You are on page 1of 10

pubs.acs.

org/bc Article

Synthesis and Biological Evaluation of Peptide-Adjuvant Conjugate


Vaccines with Increasing Antigen Content
Taylor R. Cooney, Kathryn Farrand, Sarah L. Draper, Regan J. Anderson, Phillip M. Rendle,
Ian F. Hermans, Benjamin J. Compton,* and Gavin F. Painter*
Cite This: Bioconjugate Chem. 2023, 34, 799−808 Read Online

ACCESS Metrics & More Article Recommendations *


sı Supporting Information
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via PEOPLES FRIENDSHIP UNIV OF RUSSIA on June 12, 2023 at 18:45:23 (UTC).

ABSTRACT: Synthetic vaccines that induce T cell responses to


peptide epitopes are a promising immunotherapy for both
communicable and noncommunicable diseases. Stimulating strong
and sustained T cell responses requires antigen delivery to
appropriately activated antigen presenting cells (APCs). One way
this can be accomplished is by chemically conjugating immunogenic
peptide epitopes with α-galactosylceramide (α-GalCer), a glycolipid
that acts as an immune adjuvant by inducing stimulatory interactions
between APCs and type I natural killer T (NKT) cells. Here we
investigate whether increasing the ratio of antigen:adjuvant improves
antigen-specific T cell responses. A series of conjugate vaccines was
prepared in which one, two, four, or eight copies of an immunogenic
peptide were covalently attached to a modified form of α-GalCer via
the poly(ethoxyethylglycinamide) dendron scaffold. Initial attempts
to synthesize these multivalent conjugate vaccines involved attaching the bicyclo[6.1.0]non-4-yne (BCN) group to the adjuvant-
dendron structure followed by strain-promoted azide−alkyne cycloaddition of the peptide. Although this approach was successful for
preparing vaccines with either one or two peptide copies, the synthesis of vaccines requiring attachment of four or eight BCN groups
suffered from low yields due to cyclooctyne degradation. Instead, conjugate vaccines containing up to eight peptide copies were
readily achieved through oxime ligation with adjuvant-dendron constructs decorated with the 8-oxo-nonanoyl group. When
evaluating T cell responses to vaccination in mice, we confirmed a significant advantage to conjugation over admixes of peptide and
α-GalCer, regardless of the peptide to adjuvant ratio, but there was no advantage to increasing the number of peptides attached.
However, it was notable that the higher ratio conjugate vaccines required lower levels of NKT cell activation to be effective, which
could be a safety advantage for future vaccine candidates.

■ INTRODUCTION
Infected or malignant cells can be targeted and eliminated by
and chemical compounds that trigger APCs directly via pattern
recognition receptors such as Toll-like receptors (TLRs), C-
activated CD8+ T cells that specifically recognize peptide type lectin receptors (CLRs), and nucleotide binding
epitopes presented via major histocompatibility complex oligomerization domain (NOD)-like receptors (NLRs).3
(MHC) class I proteins on the cell surface. Simple peptide- Alternatively, APC activation can be achieved through direct
based vaccines based on these epitopes can therefore be used interaction with other immune cells, notably those capable of
to prime or restimulate CD8+ T cell responses with high producing licensing signals,4 typified by the stimulation of
specificity without concerns regarding the immunogenicity of a CD40.5,6 This includes “unconventional” T cells like type I
delivery vector. The fact that peptides can be readily natural killer T (NKT) cells, which are found in high numbers
synthesized as discrete entities on large scales using automated in lymphoid tissues and respond rapidly to stimulation with
technologies with stability and/or manufacturing advantages specific agonistic antigens.7 The glycolipid compound α-
compared to antigens based on nucleic acids or whole proteins galactosylceramide (α-GalCer) is regarded as a prototypical
adds to the appeal of this vaccine platform. However, peptide
epitopes are poorly immunogenic when administered as single Received: March 5, 2023
agents and require coadministration with an immune stimulant Revised: March 25, 2023
(adjuvant) to support acquisition and/or presentation by Published: April 6, 2023
appropriately activated antigen presenting cells (APCs) in vivo.
Strategies to achieve this include oil-in-water emulsions that
aid in delivery,1 cytokines that support the function of APCs,2

© 2023 American Chemical Society https://doi.org/10.1021/acs.bioconjchem.3c00106


799 Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

Scheme 1. Synthetic Route to the 1:1 Adjuvant:Antigen (G0) GLP Vaccine 1

agonist for NKT cells8 and has been shown to act as an vaccines capable of delivering a higher antigen density, either
immune adjuvant when introduced into vaccines in preclinical through encapsulation30,33,34 or conjugation,35,36 can lead to
studies,9−11 while a variant of this structure has been shown to stronger antigen-specific responses owing to increased
have adjuvant activity in humans.12 In order to function as an circulation time and/or acting as an antigen depot.
immune adjuvant, the NKT cell agonist must be acquired and A synthetically tractable approach to generate well-defined,
presented via the MHC class I-like molecule CD1d on APCs multipeptide conjugate vaccines is to employ a dendron
to NKT cells, which in turn rapidly releases cytokines and scaffold to link the adjuvant core and the peptide antigen.
directs molecular signals, including the provision of CD40L, Dendrons offer an organized, nanosized branched framework
that transactivate APCs. If these same APCs have acquired and that can accommodate a vast array of functional groups
presented peptide epitopes from the vaccine, peptide-specific T attached to the termini, including those suitable for attaching
cell responses can be initiated or boosted.8,9,13,14 peptides. Commonly used peptide-presenting dendritic
The capacity of α-GalCer to adjuvant synthetic peptide- structures include branched lysine, 37 polyamidoamine
based vaccines in various disease models including cancer15−17 (PAMAM),38 and polyphenylene (PPDs)39 repeating units.
and infectious disease18−21 has been enhanced by strategies More recently, poly(ethoxyethylglycinamide) (PEE-G) den-
that encourage presentation of α-GalCer and peptide by the dron has been investigated for multipeptide display.40 Due to
same APC.20,22−24 This includes encapsulation into a variety of its high stability, high aqueous solubility, low cytotoxicity,
different synthetic vaccine particles,25−30 employing filamen- chemical orthogonality, and low immunogenicity,41 PEE-G is
tous bacteriophages or virus-like particles,31 and attachment to considered a particularly attractive scaffold for pharmaceutical
gold nanoparticles.32 Although these strategies to promote applications.
codelivery have proven effective in animal models, controlling To investigate whether PEE-G-based conjugate vaccines
and quantifying the ratio of adjuvant and antigen in these with increasing number of peptide epitopes (i.e., antigen load)
approaches is challenging. Self-adjuvanting α-GalCer-based attached to each α-GalCer molecule could induce stronger
glycolipid-peptide (GLP) conjugate vaccines overcome this peptide-specific T cell responses than those of equal molarity,
limitation by chemically linking the antigen and adjuvant, thus vaccines with adjuvant:peptide ratios of 1:1, 1:2, 1:4, and 1:8
providing synthetically accessible, chemically defined con- (generations (G)0 to G3, respectively) were synthesized. Here,
structs that are highly manufacturable with a prolonged shelf we adapted our previously reported approach17 that utilizes a
life as they can be stored indefinitely as lyophilized powders or modified α-GalCer compound in the vaccine design. Previous
frozen DMSO solutions. Although proven to generate strong T research has shown that GLP vaccines comprising a
cell responses in preclinical studies, current α-GalCer-peptide “rearranged” α-GalCer adjuvant, formed by the migration of
conjugate vaccines have been restricted to an equimolar ratio the cerotic acid moiety, conjugated to peptide antigen via the
of antigen and adjuvant, which may be suboptimal considering protease sensitive para-aminobenzyl-dipeptide linker are able
peptide-based therapeutics have a short circulation half-life due to induce strong antigen-specific T cell responses in vivo.42
to proteolytic degradation. Indeed, studies have shown that The activity of these vaccines is contingent on intracellular
800 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

Scheme 2. Synthesis of the G1 GLP Vaccine 14 Comprising an Adjuvant:Antigen Ratio of 1:2

Scheme 3. Synthesis of the tetra-BCN Dendron Core and Its Attempted Reaction with “Rearranged” α-GalCer Derivative

linker degradation leading to the close temporal release of both stable in circulation but are readily cleaved by proteolytic
the adjuvant and peptide antigen inside the same APC. In the cathepsin enzymes.43−45 Also, compared to citrulline, alanine
work presented here, the same α-GalCer-based vaccine design has improved solubility in organic solvents, thus offering a
was employed, except the citrulline residue in the immolative more facile synthesis of the α-GalCer-linker vaccine
dipeptide linker was substituted for alanine. This change was component. The peptide chosen for this study is an MHC
based on previous studies that have shown Val-Ala linkers are class I-binding peptide from human papillomavirus (HPV) E7
801 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

protein (E749−57; RAHYNIVTF), which has been defined as a 18, was N-capped with the exo-BCN group to give tetra-
CD8+ T cell epitope in C57BL/6 mice restricted by H2-Db. To cyclooctyne 19. Attempts to react 19 with the rearranged form
ensure facile release of the attached peptide within APCs, the of α-GalCer (5) employing HBTU, HOBt, and DIPEA,
known proteolytic cleavage site -FFRK-46 was incorporated at conditions that were successful generating the G0 (1) and G1
the peptide N-terminus alongside either the 5-azido-pentanoyl (14) vaccines, failed to give an appreciable amount of the
or aminooxy acetamido (AoA) groups for strain-promoted desired G2 tetra-cyclooctyne product 20.
azide−alkyne cycloaddition (SPAAC) or oxime ligation, To investigate this further, the reaction between α-GalCer
respectively. Multimerising the E749−57 peptide antigen on amine 5 and tetracyclooctyne 19 was monitored by LCMS.
the “migrated” α-GalCer adjuvant core was achieved via the After 2 h, approximately 55% of starting material 19 remained,
PEE-G dendron core, terminally modified with either a ring- while the appearance of a new peak, accounting for 45% of the
strained alkyne or pendant ketone that would permit reaction mixture, with a retention time and m/z of 2574.7
chemoselective ligation with an appropriately functionalized matching that expected for 20 was observed. Allowing the
peptide. reaction to progress at ambient temperature revealed that only
The work reported here describes the development and in 20% of purported 20 was present after 14 h, which diminished
vivo evaluation of such a dendron-based vaccine platform with to <15% after 24 h. The appearance of a series of new peaks in
controlled conjugation of up to eight peptide copies onto α- the chromatogram were observed (after 24 h) corresponding
GalCer. to masses 16, 32, 48, and 64 Da higher than that expected for

■ RESULTS AND DISCUSSION


Synthesis of Dendritic Vaccines Using SPAAC
G2 tetra-cyclooctyne product 20 suggested that these coupling
conditions were promoting oxidative degradation of the
dendron terminal BCN groups. Due to the nature and varying
Ligation. The synthesis of the G0 vaccine (1) comprising degrees of degradation, individual compounds were not able to
an equimolar ratio of antigen to adjuvant commenced from the be isolated from this reaction mixture.
addition of the para-nitrophenol (pNP)-activated dipeptide Degradation of the BCN unit was first reported by Gibson et
linker (2)44 and “migrated” α-GalCer (3)22 to give carbamate al.47 who observed the acidic hydration of the alkyne upon
4 (Scheme 1). Removal of the valine Fmoc protecting group addition of trichloroacetic acid to an BCN functionalized
and subsequent capping of the liberated amino group (in 5) oligonucleotide. Feng et al.48 observed a similar occurrence
with commercially available pNP-exo-BCN (6) gave cyclo- whereby BCN-containing carbamates formed cyclooctanones
octyne 7 in excellent yield. Stirring 7 with 5-azidopentanoyl- under acidic conditions. Because acidic hydration would
FFRKRAHYNIVTF (8) in DMSO at ambient temperature produce byproducts of 18 Da higher and not 16 Da as we
afforded the GLP conjugate vaccine 1 in 85% yield and 99% observed, we synthesized the model BCN propylcarbamate
purity after purification by preparative HPLC. (21) (Scheme 4) to further investigate our observations that
Increasing the antigenic load from one peptide (as in the G0
vaccine, 1) up to eight copies (G3 vaccine) was considered Scheme 4. Prolonged Exposure of the BCN
best achieved by “growing” the PEE-G dendron core through Propylcarbamate 21 to HBTU/HOBt Coupling Reagents
simple amide bond formation.41 Through this approach, it was Leads to Enone Formation
envisaged that an appropriately functionalized dendron scaffold
could be attached to the adjuvant core which, upon
chemoselective conjugation of antigen, would afford the
desired multigeneration GLP vaccines. Toward this, dendron
synthesis commenced from the commercially available
dialkylated glycine derivative (9) which was hydrogenolyzed
to liberate the carboxylic acid 1041 followed by acid-mediated
removal of the Boc protecting groups to afford bis-amine 11
(Scheme 2). Reacting 11 with pNP-exo-BCN 6 under basic
conditions gave bis-cyclooctyne 12 in 77% yield which was
subsequently coupled with α-GalCer amine 5 in the presence similar, but likely different, degradation products can form
of HBTU, HOBt, and DIPEA to afford α-GalCer bis- using HBTU and HOBt with DIPEA. Reacting pNP-exo-BCN
cyclooctyne 13 in good yield. Subjecting 13 to 3 equiv of 6 with propylamine afforded carbamate 21 which was
azido-peptide 8 in DMSO at ambient temperature for 18 h subjected to the same amide coupling conditions that lead to
gave, after purification by preparative HPLC, the G1 GLP the degradation of 20. Analyzing this reaction by HPLC
vaccine (14) bearing two peptide copies. Although 14 was revealed the formation of a major product with a mass 16 Da
obtained in an unusually low yield of 20% compared to that greater than 21 which, when isolated, was shown to have
typically reported for similar GLP conjugates (which range undergone degradation of the alkyne to an enone system (as in
from 40% to 60% with purities >95% by HPLC), a sufficient 22). A potential mechanism for this observation is proposed to
quantity of this vaccine was obtained for immunological form via HOBt which, by virtue of its N−O bond, may act as
testing. an oxidant (Supplementary Scheme 1). While the BCN moiety
Synthesis of the G2 GLP vaccine containing four peptide can survive relatively brief exposure to these conditions, as
copies was attempted using a similar to that which gave G1 exemplified for the preparation of α-GalCer bis-cyclooctyne
GLP vaccine 14. Doubling the antigenic load from two peptide 13, when couplings are slow or multiple copies of BCN are
copies (as in 14) to four was considered best achieved by present in the molecule, degradation can become a significant
doubling the branching within the dendron core (Scheme 3). problem, as we observed. For these reasons we investigated
The G2 dendron scaffold 17 was prepared as reported41 and, oxime ligation as an alternative chemoselective conjugation
upon removal of the Boc protecting groups to give tetra-amine strategy.
802 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

Scheme 5. Synthetic Route to the 1:1 Adjuvant:Antigen (G0) GLP Vaccine 26

Scheme 6. Synthesis of the G1−G3 GLP Vaccines

803 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

Figure 1. Evaluation of GLP vaccines with increasing antigen dose compared to injected admixes. Groups of C57BL/6J mice (n = 5) were
intravenously administered the indicated GLP vaccines with molar dose of conjugated NKT cell agonist at 0.5 nmol; G0 (26, 1:1), G2 (37, 1:4)
and G3 (38, 1:8). Comparator groups of mice were administered the equivalent molar ratios of unconjugated α-GalCer (α-GC) and E749−57
peptide (1:1, 1:4, 1:8), with an additional high ratio mix (1:64); control animals received PBS vehicle. (A) Assessment of cytokines released into
serum in response to treatment at indicated times, with heatmaps showing concentration normalized to sum of all values for each cytokine. (B)
Serum concentrations of indicated cytokines. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. One-way ANOVA with Dunnett’s multiple
comparisons test. (C) Flow cytometric analysis of NKT cells (CD1d tetramer+ cells) in the spleen on day 7, identified by the gating strategy in
Supplementary Figure 1, showing frequency, total number in spleen, percentage of NK1.1-negative cells (as an indication of activation), and
expression of CD69 and PD-1. One-way ANOVA with Tukey’s multiple comparisons. (D) Analysis of splenic E749−57-specific T cell response in
spleen by flow cytometry (left) using gating strategy in Supplementary Figure 2 using H2-Db/peptide pentamers to detect antigen-specific cells or
by ELISpot (right) after restimulation of splenocytes with peptide ex vivo. One-way ANOVA with Tukey’s multiple comparisons.

804 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

Revised Synthesis of Dendritic Vaccines Using Oxime triggered after NKT cell activation. This was expected to
Ligation. Oxime ligation between carbonyl-containing com- include (among others) early IFN-γ production by the NKT
pounds (i.e., aldehydes or ketones) and peptides functionalized cells themselves, and IL-12p70 production by transactivated
with an aminooxy group is a proven way to efficiently generate APCs, and a later additional IFN-γ burst by natural killer
glycolipid-peptide conjugate vaccines.20,21,49 Conditions pro- cells.51,52 Flow cytometry was used to examine NKT cell and
moting oxime bond formation include the use of aniline peptide-specific CD8+ T cell responses in spleens harvested 7
catalysts under mildly acidic conditions (∼pH 3−5).50 To days after vaccination. Fluorescent multimeric CD1d/glyco-
investigate this ligation approach, the corresponding G0 lipid complexes were used to identify NKT cells, while
vaccine construct was synthesized whereby α-GalCer amine multimeric H-2 Db/E749−57 complexes were used to identify
5 was reacted with the activated ester of 8-oxo-nonanoic acid peptide-specific T cells. Evaluation of T cell response was also
(23) to give α-GalCer ketone 24 (Scheme 5). Having conducted using ELISpot on splenocytes to identify IFN-γ-
previously shown that a solution of aniline and TFA in producing cells responding to in vitro restimulation with the
hexafluoroisopropanol (HFIP) can efficiently catalyze the E749−57 peptide. The experimental groups included the G0 (26,
oxime ligation of an appropriately functionalized glycolipid 1:1), G2 (37, 1:4), and G3 (38, 1:8) dendron conjugates and
and peptide,21 these reaction conditions were examined for admixtures of α-GalCer and E749−57 peptide (ratios of 1:1, 1:4,
synthesis of the G0 vaccine (Scheme 5). Starting with ketone 1:8, and 1:64). All vaccines were injected by the intravenous
24 and aminooxy acetyl (AoA)-FFRKRAHYNIVTF (25), a route in C57BL/6 mice, where the dose of NKT agonist was
series of concentrations of aniline (100, 200, and 400 mM) in held constant.
HFIP were assessed for the synthesis of the G0 1:1 conjugate Multiple cytokines were detected in serum after injection of
(26). Monitoring the reactions by LCMS showed that 100 the admixes as shown as fold changes from PBS controls in
mM aniline and 400 mM aniline reactions were complete heatmaps (Figure 1A). The changes observed for each
within 24 h, while the 200 mM aniline reaction exhibited a cytokine were reasonably consistent across the different ratios
faster reaction rate and was complete within 12 h likely owing of peptide to glycolipid, suggesting that the constant molar
to this system having a high catalyst loading while operating at dose of α-GalCer is the primary driver of the early immune
a favorable pH for oxime formation. Because the higher (400 response. Injection of the conjugates also induced cytokine
mM) concentration of aniline was not proven to be production, but fold changes were generally lower, particularly
detrimental (i.e., there was no observed formation of side in response to the higher generation dendron structures (G2,
products or degradation products in HPLC chromatogram for 37 and G3, 38) at 18 h. Statistical analysis of the key cytokines
this reaction), the reaction of glycolipid 24 and peptide 25 was
IFN-γ and IL-12p70 highlights this trend, with the admixes
repeated using 400 mM aniline + 130 mM TFA (pH ∼4) and
inducing significantly enhanced IFN-γ levels at 6 and 18 h
was shown by LCMS to form vaccine 26 cleanly within 12 h.
compared to PBS-injected controls and enhanced IL-12p70
Accordingly, this reaction system was selected for the synthesis
levels at 6 h, but responses to the higher generation conjugates
of the larger G1−3 conjugate vaccines.
failing to reach these same levels (Figure 1B). This likely
The G1, G2, and G3 prevaccines (27, 28, and 29,
reflects lower overall levels of NKT cell stimulation, as both the
respectively) were prepared using a common strategy. Reacting
acid 23 with either bis-amine 11 or tetra-amine 18 in DMF percentages and numbers of NKT cells that accumulated in the
with Hünig’s base afforded the G1 (30) and G2 (31) dendrons spleens of animals injected with the larger conjugates were
in good yield (Scheme 6). Coupling bis-amine 15 and tetra- lower than in animals that received the admixes, suggesting less
Boc acid 17 with HBTU and HOBt in DMF with Hünig’s base vaccine-induced NKT cell proliferation over the 7 days of the
gave the N-Boc, O-Bn protected G3 dendron core motif 32. experiment (Figure 1C, left panels). The NKT cells detected in
Hydrogenolysis (to 33)41 followed by acid-mediated N-Boc all test groups on day 7 were largely NK1.1 negative, which is a
deprotection gave octaamine 34 which was in turn coupled typical outcome after α-GalCer administration, and CD69 was
with 23 to afford the G3 dendron 35. Subsequent coupling of downregulated compared to PBS (Figure 1C, right panels).
glycolipid 5 to each G1−3 dendron core (30, 31, and 35, Although the latter is an activation marker, its expression is
respectively) gave the ketone-containing adjuvant-dendron known to peak earlier, within days of a strong stimulus, and
constructs ready for oxime ligation. The G1 and G2 vaccines then decline for several days. The expression of PD-1 was also
were prepared by treating the ketone precursor (30 and 31, increased on NKT cells in response to activation in all test
respectively) with 6 equiv of AoA-peptide 25 in 400 mM groups, but the levels were consistently lower when the
aniline + 130 mM TFA at 40 °C overnight and purified by conjugate vaccines were used (Figure 1C, bottom left panel),
preparative HPLC to give the G1 (36) and G2 (37) vaccines consistent with lower level of NKT cell activation.
in 50% and 45% yields, respectively. Employing these same When assessed by flow cytometry, peptide-specific T cell
conditions for the G3 prevaccine (29) led to incomplete responses to the admixes were limited, reaching statistical
coupling even when 12 equiv of 25 per ketone was used. The significance only over PBS-injected controls when injected at a
undercapped vaccines bearing six and seven peptides were ratio of 1:64 (Figure 1D). Analysis by ELISpot was more
separated from the desired eight peptide product by careful sensitive, with responses to all of the admixes increasing over
preparative HPLC chromatography, affording the G3 vaccine PBS controls. There were also increased T cell responses
(38) in 50% yield. associated with a higher peptide dose. However, this dose
Immunological Evaluation of Multipeptide Conju- effect was weak, with 64-fold more peptides only achieving a
gate Vaccines. With the series of conjugate vaccines in hand, ∼2-fold increase in T cell response. Conjugation significantly
we investigated their ability to activate NKT cells and induce improved T cell responses compared to equivalent admix
HPV-specific T cell responses in vivo. The experimental setup doses, highlighting a role for improving codelivery of antigen
included taking blood at 6 and 18 h to assess cytokine levels in and agonist in vivo. However, the impact of increasing peptide
serum, which typically reflect the cascade of cellular activities content was again marginal, with no consistent significant
805 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

improvement in T cell response between G0 (26) and the Sarah L. Draper − Ferrier Research Institute, Victoria
larger structures. University of Wellington, Lower Hutt 5010, New Zealand;
Our results therefore do not provide clear evidence that Maurice Wilkins Centre for Molecular Biodiscovery,
conjugating more peptide to an NKT cell agonist improves T Auckland 1010, New Zealand; orcid.org/0000-0001-
cell responses, but the associated reduction in NKT cell 8330-121X
activity observed with the higher generation structures does Regan J. Anderson − Ferrier Research Institute, Victoria
confound interpretation. An alternative delivery method such University of Wellington, Lower Hutt 5010, New Zealand;
as formulation of the individual components in particles may orcid.org/0000-0003-3467-9202
therefore be better suited for investigating the effect of Phillip M. Rendle − Ferrier Research Institute, Victoria
increased peptide:agonist ratios. The lower levels of NKT cell University of Wellington, Lower Hutt 5010, New Zealand
stimulation observed with G2 (37) and G3 (38) suggest that Ian F. Hermans − Maurice Wilkins Centre for Molecular
the higher order structures limit availability of the NKT cell Biodiscovery, Auckland 1010, New Zealand; Malaghan
agonist. It is possible that this is actually a benefit to these Institute of Medical Research, Wellington 6012, New Zealand
structures, as the corresponding reduction in NKT cell-driven Complete contact information is available at:
cytokine production is likely to be a safety advantage. The fact https://pubs.acs.org/10.1021/acs.bioconjchem.3c00106
that high levels of antigen-specific T cell priming were achieved
with less NKT cell activation may be due to more efficient Author Contributions
presentation of the peptide in activated APCs, or that higher
T.R.C. was primarily responsible for the chemical synthesis
levels of NKT cell activation are simply not required, especially
with contributions from B.J.C. and S.L.D. under the guidance
when increased amounts of peptide are provided. To further
of G.F.P. and P.M.R. T.R.C., I.F.H., and G.F.P. designed the in
investigate the implications of this, a series of studies probing
vivo studies, which were carried out and interpreted by K.J.F.
rates and efficiency of release rates of the glycolipid and
and I.F.H. B.J.C. and I.F.H. drafted the manuscript with
peptide components would be required.
editorial contributions from all authors. All authors have given

■ CONCLUSION
Our results support the concept of conjugating peptides to
approval to the final version of the manuscript.
Notes
NKT cell agonists to improve T cell responses and highlight a The authors declare no competing financial interest.
novel design for increasing the ratio of adjuvant to peptide
antigen. While increasing the amount of attached peptide does
not significantly improve the size of the T cell response, lower
■ ACKNOWLEDGMENTS
This work was funded by New Zealand Ministry of Business
levels of NKT cell activation are required for these vaccines to Innovation and Employment (RTVU1603), IROF to the
be effective, which could be a safety benefit. Malaghan Institute, and a Maurice Wilkins Centre PhD


*
ASSOCIATED CONTENT
sı Supporting Information
scholarship to TRC. The authors thank the Biomedical
Research Unit of the Malaghan Institute for animal husbandry.

The Supporting Information is available free of charge


athttps://pubs.acs.org/doi/10.1021/acs.bioconj-
■ REFERENCES
(1) Melssen, M. M.; Fisher, C. T.; Slingluff, C. L.; Melief, C. J. M.
chem.3c00106. Peptide emulsions in incomplete Freund’s adjuvant create effective
Full experimental procedures and compound character- nurseries promoting egress of systemic CD4(+) and CD8(+) T cells
ization data including supporting schemes, figures, and for immunotherapy of cancer. J. Immunother Cancer 2022, 10,
No. e004709.
tables (PDF) (2) Disis, M. L.; Bernhard, H.; Shiota, F. M.; Hand, S. L.; Gralow, J.

■ AUTHOR INFORMATION
Corresponding Authors
R.; Huseby, E. S.; Gillis, S.; Cheever, M. A. Granulocyte-macrophage
colony-stimulating factor: An effective adjuvant for protein and
peptide-based vaccines. Blood 1996, 88 (1), 202−210.
(3) Amarante-Mendes, G. P.; Adjemian, S.; Branco, L. M.; Zanetti,
Benjamin J. Compton − Ferrier Research Institute, Victoria L. C.; Weinlich, R.; Bortoluci, K. R. Pattern Recognition Receptors
University of Wellington, Lower Hutt 5010, New Zealand; and the Host Cell Death Molecular Machinery. Front Immunol 2018,
orcid.org/0000-0002-5245-9767; 9, 2379.
Email: Benji.Compton@vuw.ac.nz (4) Ridge, J. P.; Di Rosa, F.; Matzinger, P. A conditioned dendritic
Gavin F. Painter − Ferrier Research Institute, Victoria cell can be a temporal bridge between a CD4(+) T-helper and a T-
University of Wellington, Lower Hutt 5010, New Zealand; killer cell. Nature 1998, 393 (6684), 474−478.
Maurice Wilkins Centre for Molecular Biodiscovery, (5) Bennett, S. R. M.; Carbone, F. R.; Karamalis, F.; Flavell, R. A.;
Auckland 1010, New Zealand; orcid.org/0000-0002- Miller, J. F. A. P.; Heath, W. R. Help for cytotoxic-T-cell responses is
1310-7769; Email: Gavin.Painter@vuw.ac.nz mediated by CD40 signalling. Nature 1998, 393 (6684), 478−480.
(6) Schoenberger, S. P.; Toes, R. E. M.; van der Voort, E. I. H.;
Authors Offringa, R.; Melief, C. J. M. T-cell help for cytotoxic T lymphocytes
Taylor R. Cooney − Ferrier Research Institute, Victoria is mediated by CD40-CD40L interactions. Nature 1998, 393 (6684),
480−483.
University of Wellington, Lower Hutt 5010, New Zealand; (7) Bendelac, A.; Savage, P. B.; Teyton, L. The biology of NKT cells.
Maurice Wilkins Centre for Molecular Biodiscovery, Annu. Rev. Immunol. 2007, 25, 297−336.
Auckland 1010, New Zealand; orcid.org/0000-0003- (8) Kawano, T.; Cui, J. Q.; Koezuka, Y.; Toura, I.; Kaneko, Y.;
4461-2554 Motoki, K.; Ueno, H.; Nakagawa, R.; Sato, H.; Kondo, E.; et al.
Kathryn Farrand − Malaghan Institute of Medical Research, CD1d-restricted and TCR-mediated activation of V(alpha)14 NKT
Wellington 6012, New Zealand cells by glycosylceramides. Science 1997, 278 (5343), 1626−1629.

806 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

(9) Gonzalez-Aseguinolaza, G.; Van Kaer, L.; Bergmann, C. C.; (23) Cavallari, M.; Stallforth, P.; Kalinichenko, A.; Rathwell, D. C.
Wilson, J. M.; Schmieg, J.; Kronenberg, M.; Nakayama, K.; Taniguchi, K.; Gronewold, T. M. A.; Adibekian, A.; Mori, L.; Landmann, R.;
M.; Koezuka, Y.; Tsuji, M. Natural killer T cell ligand alpha- Seeberger, P. H.; De Libero, G. A semisynthetic carbohydrate-lipid
galactosylceramide enhances protective immunity induced by malaria vaccine that protects against S-pneumoniae in mice. Nat. Chem. Biol.
vaccines. J. Exp Med. 2002, 195 (5), 617−624. 2014, 10 (11), 950−956.
(10) Singh, S.; Nehete, P. N.; Yang, G.; He, H.; Nehete, B.; Hanley, (24) Wang, J.; Wen, Y.; Zhou, S. H.; Zhang, H. W.; Peng, X. Q.;
P. W.; Barry, M. A.; Sastry, K. J. Enhancement of Mucosal Zhang, R. Y.; Yin, X. G.; Qiu, H.; Gong, R.; Yang, G. F.; et al. Self-
Immunogenicity of Viral Vectored Vaccines by the NKT Cell Agonist Adjuvanting Lipoprotein Conjugate alphaGalCer-RBD Induces
Alpha-Galactosylceramide as Adjuvant. Vaccines 2014, 2 (4), 686− Potent Immunity against SARS-CoV-2 and its Variants of Concern.
706. J. Med. Chem. 2022, 65 (3), 2558−2570.
(11) Artiaga, B. L.; Whitener, R. L.; Staples, C. R.; Driver, J. P. (25) Ghinnagow, R.; De Meester, J.; Cruz, L. J.; Aspord, C.;
Adjuvant effects of therapeutic glycolipids administered to a cohort of Corgnac, S.; Macho-Fernandez, E.; Soulard, D.; Fontaine, J.;
NKT cell-diverse pigs. Vet Immunol Immunop 2014, 162 (1−2), 1− Chaperot, L.; Charles, J.; et al. Co-delivery of the NKT agonist
13. alpha-galactosylceramide and tumor antigens to cross-priming
(12) Tefit, J. N.; Crabe, S.; Orlandini, B.; Nell, H.; Bendelac, A.; dendritic cells breaks tolerance to self-antigens and promotes
Deng, S. L.; Savage, P. B.; Teyton, L.; Serra, V. Efficacy of ABX196, a antitumor responses. Oncoimmunology 2017, 6 (9), No. e1339855.
new NKT agonist, in prophylactic human vaccination. Vaccine 2014, (26) Huang, J.; Zhou, J.; Ghinnagow, R.; Seki, T.; Iketani, S.;
32 (46), 6138−6145. Soulard, D.; Paczkowski, P.; Tsuji, Y.; MacKay, S.; Cruz, L. J.; et al.
(13) Fujii, S.; Shimizu, K.; Smith, C.; Bonifaz, L.; Steinman, R. M. Targeted Co-delivery of Tumor Antigen and alpha-Galactosylcer-
Activation of natural killer T cells by alpha-galactosylceramide rapidly amide to CD141(+) Dendritic Cells Induces a Potent Tumor
induces the full maturation of dendritic cells in vivo and thereby acts Antigen-Specific Human CD8(+) T Cell Response in Human
as an adjuvant for combined CD4 and CD8 T cell immunity to a Immune System Mice. Front Immunol 2020, 11, 2043.
coadministered protein. J. Exp Med. 2003, 198 (2), 267−279. (27) Lam, P. Y.; Kobayashi, T.; Soon, M.; Zeng, B. J.; Dolcetti, R.;
(14) Hermans, I. F.; Silk, J. D.; Gileadi, U.; Salio, M.; Mathew, B.; Leggatt, G.; Thomas, R.; Mattarollo, S. R. NKT Cell-Driven
Ritter, G.; Schmidt, R.; Harris, A. L.; Old, L.; Cerundolo, V. NKT Enhancement of Antitumor Immunity Induced by Clec9a-Targeted
cells enhance CD4(+) and CD8(+) T cell responses to soluble Tailorable Nanoemulsion. Cancer Immunol Res. 2019, 7 (6), 952−
antigen in vivo through direct interaction with dendritic cells. J. 962.
Immunol 2003, 171 (10), 5140−5147. (28) Stolk, D. A.; de Haas, A.; Vree, J.; Duinkerken, S.; Lubbers, J.;
(15) Chen, P. G.; Hu, H. G.; Sun, Z. Y.; Li, Q. Q.; Zhang, B. D.; Wu, van de Ven, R.; Ambrosini, M.; Kalay, H.; Bruijns, S.; van der Vliet, H.
J. J.; Li, W. H.; Zhao, Y. F.; Chen, Y. X.; Li, Y. M. Fully Synthetic J.; et al. Lipo-Based Vaccines as an Approach to Target Dendritic
Invariant NKT Cell-Dependent Self-Adjuvanting Antitumor Vaccines Cells for Induction of T- and iNKT Cell Responses. Front Immunol
Eliciting Potent Immune Response in Mice. Mol. Pharmaceut 2019, 17 2020, 11, 990.
(2), 417−425. (29) Sainz, V.; Moura, L. I. F.; Peres, C.; Matos, A. I.; Viana, A. S.;
(16) Grasso, C.; Field, C. S.; Tang, C. W.; Ferguson, P. M.; Wagner, A. M.; Vela Ramirez, J. E.; Barata, T. S.; Gaspar, M.;
Compton, B. J.; Anderson, R. J.; Painter, G. F.; Weinkove, R.; Brocchini, S.; et al. alpha-Galactosylceramide and peptide-based nano-
Hermans, I. F.; Berridge, M. V. Vaccines adjuvanted with an NKT cell vaccine synergistically induced a strong tumor suppressive effect in
agonist induce effective T-cell responses in models of CNS melanoma. Acta Biomater 2018, 76, 193−207.
lymphoma. Immunotherapy 2020, 12 (6), 395−406. (30) Neumann, S.; Young, K.; Compton, B.; Anderson, R.; Painter,
(17) Anderson, R. J.; Compton, B. J.; Tang, C. W.; Authier-Hall, A.; G.; Hook, S. Synthetic TRP2 long-peptide and alpha-galactosylcer-
Hayman, C. M.; Swinerd, G. W.; Kowalczyk, R.; Harris, P.; Brimble, amide formulated into cationic liposomes elicit CD8(+) T-cell
M. A.; Larsen, D. S.; et al. NKT cell-dependent glycolipid-peptide responses and prevent tumour progression. Vaccine 2015, 33 (43),
vaccines with potent anti-tumour activity. Chem. Sci. 2015, 6 (9), 5838−5844.
5120−5127. (31) Sartorius, R.; D’Apice, L.; Barba, P.; Cipria, D.; Grauso, L.;
(18) Miller, D. S.; Finnie, J.; Bowden, T. R.; Scholz, A. C.; Oh, S.; Cutignano, A.; De Berardinis, P. Vectorized Delivery of Alpha-
Kok, T.; Burrell, C. J.; Trinidad, L.; Boyle, D. B.; Li, P. Preclinical GalactosylCeramide and Tumor Antigen on Filamentous Bacter-
efficacy studies of influenza A haemagglutinin precursor cleavage loop iophage fd Induces Protective Immunity by Enhancing Tumor-
peptides as a potential vaccine. J. Gen Virol 2011, 92, 1152−1161. Specific T Cell Response. Front Immunol 2018, 9, 1496.
(19) Anderson, R. J.; Li, J.; Kedzierski, L.; Compton, B. J.; Hayman, (32) Liu, Y. H.; Wang, Z. Y.; Yu, F.; Li, M. J.; Zhu, H. M.; Wang, K.;
C. M.; Osmond, T. L.; Tang, C. W.; Farrand, K. J.; Koay, H. F.; Meng, M.; Zhao, W. The Adjuvant of a-Galactosylceramide Presented
Almeida, C. F. D. S. E.; et al. Augmenting Influenza-Specific T Cell by Gold Nanoparticles Enhances Antitumor Immune Responses of
Memory Generation with a Natural Killer T Cell-Dependent MUC1 Antigen-Based Tumor Vaccines. Int. J. Nanomed 2021, 16,
Glycolipid-Peptide Vaccine. ACS Chem. Biol. 2017, 12 (11), 2898− 403−420.
2905. (33) Dolen, Y.; Kreutz, M.; Gileadi, U.; Tel, J.; Vasaturo, A.; van
(20) Holz, L. E.; Chua, Y. C.; de Menezes, M. N.; Anderson, R. J.; Dinther, E. A. W.; van Hout-Kuijer, M. A.; Cerundolo, V.; Figdor, C.
Draper, S. L.; Compton, B. J.; Chan, S. T. S.; Mathew, J.; Li, J.; G. Co-delivery of PLGA encapsulated invariant NKT cell agonist with
Kedzierski, L.; Wang, Z. F.; et al. Glycolipid-peptide vaccination antigenic protein induce strong T cell-mediated antitumor immune
induces liver-resident memory CD8(+) T cells that protect against responses. Oncoimmunology 2016, 5 (1), No. e1068493.
rodent malaria. Sci. Immunol 2020, 5 (48), No. eaaz8035. (34) Rahimian, S.; Fransen, M. F.; Kleinovink, J. W.; Christensen, J.
(21) Meijlink, M. A.; Chua, Y. C.; Chan, S. T. S.; Anderson, R. J.; R.; Amidi, M.; Hennink, W. E.; Ossendorp, F. Polymeric nano-
Rosenberg, M. W.; Cozijnsen, A.; Mollard, V.; McFadden, G. I.; particles for co-delivery of synthetic long peptide antigen and poly IC
Draper, S. L.; Holz, L. E.; et al. 6 “-Modifed alpha-GalCer-peptide as therapeutic cancer vaccine formulation. J. Controlled Release 2015,
conjugate vaccine candidates protect against liver-stage malaria. RSC 203, 16−22.
Chem. Biol. 2022, 3, 551−560. (35) Mascaraque, A.; Kowalczyk, W.; Fernandez, T.; Palomares, F.;
(22) Anderson, R. J.; Tang, C. W.; Daniels, N. J.; Compton, B. J.; Mayorga, C.; Andreu, D.; Rojo, J. Glycodendropeptides stimulate
Hayman, C. M.; Johnston, K. A.; Knight, D. A.; Gasser, O.; Poyntz, H. dendritic cell maturation and T cell proliferation: a potential influenza
C.; Ferguson, P. M.; et al. A self-adjuvanting vaccine induces cytotoxic A virus immunotherapy. MedChemComm 2015, 6 (10), 1755−1760.
T lymphocytes that suppress allergy. Nat. Chem. Biol. 2014, 10 (11), (36) Cai, H.; Sun, Z. Y.; Chen, M. S.; Zhao, Y. F.; Kunz, H.; Li, Y.
943−949. M. Synthetic Multivalent Glycopeptide-Lipopeptide Antitumor

807 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808
Bioconjugate Chemistry pubs.acs.org/bc Article

Vaccines: Impact of the Cluster Effect on the Killing of Tumor Cells. killer cells is essential for the antimetastatic effect of alpha-
Angew. Chem., Int. Ed. Engl. 2014, 53 (6), 1699−1703. galactosylceramide. Blood 2002, 99 (4), 1259−1266.
(37) Tam, J. P. Synthetic Peptide Vaccine Design - Synthesis and
Properties of a High-Density Multiple Antigenic Peptide System. Proc.
Natl. Acad. Sci. U. S. A. 1988, 85 (15), 5409−5413.
(38) Mitchell, J. P.; Roberts, K. D.; Langley, J.; Koentgen, F.;
Lambert, J. N. A direct method for the formation of peptide and
carbohydrate dendrimers. Bioorg. Med. Chem. Lett. 1999, 9 (19),
2785−2788.
(39) Mihov, G.; Grebel-Koehler, D.; Lubbert, A.; Vandermeulen, G.
W. M.; Herrmann, A.; Klok, H. A.; Mullen, K. Polyphenylene
dendrimers as scaffolds for shape-persistent multiple peptide
conjugates. Bioconjugate Chem. 2005, 16 (2), 283−293.
(40) Shrestha, R.; Petley, E. V.; Farrand, K. J.; Jamieson, S. A.; Jiao,
W. T.; Teesdale-Spittle, P. H.; Mace, P. D.; Hermans, I. F.; Rendle, P.
M. The Synthesis and Anti-tumour Properties of Poly Ethoxy Ethyl
Glycinamide (PEE-G) Scaffolds with Multiple PD-1 Peptides
Attached. ChemMedChem. 2020, 15 (13), 1128−1138.
(41) Toms, S.; Carnachan, S. M.; Hermans, I. F.; Johnson, K. D.;
Khan, A. A.; O’Hagan, S. E.; Tang, C. W.; Rendle, P. M. Poly Ethoxy
Ethyl Glycinamide (PEE-G) Dendrimers: Dendrimers Specifically
Designed for Pharmaceutical Applications. ChemMedChem. 2016, 11
(15), 1583−1586.
(42) Speir, M.; Authier-Hall, A.; Brooks, C. R.; Farrand, K. J.;
Compton, B. J.; Anderson, R. J.; Heiser, A.; Osmond, T. L.; Tang, C.
W.; Berzofsky, J. A.; et al. Glycolipid-peptide conjugate vaccines
enhance CD8(+) T cell responses against human viral proteins. Sci.
Rep 2017, 7, 14273.
(43) Dubowchik, G. M.; Firestone, R. A.; Padilla, L.; Willner, D.;
Hofstead, S. J.; Mosure, K.; Knipe, J. O.; Lasch, S. J.; Trail, P. A.
Cathepsin B-labile dipeptide linkers for lysosomal release of
doxorubicin from internalizing immunoconjugates: model studies of
enzymatic drug release and antigen-specific in vitro anticancer activity.
Bioconjugate Chem. 2002, 13 (4), 855−869.
(44) Bryden, F.; Martin, C.; Letast, S.; Lles, E.; Vieitez-Villemin, I.;
Rousseau, A.; Colas, C.; Brachet-Botineau, M.; Allard-Vannier, E.;
Larbouret, C.; et al. Impact of cathepsin B-sensitive triggers and
Recommended by ACS
hydrophilic linkers on in vitro efficacy of novel site-specific antibody-
drug conjugates. Org. Biomol Chem. 2018, 16 (11), 1882−1889. Dual-Responsive Glycopolymers for Intracellular Codelivery
(45) Poreba, M. Protease-activated prodrugs: strategies, challenges, of Antigen and Lipophilic Adjuvants
and future directions. FEBS J. 2020, 287 (10), 1936−1969. Judith De Mel, Thomas A. Werfel, et al.
(46) Flechtner, J. B.; Cohane, K. P.; Mehta, S.; Slusarewicz, P.; NOVEMBER 14, 2022
Leonard, A. K.; Barber, B. H.; Levey, D. L.; Andjelic, S. High-affinity MOLECULAR PHARMACEUTICS READ
interactions between peptides and heat shock protein 70 augment
CD8(+) T lymphocyte immune responses. J. Immunol 2006, 177 (2), Identification of a Novel Small Molecule That Enhances the
1017−1027. Release of Extracellular Vesicles with Immunostimulatory
(47) Gibson, B.; Verkade, J.; Barta, N.; Hodges, J.; van Delft, F. Potency via Induction of Calcium Influx
Bicyclo[6.1.0]nonyne Phoshoramidites for Metal-Free Conjugation of
Yukiya Sako, Tomoko Hayashi, et al.
Oligonucleotides. Chimica Oggi 2013, 31, 18−21.
APRIL 11, 2023
(48) Spangler, B.; Yang, S.; Baxter Rath, C. M.; Reck, F.; Feng, B. Y. ACS CHEMICAL BIOLOGY READ
A Unified Framework for the Incorporation of Bioorthogonal
Compound Exposure Probes within Biological Compartments. ACS
Covalently Conjugated NOD2/TLR7 Agonists Are Potent
Chem. Biol. 2019, 14 (4), 725−734.
and Versatile Immune Potentiators
(49) Burn, O. K.; Farrand, K.; Pritchard, T.; Draper, S.; Tang, C. W.;
Mooney, A. H.; Schmidt, A. J.; Yang, S. H.; Williams, G. M.; Brimble, Samo Guzelj, Žiga Jakopin, et al.
M. A.; et al. Glycolipid-peptide conjugate vaccines elicit CD8(+) T- NOVEMBER 06, 2022
JOURNAL OF MEDICINAL CHEMISTRY READ
cell responses and prevent breast cancer metastasis. Clin Transl
Immunology 2022, 11 (7), No. e1401.
(50) Dirksen, A.; Hackeng, T. M.; Dawson, P. E. Nucleophilic Nanovaccine Based on a Biepitope Antigen to Potentiate the
catalysis of oxime ligation. Angew. Chem., Int. Ed. Engl. 2006, 45 (45), Immunogenicity of a Neoantigen
7581−7584. Shuyu Chen, Guosong Chen, et al.
(51) Carnaud, C.; Lee, D.; Donnars, O.; Park, S. H.; Beavis, A.; FEBRUARY 08, 2023
Koezuka, Y.; Bendelac, A. Cutting edge: Cross-talk between cells of ACS MACRO LETTERS READ
the innate immune system: NKT cells rapidly activate NK cells. J.
Immunol 1999, 163 (9), 4647−4650.
(52) Smyth, M. J.; Crowe, N. Y.; Pellicci, D. G.; Kyparissoudis, K.;
Get More Suggestions >
Kelly, J. M.; Takeda, K.; Yagita, H.; Godfrey, D. I. Sequential
production of interferon-gamma by NK1.1(+) T cells and natural

808 https://doi.org/10.1021/acs.bioconjchem.3c00106
Bioconjugate Chem. 2023, 34, 799−808

You might also like