to the Th cell epitopes incorporated in the MultiTEP
Elite® ABC kit (Vector Laboratories, Burlingame, CA) for enzyme con- platform. Next, we tested Th cell immune responses specific to in-
jugation. Finally, sections were stained using 0.05% diaminobenzidine, dividual epitopes incorporated into the MultiTEP vaccine platform.
0.5% nickel ammonium sulfate and 0.03% H2O2. Tissue sections were ELISpot data presented in Fig. 3 demonstrated that vaccination with
mounted onto slides, dehydrated, cover slipped and scanned for ana- AV-1980R/A stimulated Th cells specific to epitopes PADRE, P23, P30,
lysis using a digital scanning microscope Axio Scan Z.1 (Zeiss Inc). P17 and P28 in this mouse strain possessing an H-2b/q immune haplo-
Digitized sections were annotated for regions of interest and positively type. Interestingly, this repertoire was slightly different in C57BL6 (H-2b)
stained area was quantified with Nearcyte software. Values for all and in THY-Tau22 strain, which was created on H-2b/k background and
sections from the same mouse were averaged to represent a single value was backcrossed with C57BL6 mice (Schindowski et al., 2006). Of note,
for that region in subsequent statistical analysis. we detected significantly higher numbers of Th cells specific to PADRE,
which is a promiscuous universal synthetic epitope, compared with P23,
2.10. Biochemical analyses P30, P17 and P28 peptides in immune splenocytes isolated from tau
vaccinated mice. High numbers of activated Th cells producing both Th1
Tissues for Western blot analysis were prepared as previously de- and Th2 types of cytokines indicated that tau vaccine induced mixed
scribed (Brownlow et al., 2014). Dissected hippocampi (HPC) and pos- Th1/Th2 types of immune responses. Importantly, no activation of po-
terior cortex (PCX) were homogenized then sonicated in RIPA buffer tentially harmful autoreactive Th cells was detected after re-stimulation
containing protease inhibitor cocktail (Sigma Aldrich) and phosphatase of immune splenocytes with tau self-epitope by ELISpot assay (Fig. 3). As
inhibitor cocktails I and II (Sigma Aldrich) and centrifuged at 40, 000 ×sg might be expected, strong cellular immune responses specific to the
for 30 min at 4 °C. The supernatant was collected (RIPA soluble fraction) MultiTEP vaccine platform should stimulate B cells to produce antibodies
and protein concentrations were determined by the BCA protein assay kit specific to tau2-18 epitope incorporated into AV-1980R/A vaccines. The
(Pierce, Rockford, IL). The remaining pellet was digested with 70% formic ELISpot data confirms this assumption and demonstrates that vaccination
acid according to the wet tissue weight, and then neutralized with NaOH generated a large numbers of anti-tau antibody-secreting B cells (ASC)
to analyze RIPA-insoluble proteins (RIPA insoluble fraction). Equal after immunization with AV-1980R/A (Fig. 4A). No cross-reactive anti-
amounts of proteins according to BCA (5 μg/well for soluble fraction, body-producing B cells were detected in splenocytes isolated from im-
1 μg/well for insoluble fraction) were loaded in each well of a 4–12% Bis- munized mice (Fig. 4A). More importantly, these tau specific B cells
tris gels and transferred to a 0.2 μm pore size nitrocellulose membrane activated by vaccination generated extremely strong humoral immune
and immunoblotted with H150 (Santacruz biotechnology, Dallas, TX), responses after only two immunizations with average concentrations of
pSer396-tau (Anaspec, Fremont, CA), N-term Tau 12 (biolegend, San antibodies close to 2 mg/ml, > 10% of total serum IgG. This level of
Diego, CA) and actin (Sigma Aldrich, St Louis, MO) at 1:1000-fold dilu- antibodies was maintained throughout the entire study (Fig. 4B).
tion. Fluorescently tagged secondary antibodies (IRDye 800CW, LI-COR Finally, to characterize the type of humoral immune responses, we
Biosciences) were used at a dilution of 1:10,000. Western Blot results measured the production of IgG1, IgG2ab, IgG2b, and IgM isotypes in mice
were quantified by scanning with a LI-COR Odyssey fluorescent scanner. immunized with AV-1980R/A vaccine. Mice from both groups generated
Band intensities were quantified by densitometric analysis using the mostly IgG antibodies (IgG1 > IgG2b > IgG2ab), while only negligible
Odyssey imaging system (LI-COR Biosciences) and normalized to the band titers of IgM antibodies were detected in the sera of immunized mice (data
intensity of β-actin (for RIPA soluble fraction) or to total protein (RIPA not shown). Of note, IgG1/IgG2ab is equal to 4 for AV-1980R/A indicating
insoluble fraction). Total protein was detected using REVERT reagent (LI- that the immune response is biased towards Th2 type.
COR) according to the manufacturer's instructions.
In addition, concentrations of human total and phosphorylated tau in 3.2. Impact of vaccination on cognitive impairments
soluble brain extracts were determined by Tau (total) Human ELISA kit,
Tau [pS396] Human ELISA Kit, Tau [pS199] Human ELISA Kit, Tau Spatial memory deficits were assessed by the 2-day radial arm water
[pT181] Human ELISA Kit, and Tau [pT231] Human ELISA Kit (all from maze test. As previously shown, repeated measure ANOVA revealed a

4
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Fig. 2. AV-1980R/A formulated in AdvaxCpG adjuvant generated both Th1 and Th2 immune responses in Tg4510 mice. Concentrations (pg/ml) of Th1 cytokines (A, B, C) and
Th2 cytokines (D, E) in response to AdvaxCpG adjuvant (open circle) and AV-1980R/A tau vaccine (black square) in Tg4510 mice. [IFN-γ: Interferon-gamma, IL-2: Interleukin 2,
TNF-α: Tumor necrosis factor alpha, IL-4: Interleukin 4, IL-5: Interleukin 5]. Cytokines were detected using Mouse Th1/Th2 Cytokine CBA kit by flow cytometry. Splenocytes
were re-stimulated in vitro with a cocktail of 12 Th epitope peptides (2 μg/ml each) for 24 h and supernatant was collected for assay. Bars represent mean ± SEM (n = 6 mice/
per group). Statistical significance was calculated against AdvaxCpG group using unpaired t-test. (*P < 0.05, ***P < 0.001 and ****P < 0.0001).

genotype effect: non-transgenic and tTA control mice learned the lo- transgenic and tTA mice, indicating no effect of the vaccine on spatial
cation of the platform by the end of day 1 and made few errors on day navigation memory.
2. Tg4510 mice treated with adjuvant were not able to remember the Conversely, we observed an improvement in short term memory
location of the platform as they made significantly more errors on day 1 assessed by novel object recognition (NOR) in mice vaccinated against
and day 2 when compared to non-transgenic or tTA control mice tau. As expected, two-way ANOVA revealed a genotype effect as non-
(Fig. 5A). There was no significant improvement in the number of er- transgenic and tTA control mice spent more time with the novel object
rors in Tg4510 vaccinated with AV-1980R/A compared to adjuvant than did the adjuvant Tg4510 mice (Fig. 5B). However, Tg4510 mice
treated Tg4510 mice. All Tg4510 mice performed worse than the non- vaccinated with AV-1980R/A displayed a significant increase in time

Fig. 3. AV-1980R/A induced strong cellular response specific to Th epitopes incorporated in the MultiTEP platform without activating potentially harmful auto-
reactive Th cells specific to tau. Splenocytes from individual mice were re-stimulated in vitro with a cocktail of 12 T-helper epitope peptides (2 μg/ml each) and
several individual peptides [PADRE, P23, P30, P17, P28] incorporated in MultiTEP platform, as well as Tau2-18 peptide (10 μg/ml). IFNγ spot-forming colonies (SFC)
were detected by ELISPOT assay and error bars indicate mean ± SEM (n = 11 per group).

5
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Fig. 4. AV-1980R/A induced specific B cells producing very high concentrations of antibodies.
Evaluation of humoral immune responses generated in Tg4510 transgenic mice after immunizations with AV-1980R vaccine formulated with AdvaxCpG adjuvant. (A)
Detection of anti-tau antibody-secreting cells (ASC), visualized as spots, was done in splenocyte cultures obtained from experimental and control mice using ELISpot
assay. (B) Concentrations of anti-tau antibodies in mouse sera were determined by ELISA. Plates were coated with Tau2-18 peptide (Genscript). Anti-tau antibody
concentrations were calculated using a calibration curve generated with 1C9 anti-tau2-18 monoclonal antibody. Bars represent average ± SD (n = 11/per group).

attending to the novel object when compared to Tg4510 adjuvant the antibodies produced during vaccination were reaching the brain,
treated mice. In addition, AV-1980R/A vaccinated mice achieved the we incubated brain sections with an anti-mouse IgG specific secondary
same discrimination ratio as tTA and non-transgenic control mice. A antibody. Quantification of positive area stained with anti-mouse IgG
battery of other cognitive tests was performed (Table 1). As previously was performed in anterior cortex (ACX), posterior cortex (PCX), and
shown, we observed a genotype effect in total distance travelled during hippocampus (HPC) calculated from digitized images is shown in
open field, number of entries in Y maze, contextual fear conditioning Fig. 6A. Tg4510 mice vaccinated with tau epitope exhibited sig-
and nesting behavior. Vaccination with AV-1980R/A had not a sig- nificantly higher levels of IgG in the ACX, HPC and PCX when compared
nificant influence on these cognitive deficits. to Tg4510 mice treated with adjuvant. When the parenchymal staining
in the posterior cortex was viewed at higher magnification, there was
3.3. Vaccination with Tau epitope induced increased levels of mouse IgG in staining of the neuropil only in Tg4510 mice vaccinated with AV-
the brain 1980R/A (Fig. 6B, panels b, d). No positive staining was detected in
Tg4510 mice treated with AdvaxCpG adjuvant (Fig. 6B, panels a, c).
As shown in Fig. 4, very high concentrations of antibodies were Mouse IgG staining was also absent in non-transgenic and tTA control
produced after vaccination with AV-1980R/A. To investigate whether mice (data not shown).

Fig. 5. Vaccination with AV-1980R/A did not improve spatial memory during radial arm water maze testing but rescued the short-term memory deficit observed in
Tg4510 during the novel object recognition test.
(A) Tg4510 mice made significantly more errors regardless of treatment with AdvaxCpG adjuvant or AV-1980R/A when attempting to locate a hidden platform during
the 2-day radial arm water maze (RAWM) test when compared to non-transgenic (Ntg) and tTA littermates. Data are presented as mean ± S.E.M., n = 12/group.
***p < 0.001. (B) Tg4510 mice treated with AdvaxCpG adjuvant spent less time with novel object compared to non-transgenic (Ntg) and tTA littermates as they
displayed a lower discrimination ratio (which is defined as the time exploring the novel object minus the time spent with familiar and divided by combined time
spent exploring both novel and familiar objects), indicating short term memory deficit. Tg4510 mice vaccinated with AV-1980R/A displayed a significant increase in
discrimination ratio compared to control Tg4510 mice treated with AdvaxCpG adjuvant indicating an improvement in short memory. Data are presented as
mean ± S.E.M., n = 12/group. *p < 0.05. 2-way ANOVA test was used for RAWM with Tukey's LSD means comparisons for significant overall effects. A t-test was
performed to compare the two Tg4510 groups for NOR.

6
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Table 1
Summary of behavior tests performances in non-transgenic (Ntg), tet only (tTA) and Tg4510 mice treated with AdvaxCpG adjuvant or AV-1980R/A. Open field total
distance travelled in meters (TDT), Ymaze entries and alternations, Radial arm water maze (RAWM) total errors, Fear conditioning (FC) percentage time freezing and
nesting behavior.
Ntg Tta AdvaxCpG AV-1980R/A

a,b a,b
Open field (TDT) 36.7 +/− 2.8 46.9 +/− 2.5 150.3 +/− 35.3 122.9 +/− 32.2
Ymaze entries 32.8 +/− 1.9a,b 34.3 +/− 1.3a,b 57.8 +/− 9 59.5 +/− 7.9
Ymaze alternation 60.7 +/− 3.2 59.9 ± 1.6 65.1 +/− 2.3 68.2 +/− 4.8
RAWM reversal total errors 25 +/− 4.7 a,b,c 40.1 +/− 2.7a,b 92.8 +/− 13.3 93.4 +/− 14.6
RAWM post dox reversal total errors 29.5 +/− 5.4a,b 52.9 +/− 8.3 81.9 +/− 14.3 70.5 +/− 7.8
FC context 25.6 +/− 5.4a,b 20.7 +/− 4.7a,b 6.6 +/− 2.3 2.6 +/− 1.2
FC cued (tone) 42.1 +/− 7 26.4 +/− 5.1 27.7 +/− 8 33.6 +/− 6.7
Nesting behavior 3.8 +/− 0.3a,b 3.9 +/− 0.2a,b 1.1 +/− 0.5 0.9 +/− 0.4

p < 0.05 when compared to AdvaxCpG adjuvant.


a

b
p < 0.05 when compared to AV-1980R/A.
p < 0.05 when compared to tTA. As expected, Tg4510 treated with AdvaxCpG adjuvant displayed hyperactivity and cognitive impairments when compared to
c

non-transgenic or tTA littermates. Immunization with AV-1980R/A did not affect these phenotypes.

Fig. 6. Tg4510 mice vaccinated with Tau vaccine AV-1980R/A displayed increased levels of mouse IgG in the brain compared to AdvaxCpG.
(A) Quantification of positive area stained for mouse IgG in anterior cortex (ACX), hippocampus (HPC) and posterior cortex (PCX) in Tg4510 treated with AdvaxCpG
adjuvant or with AV-1980R/A mice. Non-transgenic and tTA littermates displayed similar levels to Tg4510 treated with AdvaxCpG adjuvant (data not shown).
Immunostaining quantification was performed utilizing Mirax software (Zeiss Inc.). (B) Micrographic representation of Mouse IgG immunostaining in the posterior
cortex of Tg4510 mice treated with AdvaxCpG adjuvant (a, c) or AV-1980R/A (b, d). c and d are magnification of square area in a, b, respectively. Tg4510 mice
immunized with AV-1980R/A seemed to display positive staining in cell bodies particularly in the posterior cortex. Data are presented as mean ± S.E.M., n = 12/
group. Statistical significance was calculated against AdvaxCpG group using unpaired t-test *p < 0.05.

3.4. Tau pathology in Tg4510 vaccinated mice was a trend towards decrease in levels of pSer396-tau in AV-1980R/A
vaccinated mice compared to AdvaxCpG adjuvant treated mice that did
We then evaluated the impact of the different vaccines on tau pa- not reach significance (Fig. 9).
thology. Quantification of positive immunostaining for pSer396-tau ELISA assay performed for total tau and several phosphorylated tau
(phosphorylated tau) and H150 (total tau) in the entire hemisection forms in soluble extractions revealed a significant decrease in pSer396-
(total), anterior cortex (ACX), hippocampus (HPC) and posterior cortex tau and total tau but not in pSer199-, pT181- and pT231-tau, in AV-
(PCX) are shown in Fig. 7. There was a trend towards decrease in total 1980R/A vaccinated mice when compared to AdvaxCpG adjuvant treated
area as well as ACX and PCX in AV-1980R/A vaccinated mice when mice (Fig. 10).
compared to AdvaxCpG adjuvant-treated mice, but the difference did not Inflammation markers and brain weight, which have been pre-
reach significance (Fig. 7A). A similar trend was observed in AT8 or viously shown to be affected by tau pathology in Tg4510 (Wes et al.,
Gallyas silver stained sections (Table 2). No difference was observed in 2014; Joly-Amado et al., 2016) were also analyzed (Table 2). Im-
total tau levels detected by immunohistochemistry as shown in Fig. 7B, munohistochemistry for Iba-1 and CD86 (microglia) as well as GFAP
D. As expected, no positive staining was detected in non-transgenic (astrocytes) and brain weight showed genotype effects but no treatment
animals for pSer396-tau or H150 (data not shown). The adjuvant effect.
treated Tg4510 mice had values similar to historical values for un-
treated Tg4510 mice of the same age (Fig. S3). 4. Discussion
Western blot analysis revealed a decrease in pSer396-tau levels
(Fig. 8B,E) but not H150 (Fig. 8A,D) or Tau12 (anti Nterm tau aa6-18; The “amyloid cascade hypothesis” considers pathological Aβ as a
Fig. 8C,F) in the RIPA- soluble fraction of the posterior cortex in AV- triggering toxic event in AD and most clinical immunotherapy ap-
1980R/A vaccinated mice compared to AdvaxCpG adjuvant treated mice proaches have been aimed at reducing Aβ deposits. However, accu-
(see Fig. S1 for western immunoblot of all samples). This was the case mulating evidence show that pathological tau correlates with the onset
whether normalized to actin (Fig. 8) or when the phospho-epitopes of symptoms and the degree of dementia better than Aβ deposition. It is
were normalized to total tau (Fig. 8G,H and Fig. S2). There were no important to note that the lessons from clinical trials of active and
differences in the levels of these markers in PCX RIPA insoluble fraction passive immunization targeting Aβ suggest that anti-Aβ im-
(data not shown). In the hippocampus RIPA insoluble fraction, there munotherapy may be effective in the early/preclinical stages of AD,

7
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Fig. 7. Vaccination with AV-1980R/A did not affect levels of pathology in Tg4510 mice when assessed by immunohistochemistry.
Quantification of positive area stained for pSer396 (A) and H150 (B) in total area (total), anterior cortex (ACX), hippocampus (HPC) and posterior cortex (PCX) in
Tg4510 mice treated with AdvaxCpG adjuvant or with AV-1980R. Immunostaining quantification was performed utilizing Mirax software (Zeiss Inc.). Micrographic
representation of pSer396 (C) and H150 (D) immunostaining in the hippocampus of Tg4510 mice treated with AdvaxCpG adjuvant (C, D left panel), and AV-1980R (C,
D right panel). Data are presented as mean ± S.E.M., n = 12/group. Statistical significance was calculated against AdvaxCpG group using unpaired t-test *p < 0.05.

whereas in the later stages, clearing of Aβ alone may not be sufficient to domain antibodies more effective in blocking tau seeding and propaga-
stop/delay the disease. Therefore, anti-tau immunotherapy (passive tion. Thus, it seems likely that the properties of the antibody other than
and active immunizations) was proposed as an additional therapeutic its domain specificity may have more influence on efficacy. However, it
approach. seems quite feasible from the mouse studies that anti-tau immunotherapy
The first attempts at tau immunotherapy involved the active im- could reduce both tau and amyloid pathologies in cases of AD.
munization of wild type mice with full-length recombinant human tau Two active vaccines (Godyn et al., 2016; Novak et al., 2019) are
protein (Rosenmann et al., 2006), followed by experiments using a currently being tested in clinical trials with AD patients. Although these
variety of immunogens of tau and adjuvants in many different animal trials are in the early stages and are intended for safety studies, some
models of tauopathy (reviewed in (Schroeder et al., 2016). In general, preliminary efficacy data were published from AADVac1 trial, which
active immunization was efficient at reducing tau pathology in mice as reported that a trend towards slower cognitive decline and lower hip-
indicated by decreased phosphorylated tau (Boutajangout et al., 2010; pocampal atrophy rate was observed in patients with higher antibody
Troquier et al., 2012; Selenica et al., 2014) and NFTs (Boimel et al., titers (Novak et al., 2018).
2010). Some studies also demonstrated cognitive improvements There are advantages and disadvantages of both active and passive
(Boutajangout et al., 2010; Benhamron et al., 2018) or improved motor immunotherapy. Active immunotherapy has the advantage of requiring
performance (Asuni et al., 2007). Passive immunotherapy studies in fewer doses and is more cost effective than passive immunotherapy. The
animal models also showed promising results when administered early in disadvantages are that the antibody response is variable both quantita-
the disease process. Passive immunization with PHF1 or MC-1 antibody tively and qualitatively (especially in older adults) and adverse effects
decreased tau pathology in JNPL3 (Boutajangout et al., 2011; Chai et al., are challenging to reverse. Passive immunotherapy has the advantage of
2011; d'Abramo et al., 2013) and P301S (PS19) mice (Chai et al., 2011). known dosing with an antibody with known characteristics, and if ad-
Targeting oligomeric tau was efficient in JNPL3 mice (Castillo-Carranza verse events ensue, the antibody can be discontinued. Disadvantages
et al., 2014) but not in Tg4510 (Schroeder et al., 2017). include a high frequency of dosing (typically monthly by infusion) and
More recent work has identified some intriguing abilities of tau im- the considerable costs associated with antibody production and delivery.
munotherapy to clear not only tau deposits, but also Aβ deposits in mice. In this study, we tested the immunogenicity and therapeutic efficacy
In 3xTg mice, Dai et al. (Dai et al., 2017) found that an N-terminal anti- of AV-1980R/A MultiTEP platform based active vaccine targeting PAD
tau antibody was more effective than a mid-domain antibody in reducing (Tau2-18) region of tau in Tg4510 mice. MultiTEP platform composed
tau, but especially in reducing amyloid deposits also in these mice. Both of 12 foreign Th cell epitopes, was specifically designed to enhance
antibodies rescued behavioral deficits. In follow up work, Dai et al. (Dai immune responses in elderly patients with immunosenescence and
et al., 2018) also demonstrated the N-terminal antibody could block tau provide a broad coverage of human MHC class II polymorphism uti-
propagation in 3xtg mice caused by injecting AD-tau seeds. Rajamoha- lizing the wide array of tetanus toxin, hepatitis B and influenza Th
medsait H et al. (Rajamohamedsait et al., 2017) found that active im- epitopes incorporated into the MultiTEP platform.
munization of 3xTg mice partially reduced tau deposition but con- In Tg4510 mice, the vaccine induced strong cellular responses
siderably blocked amyloid deposition.Benhamron et al. (Benhamron specific to foreign Th epitopes that provided help to B cells to be acti-
et al., 2018) found that a phospho-tau vaccine was capable of reducing vated and produce very high titers of antibodies against tau, achieving
Aβ and improving memory in an APP/PS1 mouse model known to have 2 mg/ml or approximately 10% of the IgG concentration in mice.
minimal tau pathology. In a different comparison of N-terminal and mid- Moreover, no tau specific T cell response was detected indicating that
domain antibodies, Albert et al. (Albert et al., 2019) found the mid- the likelihood that this vaccine would produce autoreactive

8
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

(caption on next page)

9
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Fig. 8. Tg4510 mice vaccinated with tau vaccine AV-1980R/A display decreased levels of phospho tau in the posterior cortex soluble fraction when compared to
AdvaxCpG adjuvant treated mice.
Phospho tau (pSer396), total tau (H150) and Nterm tau (Tau12) were assessed by western blot analysis (A, B, C representative WB images) in posterior cortex in RIPA
soluble fraction.55 kda, 64 kda and combination of 55-64kda band intensity were quantified for H150 (D), pSer396 (E) and Tau12 (F) in Tg4510 mice treated with
AdvaxCpG adjuvant or with AV-1980R. All groups were normalized to the adjuvant group and to actin. No bands were detected in non-transgenic (Ntg) and tet only
(tTA) mice, as shown in image A, B, C. Ratio of Pser396 and Tau12 to H150 was calculated for bands 55–64 (G, H) and for bands 55 and 64 separately (Fig. S2). Data
are presented as mean ± S.E.M., n = 12/group. Statistical significance was calculated against AdvaxCpG group using unpaired t-test *p < 0.05.

Fig. 9. Tg4510 mice vaccinated with tau vaccine AV-1980R/A display a trend towards decrease of phospho Tau in the hippocampus insoluble fraction when
compared to AdvaxCpG.
Phospho Tau (pSer396) and total Tau (H150) was assessed by western blot analysis (A, representative WB images) in hippocampus in RIPA insoluble fraction. Briefly,
after high centrifugation, pellet was treated with formic acid and neutralized with NaOH. Band intensity was quantified for H150 (B) and pSer396 (C) as well as ratio
of pSer396 over H150 (D) in Tg4510 mice treated with AdvaxCpG adjuvant, or AV-1980R/A. All groups were normalized to the adjuvant group. No bands were
detected in non-transgenic (Ntg) and tet only (tTA) mice, as shown in image A. Data are presented as mean ± S.E.M., n = 12/group. Statistical significance was
calculated against AdvaxCpG group using non parametric Mann Whitney test.

Table 2
Brain weight at euthanasia (in mg).
Ntg tTA AdvaxCpG AV-1980R/A

Brain weight (mg) 503.9 +/− 6.1a,b,c 474.6 +/− 6.5a,b,c 427.9 +/− 13 426 +/− 11.6
Gallyas stain – – 1.0 ± 0.21 0.92 ± 0.12
Immunohistochemistry AT8 – – 1.0 ± 0.16 0.84 ± 0.12
gfap 0.54 ± 0.05a,b 0.56 ± 0.05a,b 1.0 ± 0.10 1.13 ± 0.06
iba-1 0.77 ± 0.04a,b 0.76 ± 0.05a,b 1.0 ± 0.04 0.96 ± 0.07
cd86 total 1.04 ± 0.11 0.81 ± 0.09b 1.0 ± 0.08 1.12 ± 0.09

Positive total area stained with Gallyas, or for AT8 (phospho Tau Ser202/Thr205), Glial fibrillary acidic protein (GFAP), Ionized calcium binding adaptor molecule 1
(Iba-1) and cluster of differentiation 86 (CD86). As expected, Tg4510 treated with AdvaxCpG adjuvant displayed decreased brain weight and increased positive area
ratio for GFAP, Iba-1 and CD86 when compared to non-transgenic or tTA littermates. Immunization with AV-1980R/A did not affect these phenotypes nor did it
affect levels of phospho tau (pSer396), and total tau (H150) assessed by western blot analysis in hippocampus in RIPA soluble fraction, or positive area ratio for AT8.
Statistical significance was calculated against using unpaired t-test or ANOVA.
p < 0.05 when compared to AdvaxCpG adjuvant.
a

b
p < 0.05 when compared to AV-1980R/A.
c
p < 0.05 when compared to tTA.

10
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Fig. 10. Tg4510 mice vaccinated with AV-1980R/A display decreased levels of phosphorylated tau and total tau in the posterior cortex soluble fraction when
compared to AdvaxCpG adjuvant treated mice when analyzed by ELISA.
Level of total (A) and several phosphorylated tau (pSer396 (B), pSer199 (C), pT181(D), pT231(E), in brain soluble extractions of posterior cortex from Tg4510 mice
treated with AdvaxCpG adjuvant or AV-1980R/A, were analyzed by ELISA. Error bars represent average ± SEM (n = 11–12) Statistical significance was calculated
against AdvaxCpG group using unpaired t-test. **p < 0.01.

inflammatory T cell responses against tau in brain is very low. using the phosphatase-activating domain (PAD) of tau protein as im-
The Tg4510 mice developed the expected behavioral phenotype, with munogen was efficient at causing high titers of anti-tau antibodies with
impaired performance in open field, contextual fear conditioning, novel little risk of developing auto-reactive T cells. The vaccine was effective
object recognition, radial arm water maze and nesting behavior. There at modestly reducing some aspects of tau pathology and improving
was a significant rescue of the novel object recognition behavior back to some components of cognitive performance in an aggressive model of
levels similar to nontransgenic mice. However, there was no indication of tauopathy, the Tg4510 mouse.
improvements in any of the other behavioral tests. These results are
consistent with our prior work. In Brownlow et al. (2014), we found that Declaration of Competing Interest
caloric restriction in Tg4510 mice caused significant improvements in
the novel object task but not in other measures of the behavioral phe- MGA and AG are co-founders of Capo Therapeutics that licensed
notype. This occurred with modest trends in reversal of the tau pa- MultiTEP vaccine platform technology from the Institute for Molecular
thology, none of which were statistically significant (Brownlow et al., Medicine. The remaining authors declare no financial, commercial and
2014). These results would suggest that the relatively simple novel object non-financial conflict of interests.
recognition task is easier for mice to perform. Hence even small changes Supplementary data to this article can be found online at https://
in the tau deposition are capable of rescuing this deficit, but not the doi.org/10.1016/j.nbd.2019.104636.
deficits associated with more complex behaviors.
Histological analysis indicated the presence of mouse IgG only in Acknowledgments
the tau mice vaccinated against tau. This suggested that these anti-
bodies were most likely retained by the CNS in tau deposits that de- This work was supported by funding from NIH (R01-NS050895, R01-
veloped in these mice. AG020241, U01-AG060965, R21-NS101504, R01-AG061895, R01-
This could explain the reduction of phopho-tau epitopes at pSer396 AG055524, R01-NS076308) and Byrd Alzheimer's Institute. Development
observed in vaccinated mice. There was no effect on Gallyas stained tau of AdvaxCpG adjuvant was supported by funding from NIAID/NIH
deposits or brain atrophy with the tau epitope vaccine. We also observed (Contract No. HHS-N272201400053C, HHS-N272200800039C and U01-
reductions in pSer396-tau on western blots from dissected brain regions AI061142). The content is solely the responsibility of the authors and does
and with ELISA measurements. Thus, by several measures, vaccination not necessarily represent the official views of the NIH.
against the AD domain of tau caused modest reduction in phospho-tau.
The Tg4510 mouse is a very aggressive model and manipulations References
aimed at rescuing the phenotype, short of turning off the transgene
(Santacruz et al., 2005), have failed. In prior work with a tau oligomer Agadjanyan, M.G., Petrovsky, N., Ghochikyan, A., 2015. A fresh perspective from im-
antibody, we found little impact on the tau phenotype in this mouse munologists and vaccine researchers: active vaccination strategies to prevent and
reverse Alzheimer’s disease. Alzheimers Dement. 11 (10), 1246–1259.
(Schroeder et al., 2017). To our knowledge, only one immunotherapy has Agadjanyan, M.G., Zagorski, K., Petrushina, I., Davtyan, H., Kazarian, K., Antonenko, M.,
resulted in reduction of tau pathology in this mouse model. Sankar- Davis, J., Bon, C., Blurton-Jones, M., Cribbs, D.H., Ghochikyan, A., 2017. Humanized
anarayan et al. (Sankaranarayanan et al., 2015) observed 20% reductions monoclonal antibody armanezumab specific to N-terminus of pathological tau:
characterization and therapeutic potency. Mol. Neurodegener. 12 (1), 33.
in tau deposition using a monoclonal antibody. However, in the same paper Alamed, J., Wilcock, D.M., Diamond, D.M., Gordon, M.N., Morgan, D., 2006. Two-day
they reported much larger reductions in the phenotype of the P301S-tau radial-arm water maze learning and memory task; robust resolution of amyloid-re-
model PS19. The Tg4510 mouse has 13-fold over expression of tau, while lated memory deficits in transgenic mice. Nat. Protoc. 1 (4), 1671–1679.
Albert, M., Mairet-Coello, G., Danis, C., Lieger, S., Caillierez, R., Carrier, S., Skrobala, E.,
the PS19 mouse has 5-fold over expression (Roberson, 2012). Recently it
Landrieu, I., Michel, A., Schmitt, M., Citron, M., Downey, P., Courade, J.P., Buee, L.,
has been identified that some of the Tg4510 phenotype may be related to Colin, M., 2019. Prevention of tau seeding and propagation by immunotherapy with a
insertional disruption of the Fgf14 gene by the transgene (Gamache et al., central tau epitope antibody. Brain 142 (6), 1736–1750.
Anisman, H., 1975. Time-dependent variations in aversively motivated behaviors: non-
2019). Thus, this contribution to the phenotype could not be readily re-
associative effects of cholinergic and catecholaminergic activity. Psychol. Rev. 82 (5),
versed by preventing tau deposition. We conclude that the modest reduc- 359–385.
tions in the phenotype seen here may reflect the Tg4510 model and per- Asuni, A.A., Boutajangout, A., Quartermain, D., Sigurdsson, E.M., 2007. Immunotherapy
haps not the overall efficacy of the vaccine or its intended target. targeting pathological tau conformers in a tangle mouse model reduces brain pa-
thology with associated functional improvements. J. Neurosci. 27 (34), 9115–9129.
Bachmann, M.F., Jennings, G.T., Vogel, M., 2019. A vaccine against Alzheimer’s disease:
anything left but faith? Expert. Opin. Biol. Ther. 19 (1), 73–78.
5. Conclusions Benhamron, S., Rozenstein-Tsalkovich, L., Nitzan, K., Abramsky, O., Rosenmann, H.,
2018. Phos-tau peptide immunization of amyloid-tg-mice reduced non-mutant phos-
Altogether our results show that a highly immunogenic tau vaccine tau pathology, improved cognition and reduced amyloid plaques. Exp. Neurol. 303,

11
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

48–58. Joly-Amado, A., Serraneau, K.S., Brownlow, M., Marin de Evsikova, C., Speakman, J.R.,
Boimel, M., Grigoriadis, N., Lourbopoulos, A., Haber, E., Abramsky, O., Rosenmann, H., Gordon, M.N., Morgan, D., 2016. Metabolic changes over the course of aging in a
2010. Efficacy and safety of immunization with phosphorylated tau against neuro- mouse model of tau deposition. Neurobiol. Aging 44, 62–73.
fibrillary tangles in mice. Exp. Neurol. 224 (2), 472–485. Kanaan, N.M., Morfini, G.A., LaPointe, N.E., Pigino, G.F., Patterson, K.R., Song, Y.,
Boutajangout, A., Quartermain, D., Sigurdsson, E.M., 2010. Immunotherapy Targeting Andreadis, A., Fu, Y., Brady, S.T., Binder, L.I., 2011. Pathogenic forms of tau inhibit
Pathological Tau Prevents Cognitive Decline in a New Tangle Mouse Model. 30 (49). kinesin-dependent axonal transport through a mechanism involving activation of
pp. 16559–16566. axonal phosphotransferases. J. Neurosci. 31 (27), 9858–9868.
Boutajangout, A., Ingadottir, J., Davies, P., Sigurdsson, E.M., 2011. Passive immunization Kanaan, N.M., Morfini, G., Pigino, G., LaPointe, N.E., Andreadis, A., Song, Y., Leitman, E.,
targeting pathological phospho-tau protein in a mouse model reduces functional Binder, L.I., Brady, S.T., 2012. Phosphorylation in the amino terminus of tau prevents
decline and clears tau aggregates from the brain. J. Neurochem. 118 (4), 658–667. inhibition of anterograde axonal transport. Neurobiol. Aging 33 (4) 826.e815-830.
Braak, H., Braak, E., 1991. Neuropathological stageing of Alzheimer-related changes. Kontsekova, E., Zilka, N., Kovacech, B., Skrabana, R., Novak, M., 2014. Identification of
Acta Neuropathol. 82 (4), 239–259. structural determinants on tau protein essential for its pathological function: novel
Brownlow, M.L., Joly-Amado, A., Azam, S., Elza, M., Selenica, M.L., Pappas, C., Small, B., therapeutic target for tau immunotherapy in Alzheimer’s disease. Alzheimers Res.
Engelman, R., Gordon, M.N., Morgan, D., 2014. Partial rescue of memory deficits Ther. 6 (4), 45.
induced by calorie restriction in a mouse model of tau deposition. Behav. Brain Res. Lannfelt, L., Relkin, N.R., Siemers, E.R., 2014. Amyloid-ss-directed immunotherapy for
271, 79–88. Alzheimer’s disease. J. Intern. Med. 275 (3), 284–295.
Castillo-Carranza, D.L., Gerson, J.E., Sengupta, U., Guerrero-Munoz, M.J., Lasagna- LaPointe, N.E., Morfini, G., Pigino, G., Gaisina, I.N., Kozikowski, A.P., Binder, L.I., Brady,
Reeves, C.A., Kayed, R., 2014. Specific targeting of tau oligomers in Htau mice S.T., 2009. The amino terminus of tau inhibits kinesin-dependent axonal transport:
prevents cognitive impairment and tau toxicity following injection with brain-de- implications for filament toxicity. J. Neurosci. Res. 87 (2), 440–451.
rived tau oligomeric seeds. J. Alzheimers Dis. 40 (Suppl. 1), S97–s111. Lobello, K., Ryan, J.M., Liu, E., Rippon, G., Black, R., 2012. Targeting Beta amyloid: a
Chai, X., Wu, S., Murray, T.K., Kinley, R., Cella, C.V., Sims, H., Buckner, N., Hanmer, J., clinical review of immunotherapeutic approaches in Alzheimer’s disease. Int. J.
Davies, P., O’Neill, M.J., Hutton, M.L., Citron, M., 2011. Passive immunization with Alzheimers Dis. 2012, 628070.
anti-Tau antibodies in two transgenic models: reduction of Tau pathology and delay Morgan, D., Gordon, M.N., Tan, J., Wilcock, D., Rojiani, A.M., 2005. Dynamic complexity
of disease progression. J. Biol. Chem. 286 (39), 34457–34467. of the microglial activation response in transgenic models of amyloid deposition:
Cribbs, D.H., Ghochikyan, A., Vasilevko, V., Tran, M., Petrushina, I., Sadzikava, N., implications for Alzheimer therapeutics. J. Neuropathol. Exp. Neurol. 64 (9),
Babikyan, D., Kesslak, P., Kieber-Emmons, T., Cotman, C.W., Agadjanyan, M.G., 743–753.
2003. Adjuvant-dependent modulation of Th1 and Th2 responses to immunization Novak, P., Kontsekova, E., Zilka, N., Novak, M., 2018. Ten years of Tau-targeted im-
with beta-amyloid. Int. Immunol. 15 (4), 505–514. munotherapy: the path walked and the roads ahead. Front. Neurosci. 12, 798.
d’Abramo, C., Acker, C.M., Jimenez, H.T., Davies, P., 2013. Tau passive immunotherapy Novak, P., Zilka, N., Zilkova, M., Kovacech, B., Skrabana, R., Ondrus, M., Fialova, L.,
in mutant P301L mice: antibody affinity versus specificity. PLoS One 8 (4), e62402. Kontsekova, E., Otto, M., Novak, M., 2019. AADvac1, an active immunotherapy for
Dai, C.L., Tung, Y.C., Liu, F., Gong, C.X., Iqbal, K., 2017. Tau passive immunization in- Alzheimer’s disease and non Alzheimer tauopathies: an overview of preclinical and
hibits not only tau but also Abeta pathology. Alzheimers Res. Ther. 9 (1), 1. clinical development. J. Prev. Alzheimers Dis. 6 (1), 63–69.
Dai, C.L., Hu, W., Tung, Y.C., Liu, F., Gong, C.X., Iqbal, K., 2018. Tau passive im- Orgogozo, J.M., Gilman, S., Dartigues, J.F., Laurent, B., Puel, M., Kirby, L.C., Jouanny, P.,
munization blocks seeding and spread of Alzheimer hyperphosphorylated Tau-in- Dubois, B., Eisner, L., Flitman, S., Michel, B.F., Boada, M., Frank, A., Hock, C., 2003.
duced pathology in 3 x Tg-AD mice. Alzheimers Res. Ther. 10 (1), 13. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 im-
Davtyan, H., Ghochikyan, A., Petrushina, I., Hovakimyan, A., Davtyan, A., Poghosyan, A., munization. Neurology 61 (1), 46–54.
Marleau, A.M., Movsesyan, N., Kiyatkin, A., Rasool, S., Larsen, A.K., Madsen, P.J., Panza, F., Lozupone, M., Seripa, D., Imbimbo, B.P., 2019. Amyloid-beta immunotherapy
Wegener, K.M., Ditlevsen, D.K., Cribbs, D.H., Pedersen, L.O., Agadjanyan, M.G., for alzheimer disease: is it now a long shot? Ann. Neurol. 85 (3), 303–315.
2013. Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine Petrushina, I., Ghochikyan, A., Mktrichyan, M., Mamikonyan, G., Movsesyan, N.,
(Lu AF20513) for Alzheimer’s disease: prelude to a clinical trial. J. Neurosci. 33 (11), Davtyan, H., Patel, A., Head, E., Cribbs, D.H., Agadjanyan, M.G., 2007. Alzheimer’s
4923–4934. disease peptide epitope vaccine reduces insoluble but not soluble/oligomeric Abeta
Davtyan, H., Ghochikyan, A., Petrushina, I., Hovakimyan, A., Davtyan, A., Cribbs, D.H., species in amyloid precursor protein transgenic mice. J. Neurosci. 27 (46),
Agadjanyan, M.G., 2014a. The MultiTEP platform-based Alzheimer’s disease epitope 12721–12731.
vaccine activates a broad repertoire of T helper cells in nonhuman primates. Rajamohamedsait, H., Rasool, S., Rajamohamedsait, W., Lin, Y., Sigurdsson, E.M., 2017.
Alzheimers Dement. 10 (3), 271–283. Prophylactic active tau immunization leads to sustained reduction in both tau and
Davtyan, H., Petrushina, I., Ghochikyan, A., 2014b. Immunotherapy for Alzheimer’s amyloid-beta pathologies in 3xTg mice. Sci. Rep. 7 (1), 17034.
disease: DNA- and protein-based epitope vaccines. Methods Mol. Biol. 1143, Roberson, E.D., 2012. Mouse models of frontotemporal dementia. Ann. Neurol. 72 (6),
259–281. 837–849.
Davtyan, H., Zagorski, K., Rajapaksha, H., Hovakimyan, A., Davtyan, A., Petrushina, I., Rosenmann, H., Grigoriadis, N., Karussis, D., Boimel, M., Touloumi, O., Ovadia, H.,
Kazarian, K., Cribbs, D.H., Petrovsky, N., Agadjanyan, M.G., Ghochikyan, A., 2016. Abramsky, O., 2006. Tauopathy-like abnormalities and neurologic deficits in mice
Alzheimer’s disease Advax(CpG)- adjuvanted MultiTEP-based dual and single vac- immunized with neuronal tau protein. Arch. Neurol. 63 (10), 1459–1467.
cines induce high-titer antibodies against various forms of tau and Abeta pathological Rozenstein-Tsalkovich, L., Grigoriadis, N., Lourbopoulos, A., Nousiopoulou, E., Kassis, I.,
molecules. Sci. Rep. 6, 28912. Abramsky, O., Karussis, D., Rosenmann, H., 2013. Repeated immunization of mice
DeKosky, S.T., Scheff, S.W., 1990. Synapse loss in frontal cortex biopsies in Alzheimer’s with phosphorylated-tau peptides causes neuroinflammation. Exp. Neurol. 248,
disease: correlation with cognitive severity. Ann. Neurol. 27 (5), 457–464. 451–456.
Dickey, C., Kraft, C., Jinwal, U., Koren, J., Johnson, A., Anderson, L., Lebson, L., Lee, D., Sankaranarayanan, S., Barten, D.M., Vana, L., Devidze, N., Yang, L., Cadelina, G., Hoque,
Dickson, D., de Silva, R., Binder, L.I., Morgan, D., Lewis, J., 2009. Aging analysis N., DeCarr, L., Keenan, S., Lin, A., Cao, Y., Snyder, B., Zhang, B., Nitla, M., Hirschfeld,
reveals slowed tau turnover and enhanced stress response in a mouse model of G., Barrezueta, N., Polson, C., Wes, P., Rangan, V.S., Cacace, A., Albright, C.F.,
tauopathy. Am. J. Pathol. 174 (1), 228–238. Meredith Jr., J., Trojanowski, J.Q., Lee, V.M., Brunden, K.R., Ahlijanian, M., 2015.
Dujardin, S., Colin, M., Buee, L., 2015. Invited review: animal models of tauopathies and Passive immunization with phospho-tau antibodies reduces tau pathology and
their implications for research/translation into the clinic. Neuropathol. Appl. functional deficits in two distinct mouse tauopathy models. PLoS One 10 (5),
Neurobiol. 41 (1), 59–80. e0125614.
Duyckaerts, C., Bennecib, M., Grignon, Y., Uchihara, T., He, Y., Piette, F., Hauw, J.J., Santacruz, K., Lewis, J., Spires, T., Paulson, J., Kotilinek, L., Ingelsson, M., Guimaraes, A.,
1997. Modeling the relation between neurofibrillary tangles and intellectual status. DeTure, M., Ramsden, M., McGowan, E., Forster, C., Yue, M., Orne, J., Janus, C.,
Neurobiol. Aging 18 (3), 267–273. Mariash, A., Kuskowski, M., Hyman, B., Hutton, M., Ashe, K.H., 2005. Tau suppres-
Ferrer, I., Boada Rovira, M., Sanchez Guerra, M.L., Rey, M.J., Costa-Jussa, F., 2004. sion in a neurodegenerative mouse model improves memory function. Science 309
Neuropathology and pathogenesis of encephalitis following amyloid-beta im- (5733), 476–481.
munization in Alzheimer’s disease. Brain Pathol. 14 (1), 11–20. Schilling, S., Rahfeld, J.U., Lues, I., Lemere, C.A., 2018. Passive Abeta immunotherapy:
Fox, N.C., Scahill, R.I., Crum, W.R., Rossor, M.N., 1999. Correlation between rates of current achievements and future perspectives. Molecules 23 (5).
brain atrophy and cognitive decline in AD. Neurology 52 (8), 1687–1689. Schindowski, K., Bretteville, A., Leroy, K., Begard, S., Brion, J.P., Hamdane, M., Buee, L.,
Gamache, J., Benzow, K., Forster, C., Kemper, L., Hlynialuk, C., Furrow, E., Ashe, K.H., 2006. Alzheimer’s disease-like tau neuropathology leads to memory deficits and loss
Koob, M.D., 2019. Factors other than hTau overexpression that contribute to tauo- of functional synapses in a novel mutated tau transgenic mouse without any motor
pathy-like phenotype in rTg4510 mice. Nat. Commun. 10 (1), 2479. deficits. Am. J. Pathol. 169 (2), 599–616.
Godyn, J., Jonczyk, J., Panek, D., Malawska, B., 2016. Therapeutic strategies for Schroeder, S.K., Joly-Amado, A., Gordon, M.N., Morgan, D., 2016. Tau-directed im-
Alzheimer’s disease in clinical trials. Pharmacol. Rep. 68 (1), 127–138. munotherapy: a promising strategy for treating Alzheimer’s disease and other tauo-
Gordon, M.N., Holcomb, L.A., Jantzen, P.T., DiCarlo, G., Wilcock, D., Boyett, K.L., pathies. J. Neuroimmune Pharmacol. 11 (1), 9–25.
Connor, K., Melachrino, J.O., O’Callaghan, J.P., Morgan, D., 2002. Time course of the Schroeder, S., Joly-Amado, A., Soliman, A., Sengupta, U., Kayed, R., Gordon, M.N.,
development of Alzheimer-like pathology in the doubly transgenic PS1+APP mouse. Morgan, D., 2017. Oligomeric tau-targeted immunotherapy in Tg4510 mice.
Exp. Neurol. 173, 183–195. Alzheimers Res. Ther. 9 (1), 46.
Jack Jr., C.R., Lowe, V.J., Weigand, S.D., Wiste, H.J., Senjem, M.L., Knopman, D.S., Selenica, M.-L., Davtyan, H., Housley, S.B., Blair, L., Gillies, A., Nordhues, B.A., Zhang, B.,
Shiung, M.M., Gunter, J.L., Boeve, B.F., Kemp, B.J., Weiner, M., Petersen, R.C., Liu, J., Gestwicki, J.E., Lee, D., Gordon, M.N., Morgan, D., Dickey, C., 2014. Epitope
Alzheimer’s Disease Neuroimaging, I., 2009. Serial PIB and MRI in normal, mild analysis following active immunization with tau proteins reveals immunogens im-
cognitive impairment and Alzheimer’s disease: implications for sequence of patho- plicated in tau pathogenesis.
logical events in Alzheimer’s disease. Brain 132 (Pt 5), 1355–1365. Selkoe, D.J., 2018. Light at the end of the Amyloid TunnelPublished as part of the bio-
Jeganathan, S., von Bergen, M., Brutlach, H., Steinhoff, H.J., Mandelkow, E., 2006. Global chemistry series “biochemistry to bedside”. Biochemistry 57 (41), 5921–5922.
hairpin folding of tau in solution. Biochemistry 45 (7), 2283–2293. Terry, R.D., Masliah, E., Salmon, D.P., Butters, N., DeTeresa, R., Hill, R., Hansen, L.A.,

12
A. Joly-Amado, et al. Neurobiology of Disease 134 (2020) 104636

Katzman, R., 1991. Physical basis of cognitive alterations in Alzheimer’s disease: A., Barrezueta, N.X., Polson, C., Bourin, C., Flynn, M.E., Keenan, S., Lidge, R.,
synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30 (4), Meredith, J., Natale, J., Sankaranarayanan, S., Cadelina, G.W., Albright, C.F., Cacace,
572–580. A.M., 2014. Tau overexpression impacts a neuroinflammation gene expression net-
Troquier, L., Caillierez, R., Burnouf, S., Fernandez-Gomez, F.J., Grosjean, M.E., Zommer, work perturbed in Alzheimer’s disease. PLoS One 9 (8), e106050.
N., Sergeant, N., Schraen-Maschke, S., Blum, D., Buee, L., 2012. Targeting phospho- Winblad, B., Graf, A., Riviere, M.E., Andreasen, N., Ryan, J.M., 2014. Active im-
Ser422 by active tau immunotherapy in the THYTau22 mouse model: a suitable munotherapy options for Alzheimer’s disease. Alzheimers Res. Ther. 6 (1), 7.
therapeutic approach. Curr. Alzheimer Res. 9 (4), 397–405. Wisniewski, T., Boutajangout, A., 2010. Immunotherapeutic approaches for Alzheimer’s
Ward, S.M., Himmelstein, D.S., Lancia, J.K., Binder, L.I., 2012. Tau oligomers and tau disease in transgenic mouse models. Brain Struct. Funct. 214 (2–3), 201–218.
toxicity in neurodegenerative disease. Biochem. Soc. Trans. 40 (4), 667–671. Wisniewski, T., Goni, F., 2014. Immunotherapy for Alzheimer’s disease. Biochem.
Wes, P.D., Easton, A., Corradi, J., Barten, D.M., Devidze, N., DeCarr, L.B., Truong, A., He, Pharmacol. 88 (4), 499–507.

13

You might also like