Вы находитесь на странице: 1из 18

Articles

https://doi.org/10.1038/s41565-019-0512-0

Immunization with mannosylated nanovaccines


and inhibition of the immune-suppressing
microenvironment sensitizes melanoma to
immune checkpoint modulators
João Conniot   1,2,7, Anna Scomparin   1,3,7, Carina Peres2, Eilam Yeini1, Sabina Pozzi1, Ana I. Matos2,
Ron Kleiner1, Liane I. F. Moura2, Eva Zupančič2,4, Ana S. Viana5, Hila Doron6, Pedro M. P. Gois2,
Neta Erez6, Steffen Jung4, Ronit Satchi-Fainaro   1* and Helena F. Florindo   2*

A low response rate, acquired resistance and severe side effects have limited the clinical outcomes of immune checkpoint ther-
apy. Here, we show that combining cancer nanovaccines with an anti-PD-1 antibody (αPD-1) for immunosuppression blockade
and an anti-OX40 antibody (αOX40) for effector T-cell stimulation, expansion and survival can potentiate the efficacy of mela-
noma therapy. Prophylactic and therapeutic combination regimens of dendritic cell-targeted mannosylated nanovaccines with
αPD-1/αOX40 demonstrate a synergism that stimulates T-cell infiltration into tumours at early treatment stages. However,
this treatment at the therapeutic regimen does not result in an enhanced inhibition of tumour growth compared to αPD-1/
αOX40 alone and is accompanied by an increased infiltration of myeloid-derived suppressor cells in tumours. Combining the
double therapy with ibrutinib, a myeloid-derived suppressor cell inhibitor, leads to a remarkable tumour remission and pro-
longed survival in melanoma-bearing mice. The synergy between the mannosylated nanovaccines, ibrutinib and αPD-1/αOX40
provides essential insights to devise alternative regimens to improve the efficacy of immune checkpoint modulators in solid
tumours by regulating the endogenous immune response.

A
dvances in immunotherapy have improved melanoma treat- (DCs) and consequent T-cell priming and activation could potenti-
ment outcomes, based on antitumour immune responses ate the antitumour responses and survival outcomes of the combi-
elicited by the patient’s own immune system. Cytotoxic nation of PD-1 blockade and OX40 co-stimulation.
T-lymphocyte-associated protein-4 (CTLA-4) and programmed Here, we report the design, synthesis and characterization of
cell death protein 1 (PD-1) blockade have enhanced specific T-cell- biodegradable nanoparticles made of non-mannosylated and man-
mediated antitumour immunity in clinical practice1,2. With these nosylated polylactic-co-glycolic acid (PLGA)/polyl(l-actic acid)
findings, efforts are being made not only to inhibit negative immune (PLA) (NP and man-NP) (Fig. 1a) for the in vivo co-delivery of mel-
checkpoints, but also to stimulate signalling pathways of the immune anoma-associated antigens and Toll-like receptor (TLR) agonists
system. OX40 is a co-stimulatory receptor member of the tumour- (CpG and monophosphoryl lipid A (MPLA)) to DCs, via phago-
necrosis factor (TNF) family that is transiently expressed on activated cytosis-dependent (‘passive’) and ligand-mediated (‘active’ through
T cells. Once activated, OX40 induces expansion, trafficking and the the mannose receptor (MR/CD206)) targeting, respectively12.
survival of effector T cells, and increases pro-inflammatory cytokine Under cancer-associated inflammation, an aberrant myelopoi-
secretion3,4. OX40-signalling can also block the inhibitory activity esis leads to the accumulation of myeloid-derived suppressor cells
of tumour-infiltrating regulatory CD4+ T cells by hindering Foxp3, (MDSCs), which compromise the efficient maturation of DCs and
transforming growth factor beta (TGF-β) and interleukin-10 (IL-10) thus affects the antigen presentation process and drives T-cell tran-
signalling, known to suppress vaccine-induced immune responses5–7. scriptional programmes towards T-cell anergy and exhaustion. The
Despite the promising clinical results, immune checkpoint ther- presence of MDSCs was also correlated to a decreased efficacy of anti-
apy has been associated with low percentages of effective and durable CTLA-413,14. Therefore, we further administered ibrutinib, an irrevers-
responses to single-agent therapies due to resistance or relapse8–10. ible inhibitor of Bruton’s tyrosine kinase and interleukin-2-inducible
The presence of tumour-infiltrating lymphocytes has been corre- T-cell kinase, to modulate the MDSC generation and function15,16.
lated with favourable outcomes of immune checkpoint modulation
therapies11. Based on these observations, and in an effort to integrate Polymeric nanoparticles as antitumour nanovaccines
a novel therapeutic approach, we hypothesized that cancer vaccines The synthesis of the mannose-grafted PLGA polymer was con-
that deliver tumour-associated antigens (TAAs) to dendritic cells firmed by the multiplet signal between 3.2 and 3.8 ppm in the

1
Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. 2Research Institute for Medicines (iMed.
ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal. 3Department of Drug Science and Technology, University of Turin, Turin, Italy.
4
Department of Immunology, Weizmann Institute of Science, Rehovot, Israel. 5Center of Chemistry and Biochemistry, Faculty of Sciences, Universidade de
Lisboa, Lisbon, Portugal. 6Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. 7These authors contributed equally:
João Conniot, Anna Scomparin. *e-mail: ronitsf@tauex.tau.ac.il; hflorindo@ff.ulisboa.pt

Nature Nanotechnology | www.nature.com/naturenanotechnology


Articles NATure NAnOTeCHnOlOgy

a e P = 0.001

Median fluorescence intensity


Melan A/MART-1 25
peptide NP
MPLA
20 Man-NP
(immune potentiator)
D-α-tochoperyl

(×105)
15
PEG1000 succinate
(immune potentiator) 10

5
CpG 0
(immune potentiator) Mannose 0 3 6 9 12 15 18 21 24
Incubation time (h)

b c f
NP Man-NP

2 µm
500 nm

µm
5
2.
5
1. Wheat germ agglutinin (membrane)
0 650 nm 5 Rhodamine-labelled particles
0.
DAPI (nucleus)

g h
P = 0.0018

P < 0.0001
50 Man-NP negative cells

P = 0.0026 40 Man-NP positive cells P = 0.0040


% APC with internalized NP

40
MFI of CD11b+ CD11c+ MHCII+

P = 0.0008 P = 0.0284
30
30
Plain man-NP P = 0.0013
(×102)

Antigen-loaded man-NP
20 20

10 10

0 0
CD11b+CD11c+ CD11b+CD11c– CD11b–CD11c+
Migratory DC Macrophages Resident DC CD80+ CD86+ MHCI+

Fig. 1 | NP and man-NP are potential delivery systems for vaccination. a, Schematic representation of a man-NP. b, Transmission electron microscopy
image of spherical man-NP. c, Scanning electron microscopy image of spherical man-NP. d, The AFM images of spherical man-NP show a narrow size
polydispersity. e, Particle internalization by the DCs determined by fluorescence-activated cell sorting. Non-treated cells and non-labelled empty NP and
empty man-NP were used as the negative controls. Data are presented as mean ± s.d., N = 4 independent samples with three technical replicates.
f, Confocal images of DCs after 3 h of incubation with NP (left) and man-NP (right). Z-stacks (top) and projections (bottom) (N = 3, n = 6, where N
denotes the number of independent experiments and n denotes the number of measurements per experiment). Scale bars, 50 μm. g, Percentage of
man-NP internalization 17 h after immunization with empty man-NP or antigen-loaded man-NP. Man-NP were preferentially internalized by migratory DCs
in immunized C57BL/6J mice and increased the expression of the activation and maturation markers of these APCs. h, Median florescence intensity (MFI)
of activated DCs that internalized man-NP present in the LNs 17 h after immunization. Mean ± s.d., N = 3, n = 3. Statistics: two-way analysis of variance
(ANOVA) followed by Tukey post-hoc test (g) or Bonferroni test (h).

1
H NMR spectra, which indicates the presence of mannose depending on the entrapped molecules, with a low polydispersity
(Supplementary Fig. 1)17. index (0.13 ± 0.04 to 0.18 ± 0.04) (Supplementary Table 1).
NP and man-NP presented a near-neutral surface charge and Electron microscopy and atomic force microscopy (AFM)
similar average hydrodynamic diameters (166 ± 5 nm to 181 ± 8 nm), showed homogeneous spherical particles, with a slight roughness

Nature Nanotechnology | www.nature.com/naturenanotechnology


NATure NAnOTeCHnOlOgy Articles
on the surface (Fig. 1b–d). Concanavalin A (Conc A) binds to Splenocytes collected from mice immunized with man-NP
exposed mannose residues to form aggregates (Supplementary MHCI-ag/man-NP MHCII-ag exhibited the highest ex vivo cyto-
Fig. 2a)18,19. After incubation of man-NP, Conc A induced par- toxicity when co-cultured with Ret-mCherry-sorted (RMS) murine
ticle aggregation, revealed by a fivefold increase in average size melanoma cells (Fig. 2e). Man-NP MHCI-ag/man-NP MHCII-ag
(Supplementary Fig. 2b) and confirmed by fluorescence microscopy elicited a superior CTL activity compared to man-NP MHCI-ag
(Supplementary Fig. 2c). (P = 0.014), to NP MHCI-ag (P = 0.02) and to NP MHCI-ag/NP
NP and man-NP carried the Melan-A/MART-1(26-35(A27L)) MHCII-ag (P = 9.6 × 10–6). MHCI-ag/MHCII-ag/CpG/MPLA (free
major histocompatibility complex class I (MHCI)-restricted peptide antigens/immune potentiators) or empty particles induced a CTL
(MHCI-ag) and the Melan-A/MART-1(51-73) MHCII-restricted activity similar to that of PBS (Fig. 2e).
peptide (MHCII-ag) aimed at the MHC class I and class II antigen The CTL activity induced by man-NP MHCI-ag/man-NP
presentation pathways, respectively. These were shown to potentiate MHCII-ag is clearly shown by the increased fluorescent staining
active vaccination strategies by engagement of both CD4+ and CD8+ related to caspase 3/7 activation, a marker of apoptosis (Fig. 2f).
T  cells18. NP and man-NP showed high levels of entrapment effi- The pro-inflammatory cytokine secretion profile induced
ciency (EE) and loading capacity (LC) (Supplementary Table 2). For by our nanovaccines clearly correlated with this enhanced ex
the MHCI-ag, EE >97.5 ± 0.2% and LC >48.8 ± 0.1 μg mg–1, whereas vivo CTL activity. Importantly, the presence of mannose on the
for the MHCII-ag EE >74.6 ± 3.5% and LC >37.3 ± 1.7 μg mg–1. The surface and the particulate nature of our nanovaccines co-entrap-
EE of CpG was above 80.8 ± 2.5%, which corresponds to a LC of ping the antigens and TLR agonists were crucial to the recogni-
8.1 ± 0.3 µg mg–1. tion and destruction of target cells by activating splenocytes in
NP and man-NP did not affect DC viability (>90%) in the con- healthy animals.
centration range tested (Supplementary Fig. 3a), which supports There was no evidence of body weight change in all the
their physiological biocompatibility. Also, NP and man-NP did groups, which attests to the vaccine tolerability and safety
not disrupt red blood cell membranes at concentrations up to (Supplementary Fig. 6).
20 mg ml–1 (Supplementary Fig. 3b).
Man-NP exhibited higher levels of internalization (P = 0.001) Combination of nanovaccines with αPD-1/αOX40
into murine immature DC (JAW SII) than did NP (Fig. 1e). Confocal The combination dosing schedule of man-NP MHCI-ag/man-
microscopy corroborated these fluorescence-activated cell-sorting NP MHCII-ag with αPD-1/αOX40 (Fig. 3a) induced a strong
(FACS) results (Fig. 1f). We observed an increase in Cy5.5-labelled and safe tumour growth inhibition (Fig. 3b–e and Supplementary
NP and man-NP uptake when d-α-tocopherol polyethylene glycol Figs. 7 and 8).
1000 succinate (TPGS) was used to prepare these formulations, At day 17 after the tumour inoculation, the tumour volume
compared with poly(vinyl alcohol) (PVA) (Supplementary Fig. 4). in animals treated only with αPD-1/αOX40 or with the combina-
The higher uptake obtained for man-NP (TPGS) was reduced in the tion of non-mannosylated (NP MHCI-ag/NP MHCII-ag) or man-
presence of soluble mannose. Therefore, the higher internalization nosylated (man-NP MHCI-ag/man-NP MHCII-ag) nanovaccines
obtained for man-NP was mediated by the interaction of mannose with αPD-1/αOX40 was much lower compared to animals treated
with the MR/CD206 receptor at the DC surface. with PBS (P < 0.0016) (Fig. 3d). At day 27, the tumour volume
Our man-NP (TPGS) was preferentially taken-up in  vivo by intragroup variability increased for all the groups, except for that
migratory DCs, compared to macrophages and resident DCs (Fig. treated with man-NP MHCI-ag/man-NP MHCII-ag  + αPD-1/
1g and Supplementary Fig. 5). This nanovaccine was able to signifi- αOX40, in which the tumours remained small (Fig. 3e and
cantly increase the expression of activation/maturation markers at Supplementary Fig. 8).
the DC surface (Fig. 1h). The combination man-NP MHCI-ag/man-NP MHCII-ag  + 
αPD-1/αOX40 resulted in 100% survival 42 days after the tumour
Cytotoxic activity induced by nanovaccines inoculation, with a survival rate of 50% at day 60. The treatment
Cy5.5-labelled NP or man-NP remained at the site of immuni- with αPD-1/αOX40 resulted in only 20% survival (Fig. 3c).
zation 48 h after subcutaneous (s.c.) injection, which promoted In clinical trials, the use of MHCII-ag peptides in cancer vacci-
contact with DCs at the periphery (inguinal lymph nodes (LNs)) nation has been associated with poor prognosis due to the increased
as well as proximity to the popliteal and axillary LNs. This prefer- activity of CD4+CD25+ Treg cells or to the apoptosis of activated
ential accumulation in the LNs is important for the induction of CD8+ T cells29,30. In our study, the use of MHCI-ag and MHCII-ag
antigen-specific adaptive immune responses and vaccine efficacy20. peptides was essential for the induction of a robust antitumour
Additionally, by presenting a mean average diameter below 200 nm, response31, but only when delivered by our nanoplatform, as the free
these nanovaccines may also be suitable for trafficking through administration of the same antigens induced a much lower efficacy.
the lymphatic drainage directly to the lymphoid organs21. Residual This is an indication that the antitumour immune-mediated effect
accumulation was detected in the liver and the kidney due to the resulted from the epitopes’ increased immunogenicity conferred by
excretion of metabolic particle derivatives (Fig. 2a,b). the adjuvant effect of our nanovaccine.
The groups treated with man-NP (Fig. 2c) showed the highest Immunohistochemistry staining revealed high levels of cas-
levels of interferon gamma (IFN-γ) and granulocyte–macrophage pase-3 and tumour-infiltrating CD4+ and CD8+ T  cells in all the
colony-stimulating factor (GM-CSF), which are associated with groups immunized with nanovaccines (Supplementary Fig. 9).
enhanced antigen priming and subsequent presentation by antigen- The highest level of tumour-infiltrating CD8+ T  cells was
presenting cells (APCs)22,23. IFN-γ is a predictor of antigen-specific observed in groups treated with mannosylated nanovaccines alone
cytotoxic CD8+ T-lymphocyte (CTL)-mediated responses24 (Fig. 2d). or in combination with αPD-1/αOX40, and with non-mannosyl-
Our nanovaccines also increased the secretion of IL-6, which is ated nanovaccines alone. Therefore, the nanovaccines are capable
involved in T-cell recruitment and differentiation, and in the sup- of reactivating cell-mediated cytotoxicity in the tumour microenvi-
pression of regulatory T (Treg) cells25,26. Additionally, our nano- ronment (TME), as previously observed ex vivo.
vaccines also led to a lower secretion of the T helper cell 2 (TH2) The synergism observed with the combination of prophylactic
cytokines macrophage inflammatory protein (MIP-1β)/chemokine nanovaccines with αPD-1/αOX40 prompted the design of a thera-
(C–C motif) ligand 4 (CCL4) and CCL12, also associated with peutic intervention strategy in mice inoculated with Ret-melanoma
the inhibition of Treg cells, reduction of tumour angiogenesis and cells or RMS cells (Fig. 4a), which induced similar tumour growth
enhanced antitumour efficacy27,28. profiles (Supplementary Fig. 10).

Nature Nanotechnology | www.nature.com/naturenanotechnology


Articles NATure NAnOTeCHnOlOgy

a b
3h 48 h 3h 48 h

3 NP

(scaled counts s–1 mm–2)


man-NP

Total signal
1

rt

en

LN
ve

ng

ey
ea

le
Li

dn
Lu

Sp
Ki
c d 15 IFN-γ

to standard control (%)


Secretion relative
Splenocyte harvesting
–24 –17 –10 10
0 6
5
1st 2nd 3rd
Nanovaccine Cytokine and Cytotoxic activity 0
chemokine secretion on RMS cells

40 GM-CSF TNF-α 8 IL-2 15 TARC/CCL17


120
to standard control (%)
Secretion relative

30 6
80 10
20 4
40 5
10 2

0 0 0 0
IL-6 CCL1/TCA-3 80 MIP-1β/CCL4 150 MCP-5/CCL12
80 10
to standard control (%)

120
Secretion relative

8 60
60
6 90
40 40
4 60
20 2 20 30

0 0 0 0

PBS NP (empty) MHCI-ag/CpG/MPLA (free) MHCI-ag/MHCII-ag/CpG/MPLA (free)


NP MHCI-ag NP MHCI-ag/NP MHCII-ag Man-NP MHCI-ag Man-NP MHCI-ag/man-NP MHCII-ag

e 40 f
0h 8h 12 h
P = 0.014 P = 0.010
Total cell death area

30
(×105 µm2 per well)

P < 0.0001

P < 0.0001

20

10 0h 8h 12 h

0
0 2 4 6 8 10 12
Time (h in co-culture)
RMS cells Caspase 3/7
Untreated PBS
NP (empty) MHCI-ag (free)
MHCI-ag/MHCII-ag (free) NP MHCI-ag
NP MHCI-ag/NP MHCII-ag Man-NP MHCI-ag
Man-NP MHCI-ag/man-NP MHCII-ag

Fig. 2 | NP and man-NP vaccines induce splenocyte activation and ex vivo cytotoxicity against melanoma cells. a, Non-invasive intravital fluorescence
imaging of C57BL/6J mouse 3 h and 48 h after hock immunization with NP (left) and man-NP (right). b, Organ biodistribution according to a fluorescence
signal (N = 3 animals) with NP and man-NP. Data represent mean ± s.d. c, Immunization scheme of C57BL/6J mice and ex vivo splenocyte cytotoxic
activity timeline. d, Secretion of IFN-γ, GM-CSF, TNF-α, IL-2, IL-6, CCL1/TCA-3, MIP-1β, monocyte chemotactic protein 5 (MCP-5)/CCL12 and thymus-
and activation-regulated chemokine (TARC/CCL17) on the restimulation of splenocytes in culture. The highest levels of IFN-γ and GM-CSF were induced
by man-NP. Elevated levels of CCL1/TCA-3 and TARC/CCL17, which are chemokines associated with activated T cells, also suggested that our particles
played a major role in priming antigen-specific CD8+ T cells. An inclusive assessment of the triad IFN-γ, IL-2 and TNF-α predicted an improved cytotoxic
CD8+ T-cell activity for antigen-loaded man-NP. These nanovaccines also presented a role in the modulation of the TH2 chemokine secretion profile.
NP MHCI-ag/NP MHCII-ag and man-NP MHCI-ag/man-NP MHCII-ag decreased the MIP-1β/CCL4 secretion, whereas both man-NP MHCI-ag and
the combination man-NP MHCI-ag/man-NP MHCII-ag equally reduced the MCP-5/CCL12 levels. e, Cytotoxic activity of splenocytes harvested from
immunized C57BL/6J mice after incubation with Melan-A/MART-1 and CD28 in solution for 6 days. Data are presented as mean ± s.e.m., N = 6 biological
independent samples. One-way ANOVA with Tukey test. f, Images of RMS cells co-cultured with reactivated splenocytes from the group immunized
with NP MHCI-ag/NP MHCII-ag (top) and the group immunized with man-NP MHCI-ag/man-NP MHCII-ag (bottom). Cell death was detected with an
apoptosis reagent that couples to activated caspase-3/7 recognition motif and quantifies apoptosis (in green). The experiment was repeated five times
with similar results. Scale bars, 300 μm.
Nature Nanotechnology | www.nature.com/naturenanotechnology
NATure NAnOTeCHnOlOgy Articles
a b 2,000

Tumour volume (mm3)


1,600
αPD-1/αOX40
1,200
P = 0.029
–28 –21 –14 0 9 13 17 20 24 800 P = 0.016

4.5 × 105 400


Nanovaccine RMS
0
6 10 14 18 22 26 30 34 38 42
Time (days after tumour inoculation)

c d P = 0.0049 e
100
P = 0.0016
P = 0.0081
80 2,000 2,500
P = 0.0041

Tumour volume (mm3)


Survival (%)

Tumour volume (mm3)


60 1,500 2,000
P = 0.047
1,500
40 1,000
1,000
P = 0.023
20 500
500

0 0 0
6 12 18 24 30 36 42 48 54 60 66 Treatment groups Treatment groups
Time (days after tumour inoculation)
PBS αPD-1/αOX40
MHCI-ag/MHCII-ag/CpG/MPLA (free) MHCI-ag/MHCII-ag/CpG/MPLA (free) + αPD-1/αOX40
NP MHCI-ag/NP MHCII-ag NP MHCI-ag/NP MHCII-ag + αPD-1/αOX40
Man-NP MHCI-ag/man-NP MHCII-ag Man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40

Fig. 3 | Prophylactic nanovaccines have a synergistic effect with PD-1 blockade and OX40 activation, which restricts melanoma growth and prolongs
survival. a, Timeline (days) of immunization, tumour inoculation and immune checkpoint therapy. b, Tumour growth curve. P values correspond to tumour
volume at day 17. Data are presented as mean ± s.e.m. (N = 4 animals for the PBS group and N = 5 animals for the remaining groups). One-way ANOVA.
c, Kaplan–Meier overall survival over time graph for mice inoculated with 4.5 × 105 RMS cells (N = 4 animals for the PBS group and N = 5 animals for the
remaining groups). Log-rank test, P < 0.05 for the combination man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40 compared to all other treatment
groups. d, Individual tumour volume at day 17 (N = 4 animals for the PBS group and N = 5 animals for the remaining groups) with mean ± s.e.m. Unpaired
two-tailed t test. e, Day 27 after tumour inoculation (N = 5 animals) with mean ± s.e.m.

The groups αPD-1/αOX40 and man-NP MHCI-ag/man-NP αOX40-treated group. These extremely elevated levels of MDSCs
MHCII-ag + αPD-1/αOX40 showed a similar average tumour vol- seem to correlate with the marked decrease of CD8+ T-cell infil-
ume at day 18, which was sixfold significantly smaller compared tration (Fig. 4d and Supplementary Fig. 12) and with the high
to the PBS-treated group (P < 0.0012) (Fig. 4b), with negligible percentage of Treg cells (Fig. 4f and Supplementary Fig. 13) at the
systemic toxicity (Supplementary Fig. 11). The animals treated final end-point.
with the combination had the highest levels of tumour-infiltrat- The infiltration of MDSCs hindered the early effect of the
ing CD8+ T cells (P < 0.001) (Fig. 4d and Supplementary Fig. 12), CD8+ T-cell stimulation and proliferation, and so inhibited T-cell
which supports the synergism previously observed in the prophy- infiltration and CTL activity32,33. This imbalance promoted an
lactic regimen. immunosuppressive TME, and the combination of mannosylated
The treatment with αPD-1/αOX40 induced the lowest lev- nanovaccines + αPD-1/αOX40 failed to show a benefit in compari-
els of CD4+ T-cell infiltrates (Fig. 4e and Supplementary Fig. 12), son with αPD-1/αOX40 alone.
which displayed a reduced infiltration of Treg cells (Fig. 4f and Ibrutinib was shown to limit the generation and migration of
Supplementary Fig. 13). Animals treated with mannosylated nano- MDSCs, and was proposed as an approach to enhance the efficacy
vaccines, alone or in combination, had higher levels of Treg cells of cancer immunotherapies15,34. We hypothesized that the inhibition
compared to the αPD-1/αOX40-treated group. Therefore, the high- of these MDSCs with ibrutinib could improve the clinical outcomes
est CD8:Treg ratio was observed for the αPD-1/αOX40 treatment, of our strategy.
followed by the combination of mannosylated nanovaccines with
αPD-1/αOX40 (Fig. 4g). Combination of nanovaccines, αPD-1/αOX40 and ibrutinib
At the final end-point, the animals treated with αPD-1/αOX40 Melanoma-bearing mice were treated according to the schedule
remained with the highest CD8:Treg ratio. This was sevenfold higher in Fig. 5a. On day 20 after the tumour inoculation, the average
compared to the groups treated with mannosylated nanovaccines, tumour volume of groups treated with ibrutinib or with man-
alone or in combination with αPD-1/αOX40 (P < 0.01). nosylated nanovaccines + ibrutinib were identical to that of the
Interestingly, from day 18 to the final end-point, the infil- PBS-treated group. However, groups treated with the trivalent
tration of CD11b+Gr-1+ MDSCs increased significantly in the therapy (man-NP + αPD-1/αOX40 + ibrutinib) or with αPD-1/
two groups treated with mannosylated nanovaccines, alone or in αOX40 + ibrutinib showed a significantly reduced tumour vol-
combination (Fig. 4h and Supplementary Fig. 14). At the final ume by more than fivefold (P = 0.0058 and P = 0.0060, respec-
end-point, the levels of MDSCs in these groups were approxi- tively) (Fig. 5b). Importantly, there was no difference between
mately fourfold higher than those obtained for the αPD-1/ this antitumour response induced by our triple combination

Nature Nanotechnology | www.nature.com/naturenanotechnology


Articles NATure NAnOTeCHnOlOgy

a b 2,000

Tumour volume (mm3)


1,500

αPD-1/αOX40
1,000
P = 0.012

0 7 14 21 24 27 30 500
P = 0.007

3 × 10 5 Nanovaccine 0
5 10 15 20 25 30 35
RMS cells
Time (days after tumour inoculation)

PBS αPD-1/αOX40 Man-NP MHCI-ag/man-NP MHCII-ag Man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40

c P = 0.01 d P < 0.001


e
P = 0.015
P = 0.02 P < 0.001 P = 0.003

P = 0.001 P < 0.001 P = 0.003 P = 0.007


P = 0.01 P = 0.01
60 50
P < 0.001

P < 0.001 P < 0.001 40


P = 0.003 P = 0.001

40
(% of CD3+ T cells)

P = 0.006

(% of CD3+ T cells)
CD3+ T cells per

30
40
total live cells

P = 0.003
30
CD8+

CD4+
20
20
20
10
10

0 0 0

Day 18 Final end-point Day 18 Final end-point Day 18 Final end-point

f g P = 0.029 h
P = 0.011 P = 0.016 P = 0.016 P < 0.001
40 8 100
P = 0.003 P < 0.001
(% of CD3+ CD4+ T cells)

30 80 P = 0.007
6
(% of single cells)
CD8 : Treg ratio

P = 0.002 P = 0.05 P = 0.011


60
MDSCs
Treg

20 P = 0.03
4
P = 0.002
40
+

P < 0.001
10 2
20

0 0 0

Day 18 Final end-point Day 18 Final end-point Day 18 Final end-point

PBS αPD-1/αOX40 Man-NP MHCI-ag/man-NP MHCII-ag Man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40

Fig. 4 | Low CD8+:Treg ratio and high infiltration of MDSCs (CD11b+Gr-1+MDSC) compromise the therapeutic efficacy of the combination of
mannosylated nanovaccines with αPD-1/αOX40. a, Timeline (days) of tumour inoculation and treatments. b, Tumour growth curve. Data are presented
as mean ± s.e.m. (N = 7 animals). P values correspond to tumour volume at day 18 after the tumour inoculation. One-way ANOVA. c–h, Tumour-infiltrating
immune cell populations for CD3+ (c), CD8+ (d), CD4+ (e), Treg (f), CD8+:Treg ratio (g) and MDSCs (h). Tumours were isolated on day 18 after the tumour
inoculation and when the tumour volume for the final end-point was reached. Quantification was performed by flow cytometry. Data are presented as
mean ± s.d., N ≥ 3 animals. Unpaired two-tailed t test.

therapy in animals inoculated with Ret melanoma cells or RMS Melan-A/MART-1 specificity was further confirmed by the IFN-γ
cells (Supplementary Fig. 10). ELISpot assay (Supplementary Fig. 15e). RMS-bearing mice treated
In addition, man-NP was significantly superior to NP from day with man-NP presented the highest levels of antigen-specific intra-
19 onwards (P = 0.005), which demonstrates the crucial role of the cellular IFN-γ+ stained in tumour-infiltrating cytotoxic CD8+
mannose-targeting moieties in achieving an effective immune- T lymphocytes, at the first end-point (Supplementary Fig. 15f).
mediated control of the tumour growth (Supplementary Fig. 15a,b). On day 33, animals treated with man-NP MHCI-ag/man-NP
In fact, on day 23, the man-NP-treated group had an average tumour MHCII-ag + αPD-1/αOX40 + ibrutinib presented a threefold higher
volume of 54 mm3, in contrast to the NP-treated group, which had survival percentage (P =  0.0049) compared to animals treated
an average tumour volume of 638 mm3. with NP MHCI-ag/NP MHCII-ag + αPD-1/αOX40 + ibrutinib, of
The trivalent therapy that included man-NP induced the which 8 out of 13 had already reached a tumour volume of at least
most potent antigen-specific antitumour immune response 1,000 mm3 (Supplementary Fig. 15g).
(P < 0.001 in Fig. 5c,d; and P < 0.01 in Supplementary Fig. 15c,d). Of the 15 animals of the group mannosylated nanovac-
The superior antigen-dependent immune response was irrespec- cine + αPD-1/αOX-40 + ibrutinib, 14 remained alive at day 40,
tive of the antigen used (gp100 or Melan-A/MART-1), as shown by in comparison to 7 out of 15 animals of the group αPD-1/αOX-
a tetramer staining assay (Fig. 5c and Supplementary Fig. 15c,d). 40 + ibrutinib (Fig. 5e). The latter presented higher variability in

Nature Nanotechnology | www.nature.com/naturenanotechnology


NATure NAnOTeCHnOlOgy Articles
terms of tumour size (Fig. 5f). Moreover, 7% of the animals treated Supplementary Figs. 14 and 17). No clear effect was observed on
with mannosylated nanovaccine + αPD-1/αOX-40 and 23% of the tumour-infiltrating APCs and macrophages (Fig. 5l,n).
animals treated with αPD-1/αOX-40 + ibrutinib remained alive The trivalent combination also showed a superior antitumour
after 65 days, whereas 67% of animals treated with the trivalent efficacy in two independent studies using a second mouse model
combination mannosylated nanovaccine + αPD-1/αOX-40 + ibru- that bore an orthotopic s.c. B16-F10 melanoma (Supplementary
tinib survived during that period. The survival curves of the triple Fig. 18a). In this case, the dosing regimen started once the ani-
regimen are statistically different from those obtained for the PBS mals presented palpable tumours, and led to 79% (first study,
(P = 0.0001), man-NP + αPD-1/αOX-40 (P = 0.0001) and αPD-1/ Supplementary Fig. 18b) and 81% (second study, Supplementary
αOX-40 + ibrutinib (P =  0.0256) treatments. Only slight body Fig. 18c) tumour growth inhibition at day 19 for the combina-
weight changes relative to the initial body weight were detected tion of man-NP with ibrutinib and αPD-1/αOX40, versus 67%
throughout the study, which reflects a negligible systemic toxicity (Supplementary Fig. 18b) and 68% (Supplementary Fig. 18c) for
(Supplementary Fig. 11). man-NP + αPD-1/αOX40 combination, and 70% (Supplementary
A higher infiltration of T lymphocytes, as expected, was asso- Fig. 18b) and 57% (Supplementary Fig. 18c) for the αPD-1/
ciated with a stronger tumour growth inhibition (Fig. 5g and αOX40 + ibrutinib combination. The proposed mechanism was
Supplementary Fig. 12). The number of CD8+ T lymphocytes confirmed in this second melanoma model by the reduction of
was more than 20-fold higher for mice treated with αPD-1/ infiltrating MDSCs and the increase of infiltrating T  cells in the
αOX40 + ibrutinib or with man-NP + αPD-1/αOX40 compared to TME (Supplementary Fig. 19). We selected day 19 for comparison
the PBS-treated group (Fig. 5h and Supplementary Fig. 12). as this was the last day of the studies at which all the animals in all
The highest level of CD8+ tumour-infiltrating lymphocytes, at the groups were still present.
the second end-point, was induced by αPD-1/αOX40 + ibrutinib.
At this time point, low levels of Treg cells were observed within the Conclusions
tumours of animals treated with αPD-1/αOX40 + ibrutinib or man- The recent clinical validation of different immune checkpoint mod-
NP + αPD-1/αOX40 (Fig. 5i,j, Supplementary Fig. 13). Both treat- ulators has revolutionized cancer therapy. However, low response
ments also resulted in high CD8:Treg ratios, which correlated with rates, adverse effects and resistance led to the clinical need for alter-
the enhanced therapeutic efficacy (Fig. 5k). native combination strategies to overcome these limitations35.
The prominent infiltration of CD8+ T  cells at day 20 after the These nanovaccines, as an active immunotherapeutic strategy,
tumour inoculation in groups that received man-NP with αPD-1/ have the unique ability to expand the host antitumour specific
αOX40 was further confirmed by immunohistochemistry staining T-effector phenotype, which improves sensitivity and long-term
of RMS tumour sections for CD4 and CD8. Man-NP also induced tumour recognition. The synergistic effect of αPD-1/αOX40,
a higher expression of the OX40 receptor, as shown in the groups already reported in an ovarian cancer model36, could be relevant for
treated with man-NP MHCI-ag/man-NP MHCII-ag  + ibruti- melanoma37 and be potentiated by cancer vaccines that co-deliver
nib and man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40 TAAs and TLR ligands to DCs. The co-delivery of TAAs and TLR
(Supplementary Fig. 16). The co-stimulatory OX40 was then ligands by the same nanoparticle enhances antigen internalization,
available at a greater extent to be targeted by the immune processing and subsequent presentation. This is a key step to over-
checkpoint therapy. come host tolerance to tumour cells by improving the effective
Note that the expression of PD-1/PD-L1 was the lowest for T-cell priming and lymphocyte expansion18.
the tumours of animals treated with man-NP MHCI-ag/man-NP The TLR synergy has been reported previously38. The co-delivery
MHCII-ag + αPD-1/αOX40 + ibrutinib, even when compared to of TLR agonists to a single DC potentiates the efficient maturation
those immunized with the NP (Supplementary Fig. 17). towards balanced TH1- and TH2-type immune responses, which are
High levels of caspase-3 were observed in the tumours of the crucial to control homeostasis within the tumour site and thereby to
groups that received man-NP MHCI-ag/man-NP MHCII-ag + ibru- induce tumour-specific T-cell responses39–41.
tinib, man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40 and Our nanoparticles allowed the multitargeting TLR co-stimu-
αPD-1/αOX40 + ibrutinib, although only man-NP MHCI-ag/man- latory effect by entrapping both CpG and MPLA within the same
NP MHCII-ag  + αPD-1/αOX40 and αPD-1/αOX40 + ibrutinib polymeric matrix, in contrast to the administration of the free com-
strongly inhibited the tumour growth and significantly prolonged pounds. The role of TLR4 agonists (MPLA) on Treg cells is underex-
the survival of the mice (Fig. 5b,e and Supplementary Fig. 16). plored, but it was shown to be essential for the immune-mediated
In groups treated with αPD-1/αOX40 + ibrutinib and those with destruction of cancer cells42. CpG, a TLR9-agonist, however, was
man-NP + ibrutinib, we observed a decrease in MDSC infiltrates shown to suppress the activation of those Treg cells, increase CD4+
from the first to the second end-point. This suggests that ibrutinib T-cell survival and promote the infiltration of CD8+ T  cells into
was restricting the migration of MDSCs into the TME (Fig. 5m and the tumour43. Also, it was previously reported that the intratumour

Fig. 5 | Trivalent combination of mannosylated nanovaccines with ibrutinib and αPD-1/αOX40 strongly restricts melanoma growth, which leads
to long-term survival. a, Timeline (days) of tumour inoculation and treatments. b, Tumour growth curve. Data are presented as mean ± s.e.m. (N = 7
animals). One-way ANOVA. P values correspond to tumour volume at day 20 after the tumour inoculation. c, Percent of EGSRNQDWL (gp100)-specific
CD8+ T cells in the LNs. Data presented as mean ± s.e.m. Unpaired two-tailed t test. d, ELISpot representative images of IFN-γ spot forming cells among
splenocytes after ex vivo restimulation with Melan-A/MART-1 peptides on day 22. Each condition was repeated at least three times. e, Kaplan–Meier
overall survival over time graph for mice inoculated with 3 × 105 RMS cells (N = 13 animals for PBS and N = 15 animals for the remaining groups), replicated
in two independent experiments. f, Individual tumour volume at days 27, 36 and 43 after the tumour inoculation, with the mean ± s.e.m. represented
(N = 13 animals for PBS and N = 15 animals for the remaining groups), replicated in three independent experiments. g–n, Tumour-infiltrating immune cell
populations. Tumours were isolated on the first end-point, day 20 after the tumour inoculation, which corresponds to the day of the first death in the
PBS-treated group, whereas the second end-point was tumour-size matched: day 27 for PBS, ibrutinib only and mannosylated nanovaccines + ibrutinib;
day 35 for αPD-1/αOX40 + ibrutinib and mannosylated nanovaccines + αPD-1/αOX40. The quantification was performed by flow cytometry. During this
period, the tumours of animals treated with mannosylated nanovaccines + αPD-1/αOX40 + ibrutinib were very small and, therefore, these were kept for
the immunohistochemistry analysis. Data are presented as mean ± s.d., N ≥ 3 animals. Unpaired two-tailed t test.

Nature Nanotechnology | www.nature.com/naturenanotechnology


Articles NATure NAnOTeCHnOlOgy

a b 1,500

αPD-1/αOX40 1,250

Tumour volume (mm3)


0 7 10 14 17 21 24 28 31 1,000 P = 0.0060 P = 0.0058

750
3 × 105 Nanovaccine
500 P = 0.038
RMS cells
Ibrutinib
250

0
5 10 15 20 25 30 35 40 45 50 55
PBS Ibrutinib
Time (days after tumour inoculation)
αPD-1/αOX40 + ibrutinib Man-NP MHCI-ag/man-NP MHCII-ag + ibrutinib

Man-NP MHCI-ag/man-NP MHCII-ag Man-NP MHCI-ag/man-NP MHCII-ag


+ αPD-1/αOX40 + αPD-1/αOX40 + ibrutinib

c 15 P < 0.001 d
(among CD3+ CD8+ T cells in the LN)
% EGSRNQDWL-specific T cells

PBS

10
MHCI-ag/MHCII-ag/
CpG/MPLA (free)
+ αPD-1/αOX40
5 + ibrutinib

Man-NP MHCI-ag/
Man-NPMHCII-ag/
+ αPD-1/αOX40
0
+ ibrutinib
PBS
gp100 MHCI-ag/MHCII-ag/CpG/MPLA (free)
+ αPD-1/αOX40 + ibrutinib
Man-NP gp100 MHCI-ag/man-NP gp100 MHCII-ag
+ αPD-1/αOX40 + ibrutinib

e 100
f
3,500
Tumour volume (mm3)

80 3,000
P = 0.0001
Survival (%)

2,500
60 P = 0.0256
2,000
40 1,500
P = 0.0003
1,000
20
P = 0.0001 P = 0.0001 500
0 0
10 15 20 25 30 35 40 45 50 55 60 65 36 43
27
Time (days after tumour inoculation) Time (days after tumour inoculation)

g P = 0.001 h i
P < 0.001 P = 0.002
P = 0.001
P < 0.001 P = 0.002
P = 0.002
250 P < 0.001 40 P < 0.001
100
P < 0.001

P = 0.004
tumour volume (mm3)

P < 0.001 P < 0.001 P = 0.002


tumour volume (mm3)
tumour volume (mm3)

P < 0.001 P < 0.001 P = 0.001

200 P = 0.01 P = 0.01 P < 0.001


80
CD8+ (×10) per

CD4+ (×10) per


CD3+ (×10) per

30
150 60
P < 0.001 20 P = 0.003
100 P = 0.03 P = 0.03 P = 0.03
40
P = 0.01
P = 0.02 P = 0.02 P < 0.001
10
50 20 P = 0.008 P = 0.009

0 0 0
Day 20 Final end-point Day 20 Final end-point Day 20 Final end-point

j P = 0.008 k
P = 0.007
P = 0.03
4 P = 0.04 P = 0.03 P = 0.007 50 P = 0.048 P = 0.045
tumour volume (mm3)

40 PBS
3
Treg (×10) per

CD8+: Treg

30 Ibrutinib
2
20 αPD-1/αOX40 + ibrutinib

1 Man-NP MHCI-ag/man-NP MHCII-ag + ibrutinib


10
Man-NP MHCI-ag/man-NP MHCII-ag + αPD-1/αOX40
0 0
Day 20 Final end-point Day 20 Final end-point

l m P = 0.004 P < 0.001


n
P = 0.001
P = 0.015 P = 0.002
P < 0.001
20 P < 0.001 60 60
P = 0.03 P = 0.005 P < 0.001 P = 0.004 P = 0.002
P = 0.007 P < 0.001 P = 0.006 P = 0.03
(% of single cells)

P < 0.001
(% of single cells)
(% of single cells)

15 P < 0.001
Macrophages

P = 0.02 P = 0.017
40 40
MDSCs
APC

10
20 20
5

0 0 0
Day 20 Final end-point Day 20 Final end-point Day 20 Final end-point

Nature Nanotechnology | www.nature.com/naturenanotechnology


NATure NAnOTeCHnOlOgy Articles

Skin

αPD-1 Ibrutinib Tumour


Nanovaccine
αOX40
Inhibition MDSC

Stimulation

Activation
and maturation
DC CD8+ T cell

PDL1 MHC

Migration and TCR Cytotoxic


priming T-cell activity
LN
against tumour cells
CD4+ T cell

Modulation of cytokine
Mature DC
MHC gradient

OX40
CD4+ T cells PD-1 Increased
Migration T-cell proliferation
and expansion

CD8+ T cells

Fig. 6 | Proposed model for the trivalent therapeutic strategy that combines mannosylated nanovaccines with ibrutinib and αPD-1/αOX40.
TCR, T-cell receptor.

administration of CpG, in combination with αOX40, induced a and CD4+ T-cell proliferation, which increased the secretion of
superior antitumour effect in melanoma compared to each of these cytokines essential for the expansion of CD8+ T cells while traffick-
agonists alone44. Our nanovaccine constitutes an alternative for the ing into the TME.
targeted delivery of CpG oligonucleotide, as our ex vivo and in vivo Overall, the antitumour effect obtained with the trivalent treat-
studies support the successful protection of CpG, once entrapped ment resulted from the modulation of immune-cell activity within
within our nanovaccines’ polymeric matrix. TME, rather than from the direct effect on tumour cells. It led to
Clinical studies that combine antitumour cellular vaccines with DC recruitment and differentiation and reshaped the TME cellular
immune checkpoint therapy have had promising outcomes in pan- plasticity (Fig. 6).
creatic cancer45, colorectal cancer46, prostate cancer47 and mela- Although each of these components had modest results as
noma48. Our therapeutic strategy could offer a clinical alternative monotherapy, their combination resulted in robust and widespread
to those therapies with autologous DCs, which are associated with complementary outcomes. These results provide a strong basis to
complex, long and expensive procedures49. devise novel regimens and combination therapies for solid tumours,
Our results report the synergism of mannosylated antitu- and unravel the full potential of particulate mannosylated cancer
mour nanovaccines with ibrutinib and PD-1/OX40 immune nanovaccines.
checkpoint therapy, which was validated in two tumour-bear-
ing mouse models, with a restricted melanoma growth and a Online content
long-term survival. Any methods, additional references, Nature Research reporting
Of note, this strong therapeutic synergism was only obtained summaries, source data, statements of code and data availability and
when mannose was grafted on the surface of the nanovaccines. associated accession codes are available at https://doi.org/10.1038/
Therefore, the mannosylation of our nanovaccine was crucial s41565-019-0512-0.
for the DC-mediated antigen presentation and the activation of
tumour-antigen-specific T  cells. Recently, ovalbumin-conjugated Received: 24 April 2018; Accepted: 18 June 2019;
mannosylated polymers showed increased DC activation in Published: xx xx xxxx
malaria50. Also, αPD-1/αOX40 + ibrutinib contributed to a homeo-
stasis in the TME ideal for antitumour vaccination. By depleting
References
MDSCs, ibrutinib revealed the full potential of our mannosylated 1. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1
nanovaccines by enhancing the T-cell responses at the TME, which antibody in cancer. New Engl. J. Med. 366, 2443–2454 (2012).
rendered them available for αPD-/αOX40 immune checkpoint 2. Hodi, F. S. et al. Improved survival with ipilimumab in patients with
therapy. The addition of ibrutinib also improved DC maturation metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).

Nature Nanotechnology | www.nature.com/naturenanotechnology


Articles NATure NAnOTeCHnOlOgy
3. Gramaglia, I. et al. The OX40 costimulatory receptor determines the 31. Shedlock, D. J. & Shen, H. Requirement for CD4 T cell help in generating
development of CD4 memory by regulating primary clonal expansion. functional CD8 T cell memory. Science 300, 337–339 (2003).
J. Immunol. 165, 3043–3050 (2000). 32. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators
4. Arch, R. H. & Thompson, C. B. 4-1BB and Ox40 are members of a tumor of the immune system. Nat. Rev. Immunol. 9, 162–174 (2009).
necrosis factor (TNF)-nerve growth factor receptor subfamily that bind TNF 33. Nagaraj, S., Schrum, A. G., Cho, H. I., Celis, E. & Gabrilovich, D. I.
receptor-associated factors and activate nuclear factor kappaB. Mol. Cell Biol. Mechanism of T cell tolerance induced by myeloid-derived suppressor cells.
18, 558–565 (1998). J. Immunol. 184, 3106–3116 (2010).
5. Aspeslagh, S. et al. Rationale for anti-OX40 cancer immunotherapy. 34. Sagiv-Barfi, I. et al. Therapeutic antitumor immunity by checkpoint blockade
Eur. J. Cancer 52, 50–66 (2016). is enhanced by ibrutinib, an inhibitor of both BTK and ITK. Proc. Natl Acad.
6. Sarff, M. et al. OX40 (CD134) expression in sentinel lymph nodes Sci. USA 112, E966–E972 (2015).
correlates with prognostic features of primary melanomas. Am. J. Surg. 195, 35. Swart, M., Verbrugge, I. & Beltman, J. B. Combination approaches
621–625 (2008). with immune-checkpoint blockade in cancer therapy. Front. Oncol. 6,
7. Vetto, J. T. et al. Presence of the T-cell activation marker OX-40 on tumor 233 (2016).
infiltrating lymphocytes and draining lymph node cells from patients with 36. Guo, Z. et al. PD-1 blockade and OX40 triggering synergistically protects
melanoma and head and neck cancers. Am. J. Surg. 174, 258–265 (1997). against tumor growth in a murine model of ovarian cancer. PLoS ONE 9,
8. Spranger, S. et al. Mechanism of tumor rejection with doublets of CTLA-4, e89350 (2014).
PD-1/PD-L1, or IDO blockade involves restored IL-2 production and 37. Woods, D. M., Ramakrishnan, R., Sodré, A. L., Berglund, A. & Weber, J.
proliferation of CD8+ T cells directly within the tumor microenvironment. Abstract A067: PD-1 blockade enhances OX40 expression on regulatory
J. Immunother. Cancer 2, 3 (2014). T-cells and decreases suppressive function through induction of phospho-
9. Gajewski, T. F. The next hurdle in cancer immunotherapy: overcoming STAT3 signaling. Cancer Immunol. Res. 4, A067–A067 (2016).
the non-T-cell-inflamed tumor microenvironment. Semin. Oncol. 42, 38. Zhu, Q. et al. Using 3 TLR ligands as a combination adjuvant induces
663–671 (2015). qualitative changes in T cell responses needed for antiviral protection in
10. Minn, A. J. & Wherry, E. J. Combination cancer therapies with immune mice. J. Clin. Invest. 120, 607–616 (2010).
checkpoint blockade: convergence on interferon signaling. Cell 165, 39. Chen, L. & Flies, D. B. Molecular mechanisms of T cell co-stimulation and
272–275 (2016). co-inhibition. Nat. Rev. Immunol. 13, 227–242 (2013).
11. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive 40. Hanahan, D. & Coussens, L. M. Accessories to the crime: functions of cells
immune resistance. Nature 515, 568–571 (2014). recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).
12. van Kooyk, Y. C-type lectins on dendritic cells: key modulators for the 41. Burkholder, B. et al. Tumor-induced perturbations of cytokines and immune
induction of immune responses. Biochem Soc. Trans. 36, 1478–1481 (2008). cell networks. Biochim. Biophys. Acta 1845, 182–201 (2014).
13. Kodumudi, K. N., Weber, A., Sarnaik, A. A. & Pilon-Thomas, S. Blockade of 42. Fang, H. et al. TLR4 is essential for dendritic cell activation and anti-tumor
myeloid-derived suppressor cells after induction of lymphopenia improves T-cell response enhancement by DAMPs released from chemically stressed
adoptive T cell therapy in a murine model of melanoma. J. Immunol. 189, cancer cells. Cell. Mol. Immunol. 11, 150–159 (2013).
5147–5154 (2012). 43. Beyersdorf, N., Kerkau, T. & Hunig, T. CD28 co-stimulation in T-cell
14. Meyer, C. et al. Frequencies of circulating MDSC correlate with clinical homeostasis: a recent perspective. Immunotargets Ther. 4, 111–122 (2015).
outcome of melanoma patients treated with ipilimumab. Cancer Immunol. 44. Sagiv-Barfi, I. et al. Eradication of spontaneous malignancy by local
Immunother. 63, 247–257 (2014). immunotherapy. Sci. Transl. Med. 10, eaan4488 (2018).
15. Stiff, A. et al. Myeloid-derived suppressor cells express Bruton’s tyrosine 45. Maher, E. A., Mietz, J., Arteaga, C. L., DePinho, R. A. & Mohla, S. Brain
kinase and can be depleted in tumor-bearing hosts by ibrutinib treatment. metastasis: opportunities in basic and translational research. Cancer Res. 69,
Cancer Res. 76, 2125–2136 (2016). 6015–6020 (2009).
16. Natarajan, G. et al. A Tec kinase BTK inhibitor ibrutinib promotes maturation 46. Santarelli, J. G., Sarkissian, V., Hou, L. C., Veeravagu, A. & Tse, V. Molecular
and activation of dendritic cells. Oncoimmunology 5, e115159 (2016). events of brain metastasis. Neurosurg. Focus 22, E1 (2007).
17. Alonso-Sande, M. et al. Development of PLGA–mannosamine nanoparticles 47. van den Eertwegh, A. J. et al. Combined immunotherapy with granulocyte-
as oral protein carriers. Biomacromolecules 14, 4046–4052 (2013). macrophage colony-stimulating factor-transduced allogeneic prostate cancer
18. Silva, J. M. et al. In vivo delivery of peptides and Toll-like receptor ligands by cells and ipilimumab in patients with metastatic castration-resistant prostate
mannose-functionalized polymeric nanoparticles induces prophylactic and cancer: a phase 1 dose-escalation trial. Lancet Oncol. 13, 509–517 (2012).
therapeutic anti-tumor immune responses in a melanoma model. J. Control 48. Hodi, F. S. et al. Ipilimumab plus sargramostim vs ipilimumab alone for
Release 198, 91–103 (2015). treatment of metastatic melanoma: a randomized clinical trial. J. Am. Med.
19. Wang, X., Ramstrom, O. & Yan, M. Dynamic light scattering as an Assoc. 312, 1744–1753 (2014).
efficient tool to study glyconanoparticle–lectin interactions. Analyst 136, 49. Kaiser, A. D. et al. Towards a commercial process for the manufacture of
4174–4178 (2011). genetically modified T cells for therapy. Cancer Gene Ther. 22, 72–78 (2015).
20. De Koker, S. et al. Engineering polymer hydrogel nanoparticles for lymph 50. Wilson, D. S. et al. Antigens reversibly conjugated to a polymeric glyco-
node-targeted delivery. Angew. Chem. Int. Ed. 55, 1334–1339 (2016). adjuvant induce protective humoral and cellular immunity. Nat. Mater. 18,
21. Azzi, J. et al. Targeted delivery of immunomodulators to lymph nodes. Cell 175–185 (2019).
Rep. 15, 1202–1213 (2016).
22. Seliger, B., Ruiz-Cabello, F. & Garrido, F. IFN inducibility of major Acknowledgements
histocompatibility antigens in tumors. Adv. Cancer Res. 101, 249–276 (2008). The MultiNano@MBM project was supported by The Israeli Ministry of Health and
23. Dranoff, G. et al. Vaccination with irradiated tumor cells engineered to The Fundação para a Ciência e Tecnologia-Ministério da Ciência, Tecnologia e Ensino
secrete murine granulocyte-macrophage colony-stimulating factor stimulates Superior (FCT-MCTES) under the framework of EuroNanoMed-II (ENMed/0051/2016
potent, specific, and long-lasting anti-tumor immunity. Proc. Natl Acad. Sci. to H.F.F., R.S.-F. and S.J.). R.S.-F. thanks the European Research Council (ERC)
USA 90, 3539–3543 (1993). Consolidator Grant Agreement no. [617445]-PolyDorm and ERC Advanced Grant
24. Kowalczyk, D. W. et al. Vaccine-induced CD8+ T cells eliminate tumors by a Agreement no. [835227] - 3DBrainStrom, The Israel Science Foundation (Grant
two-staged attack. Cancer Gene Ther. 10, 870–878 (2003). nos. 918/14 and 1969/18), The Melanoma Research Alliance (the Saban Family
25. Pasare, C. & Medzhitov, R. Toll-like receptors: balancing host resistance with Foundation–MRA Team Science Award to R.S.-F. and N.E., and Established
immune tolerance. Curr. Opin. Immunol. 15, 677–682 (2003). Investigator Award to R.S.-F.) and the Israel Cancer Research Fund (ICRF). J.C.,
26. Scheller, J., Chalaris, A., Schmidt-Arras, D. & Rose-John, S. The pro- and A.I.M., C.P., E.Z. and L.I.F.M. are supported by the FCT-MCTES (Fellowships SFRH/
anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. BD/87150/2012, PD/BD/113959/2015, SFRH/BD/87591/2012, SFRH/BD/78480/2011
Acta 1813, 878–888 (2011). and SFRH/BPD/94111/2013, respectively). This project has received funding from
27. Fridlender, Z. G. et al. CCL2 blockade augments cancer immunotherapy. European Structural & Investment Funds through the COMPETE Programme and
Cancer Res. 70, 109–118 (2010). from National Funds through FCT under the Programme grant SAICTPAC/0019/2015
28. Tsui, P. et al. Generation, characterization and biological activity of CCL2 (H.F.F.). We thank E. Haimov and B. Redko from the Blavatnik Center for Drug
(MCP-1/JE) and CCL12 (MCP-5) specific antibodies. Hum. Antibodies 16, Discovery at the Tel Aviv University for their professional and technical assistance with
117–125 (2007). the peptide synthesis.
29. Phan, G. Q. et al. Immunization of patients with metastatic melanoma using
both class I- and class II-restricted peptides from melanoma-associated
antigens. J. Immunother. 26, 349–356 (2003). Author contributions
30. Slingluff, C. L. Jr. et al. A randomized phase II trial of multiepitope J.C. and A.S. synthesized the nanovaccines and performed the in vitro and the animal
vaccination with melanoma peptides for cytotoxic T cells and helper T cells studies; J.C. synthesized the man-PLGA polymers and carried out the physicochemical
for patients with metastatic melanoma (E1602). Clin. Cancer Res. 19, characterization of the nanovaccines; R.K. helped with the nanovaccine formulation;
4228–4238 (2013). A.S., S.P. and R.K. performed the ELISPOT experiments; C.P. and A.I.M. analysed

Nature Nanotechnology | www.nature.com/naturenanotechnology


NATure NAnOTeCHnOlOgy Articles
the flow cytometry experiments; E.Y. and S.P. performed the immunohistochemistry Additional information
experiments; J.C. and A.I.M. carried out the tetramer assay; L.I.F.M. and E.Z. helped Supplementary information is available for this paper at https://doi.org/10.1038/
with the animal experiments; A.S.V. performed the AFM experiments; H.D. and N.E. s41565-019-0512-0.
contributed with the Ret cells; P.M.P.G. advised on the polymer synthesis; S.J. critically
Reprints and permissions information is available at www.nature.com/reprints.
advised and contributed in interpreting the results. J.C., A.S., R.S.-F. and H.F.F. conceived
and designed the experiments, analysed the data and wrote and revised the manuscript. Correspondence and requests for materials should be addressed to R.S. or H.F.F.
R.S.-F. and H.F.F. were in charge of the overall direction and planning of this study, and Peer review information: Nature Nanotechnology thanks Walter Storkus and other,
all the authors commented on the manuscript. anonymous, reviewer(s) for their contribution to the peer review of this work.
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
Competing interests published maps and institutional affiliations.
The authors declare no competing interests. © The Author(s), under exclusive licence to Springer Nature Limited 2019

Nature Nanotechnology | www.nature.com/naturenanotechnology


Articles NATure NAnOTeCHnOlOgy

Methods the particles was measured by laser Doppler velocimetry in combination with
Materials. PLA (2,000 Da) with a weight-averaged molecular mass (Mw) of phase analysis light scattering with the same equipment. Particles were diluted in
2,000 was purchased from PolySciences, Inc. The tumour-associated peptides ultrapure water and the electrophoretic mobility was determined at 25 °C with the
MHCI-restricted Melan-A:26-35(L27), ELAGIGILTV, (MHCI-ag), MHCII- Helmholtz–Smoluchowski model.
restricted Melan-A:51-73(RR-23) and RNGYRALMDKSLHVGTQCALTRR
(MHCII-ag) were purchased from GeneCust. Alternatively, the synthesis and Particle morphology. AFM. Particles were diluted at 5 mg ml–1 in ultrapure water.
purification of these Melan-A peptides was performed by the Blavatnik Center A drop of the sample was placed onto freshly cleaved mica for 20 min and dried
for Drug Discovery at the Tel Aviv University. The synthesis and purification with pure N2. Samples were analysed by AFM in tapping mode in air at room
of the gp100 peptides (MHCI-restricted gp100:25-33 (EGSRNQDWL) and temperature using a Nanoscope IIIa Multimode (Digital Instruments/Veeco) atomic
MHCII-restricted gp100:44-59 (WNRQLYPEWTEAQRLD) was performed force microscope and etched silicon tips (~300 kHz) at a scan rate of ~1.6 Hz.
by the Blavatnik Center for Drug Discovery at Tel Aviv University. The H-2Db-
restricted EGSRNQDWL PE-labelled tetramer was supplied by Quimigen S.L. CpG Scanning electron microscopy. Particles were diluted in trehalose 5% (m/v) and fast
ODN 1826 (TCCATGACGTTCCTGACGTT) was purchased from InvivoGen. frozen at −80 °C for 2 h. Samples were dried under vacuum, first at −20 °C for 14 h
MEM α, nucleosides and ascorbic acid were purchased from Invitrogen. RPMI and then at 20 °C for 2 h. Dried specimens were coated with gold on a Peltier cold-
1640, heat-inactivated fetal bovine serum, trypsin EDTA 0.05%, penicillin stage sputter coater and examined using a FEI Quanta 200 FEG ESEM Phillips 500
(10,000 unit ml–1) and streptomycin (10,000 μg ml–1), sodium pyruvate (100 mM), scanning electron microscope at a 5 kV accelerating voltage.
GM-CSF recombinant mouse protein (5 ng ml–1), HEPES buffer (1 M), ACK lysing
buffer, paraformaldehyde (PFA) 4% (v/v), wheat germ agglutinin, Alexa Fluor Transmission electron microscopy. Particles were diluted in PBS and placed on a
633 Conjugate, Hoechst 33342 and AlamarBlue reagent were purchased from carbon-coated copper grid and dried. The samples were analysed with a Philips
Thermo Fisher Scientific. The Proteome Profiler Mouse XL Cytokine Array Kit was CM 120 Bio-Twin transmission electron microscope.
purchased from R&D Systems, Inc. Matrigel Matrix (catalogue no. 356231) was
purchased from BD Biosciences-Europe. IncuCyte Caspase-3/7 Apoptosis Assay EE and LC of antigens and immune potentiators. Supernatants collected from
Reagent was acquired from Biological Industries Israel Beit-Haemek Ltd PLGA the centrifugations were used for an indirect quantification of the entrapped
(Resomer 503H, Mw 24,000–38,000), mannosamine·HCl, dimethylformamide, antigens and immune potentiators. The EE (equation (1)) and LC (µg mg–1)
4-dimethylaminopyridine, N,N′-dicyclohexylcarbodiimide, methanol, anhydrous (equation (2)) of Melan-A/MART-1(26-35(A27L)) and Melan-A/MART-1(51-
sodium sulfate, Conc A, FITC-labelled Conc A from Canavalia ensiformis (jack 73) were determined with FAM-labelled Melan-A/MART-1(26-35(A27L)) and
bean), Type IV lyophilized powder, lipid A monophosphoryl from Salmonella Melan-A/MART-1(51-73), respectively. The relative fluorescence units were
enterica serotype Minnesota Re 595 (Re mutant) (MPLA), bovine serum albumin, measured with a SpectraMax M5e plate reader (Molecular Devices) at 498/518 nm
TPGS, PVA (Mw 13,000–23,000 Da, 99% hydrolysed), dichloromethane (DCM) and for the excitation/emission wavelengths. The amount of CpG in the supernatant
chloroform-d were purchased from Sigma Aldrich. PD-1 and OX40 monoclonal was determined with an Oligreen ssDNA quantitation kit18 with the relative
antibodies were acquired from Bio X Cell. Rabbit monoclonal anti-caspase 3 was fluorescence units measured using the fluorometer at 485 nm excitation and
purchased from Epitomics. Antibodies anti-mouse CD4 (clone CK1.5), CD8α 530 nm emission wavelengths.
(clone 53-6.7) and DAPI were acquired from BioLegend. Streptavidin−horseradish
peroxidase conjugate was purchased from Histostain, Life Technologies. EE (%) =
Fluorochrome-labelled antibodies for flow cytometry were purchased from initial amount of agent−amount of agent in the supernatant
(1)
Miltenyi Biotech, unless otherwise specified. ELISpot kit was purchased from initial amount of agent
R&D Systems Inc.
× 100
Synthesis and characterization of mannose-PLGA polymer. Mannose-PLGA
(man-PLGA) was synthesized from PLGA (Resomer 503H). The carboxylic acid LC(μg mg −1)
terminal groups of PLGA were modified with mannosamine under a nitrogen initial amount of agent−amount of agent in the supernatant (2)
atmosphere in mild conditions17. Briefly, mannosamine·HCl (10.8 mg, 0.05 mmol, =
total amount of polymer
3.5 equiv.) and 4-dimethylaminopyridine (6.5 mg, 0.05 mmol, 4 equiv.) were
added to 4 ml of dimethylformamide. The mixture was stirred for 10 min at room
temperature to achieve complete dissolution of mannosamine. PLGA (Resomer Mannose detection on the particle surfaces by the lectin recognition assay. The
503H) (400 mg, 0.0133 mmol, 1 equiv.) was added to the previous solution and details are described in the Supplementary Methods.
stirred for 10 min at room temperature. N,N′-Dicyclohexylcarbodiimide (5.5 mg,
0.027 mmol, 2 equiv.) was added to induce the reaction between PLGA and Cell lines. The details are described in the Supplementary Methods.
mannosamine. The reaction was allowed to stir for 48 h at room temperature under
an argon atmosphere. The polymer was precipitated with water and recovered In vitro cell viability in the presence of NP or man-NP. The details are described
by centrifugation. Man-PLGA was dissolved in DCM and dried over anhydrous in the Supplementary Methods.
sodium sulfate. The solution was filtered. DCM was evaporated through rotary
evaporation. Methanol was added to precipitate the polymer and wash the reaction Haemolysis assay. The details are described in the Supplementary Methods.
crude. Man-PLGA was dissolved in DCM and the precipitation was repeated.
Finally, man-PLGA was dried under vacuum overnight and weighed after 24 h In vitro particle internalization by DCs. JAW SII DCs (5 × 104 cells per well) were
(186.7 mg, η = 47%). seeded in 96-well plates and incubated overnight. Cells were then incubated with
rhodamine-grafted NP or man-NP (500 μg ml–1) for 4, 12 and 24 h. Cells were then
NMR spectroscopy. Details are described in the Supplementary Methods. washed with Dulbecco’s PBS and resuspended in the flow cytometry buffer. Non-
treated cells and non-labelled NP were used as the negative controls. An excess
Synthesis of NP and man-NP. PLGA/PLA NP was formulated by the double of soluble mannose (5 mM) was added to the medium as a control to confirm its
emulsion–solvent evaporation method17. A PLGA/PLA (2:8) blend was dissolved ability to compete with man-NP for MR/CD206 at the DC surface. The individual
in DCM at 50 mg ml–1. MPLA (100 µg) was added to the DCM polymer solution. fluorescence JAW SII DCs was collected for each sample using an LSR Fortessa
A 10% (m/v) PVA aqueous solution (100 µl) that contained CpG at 0.5 mg ml–1 cytometer (BD Biosciences) by Facs Diva, and analysed with FlowJo software
and Melan-A/MART-1(26-35(A27L), Melan-A/MART-1(51–73), gp100(25–33) version 7.6.5 for Microsoft (TreeStar).
or gp100(44–59) at 5 mg ml–1 was added to DCM. For the empty NP, 100 µl of The eight-well Ibidi µ-Slide microscopy chambers were incubated with 300 µl
10% (m/v) PVA aqueous solution was added. The mixture was emulsified with a of fibronectin (10 µg ml–1) per well during 30 min in a humidified incubator with
microprobe ultrasonic processor for 15 s at 20% amplitude. A 2.5% (m/v) TPGS 5% CO2 at 37 °C. After discarding the volume of fibronectin, JAW SII cells (5 × 104
aqueous solution (400 µl) was added and the second emulsion was formed using cells per well) were seeded in eight-well Ibidi µ-Slide microscopy chambers for
the same conditions. The double emulsion was added dropwise into a 0.25% (m/v) 6 h in a humidified incubator with 5% CO2 at 37 °C. The cells were incubated with
PVA aqueous solution and stirred for 1 h at room temperature. Particle suspension rhodamine-grafted NP or man-NP (500 μg ml–1) for 3 h. Non-treated cells and
was collected by centrifugation at 20,000g for 45 min, 4 °C (SORVALL RC-5B PLUS non-labelled empty NP and empty man-NP were used as the negative controls.
Superspeed centrifuge). Particles were washed with ultrapure water, collected by Cells were washed and fixed at room temperature in 4% PFA that contained
centrifugation and finally resuspended in PBS or ultrapure water. Man-NP were Hoechst 332 at 2 µg ml–1 and wheat germ agglutinin Alexa Fluor 633 at 5 µg ml–1
prepared as previously described with a man-PLGA/PLA (2:8) blend instead. Cy5.5- to stain the nucleus and the cell membrane, respectively. Each well was washed
labelled NP and man-NP were synthesized by adding 0.5 μg to the polymer blend. three times with PBS and 100 µl of fluoromount were added to each one. Particle
internalization was observed by confocal microscopy using a Zeiss LSM 710 with
Size distribution and ζ potential measurements. Particle size was measured ×63 amplification in oil. Images were processed with ImageJ software. Three-
by dynamic light scattering with a Malvern Nano ZS (Malvern Instruments). dimensional projection images were obtained from 0.4 μm Z-stacks and processed
The Z-average size was determined by cumulative analysis. The ζ potential of using the Leica Application Suite-Advanced Fluorescence software.

Nature Nanotechnology | www.nature.com/naturenanotechnology


NATure NAnOTeCHnOlOgy Articles
Animal studies. All the animal procedures were performed in compliance (1:1) were inoculated subdermally on the right dorsal region as reported before51.
with Sackler Faculty of Medicine, Tel Aviv University guidelines and with the For the intervention studies, the amount of murine RMS and Ret-melanoma
Portuguese competent authority for animal protection, Direcção Geral de cells that were inoculated on day 0 was 3 × 105. For the intervention study on the
Alimentação e Veterinária. The protocols were approved by the Institutional second melanoma model, 100 µl of saline with 1.5 × 105 B16-F10 cells in suspension
Animal Care and Use Committee and performed in accordance with National were inoculated subdermally in the right flank. For both tumour models, mice
Institutes of Health guidelines. were anaesthetized with ketamine (100 mg kg–1) and xylazine (12 mg kg–1). The
Male C57BL/6J mice (8 weeks old) were purchased from Envigo Ltd or Charles right dorsal area was treated with depilatory cream before the injection. αPD-1
River and housed in the animal facility of Tel Aviv University or at the Faculty and αOX40 were administered intraperitoneally at 10 mg kg–1. Ibrutinib was also
of Pharmacy, University of Lisbon. The number of animals in each group was administered intraperitoneally at 6 mg kg–1. The tumour size was measured every
determined according to previous studies18. 3 days with a calliper. The tumour volume was determined by X2Y0.5 (X, small
Mice body weight change was monitored three times per week. Mice were diameter; Y, large diameter) and body weight was monitored twice a week.
euthanized according to ethical protocols when they showed signs of distress
or with rapid weight loss (above 10% within a few days or 20% from the initial Immunohistochemistry. The details are described in the Supplementary Methods.
weight). Tumour-bearing mice were euthanized in the case that the tumour
size exceeded 2,000 mm3 or if the tumour was necrotic or ulcerative. Mice were Flow cytometric analysis of tumour-infiltrated immune populations. RMS
perfused intracardially with PBS immediately after euthanasia, the tumours were tumours were isolated from the animals directly after euthanasia. Tumour single-cell
dissected and incubated in 4% PFA and collected for histology. suspensions were obtained by mechanical disruption of the tissues and enzymatic
digestion in RPMI medium with 0.5% BSA, 0.1% collagenase type II (LS004177,
In vivo study of man-NP internalization by DC in draining LNs. Male Worthington), 0.1% dispase (LS02109, Worthington) and DNase (LS002007,
C57BL/6, 8-week-old mice (n = 3 per group) were injected with one of the Cy5.5 Worthington) for 1 h at 37 °C. After digestion, the suspension was filtered through a
fluorescently-labelled plain and antigen-loaded nanoparticle formulations (2 mg 70 μm filter (BD Biosciences) to remove the debris. For the RMS cells, the obtained
of nanoparticle per mouse) into the right flank by s.c. hock immunization. The single-cell suspension was then stained with fluorochrome-labelled antibodies and
left non-injected flank served as a negative control. All the formulations contained analysed using an LSR Fortessa (BD Biosciences) and FlowJo software (TreeStar).
MART-1 antigens and TLR ligands. Popliteal and inguinal LNs were harvested 16 h Intracellular staining was performed with the Inside stain kit (Miltenyi Biotec,
postimmunization. A single-cell suspension was stained with fluorescent-labelled catalogue no. 130-090-477) according to the manufacturer’s protocol.
anti-mouse antibodies against CD11c, MHCII (I-Ab), MHCI (H-2Kb), CD80 Details of the antibodies used in each panel are given in the Supplementary
and CD86 for 20 min at 4 °C. Samples were acquired with an LSR II Fortessa flow Information.
cytometer (BD Bioscience) and analysed with FlowJo software (TreeStar).
Functional assessment of T cells. For the ELISpot assay, on day 0, 50 µl of a cell
Immunization of animals with TAAs. For immunization studies, suspension that contained 4.5 × 105 murine RMS cells was inoculated subdermally.
8-week-old C57BL/6J male mice were randomized into eight groups (N = 6) Mice were randomized into 5 groups, as reported in Supplementary Table 5, and
(Supplementary Table 3). treated according to the schedule used for the intervention combinatorial
Treatments (100 µl) were injected into each mouse by hock immunization studies (Fig. 5a).
via s.c. injection proximal to popliteal LNs. A half dose (50 μl) was injected On day 21, mice were euthanized, the spleens harvested and the splenocytes
into the right side and the other half into the left side. When the two peptide isolated. Splenocytes were seeded at 2 × 105 cells per well in 96-well plates coated
antigens Melan-A/MART-1(26-35(A27L)) and Melan-A/MART-1(51-73) were with IFN-γ antibody (R&D Systems Inc.) and incubated for 20 h with 1 mg ml–1
administered to the same mouse (groups 4, 6 and 8), 25 µl of each treatment of Melan-A/MART-1 MHCI-ag peptide. The secreted and captured IFN-γ was
was administered on each side. Each dose contained 100 µg of antigen (50 µg of subsequently detected using a biotinylated antibody specific for IFN-γ and
Melan-A/MART-1(26-35(A27L)) and 50 µg of Melan-A/MART-1(51-73) when two an alkaline-phosphatase conjugated to streptavidin. After the addition of the
antigens were used) plus 20 µg of CpG and 20 µg of MPLA, either free in solution substrate solution, a blue precipitate formed and appeared as spots at the sites of
or entrapped in 2 mg of particles (20 mg ml–1). cytokine localization. Automated spot quantification was performed using a UV
ImmunoSpot S6 Ultra-V.
Tumour-antigen-specific proliferation of splenocytes from immunized mice. For the tetramer staining assay, male C57BL/6J, 8-week-old mice were s.c.
Splenocytes from whole spleens of treated C57BL/6J mice (N = 6) were harvested immunized into both the right and left inguinal areas twice (once a week) with
and seeded in sterile 60 mm Petri dishes 10 days after the final immunization. 100 µl of man-NP or NP (100 µg of gp100 antigens, 20 µg of CpG and 20 µg of
Splenocytes were seeded for 6 days in complete RPMI medium in the presence of MPLA per mouse, 50 µl in each side), free gp100 antigens with adjuvants CpG and
Melan-A/MART-1(26-35(A27L)) or Melan-A/MART-1(26-35(A27L)) + Melan-A/ MPLA or PBS. Inguinal LNs were harvested 10 days after the second injection,
MART-1(51-73) (0.1 mg ml–1) (for groups 4, 6 and 8) and CD28 (2 μg ml–1). homogenized in a single-cell suspension and plated in a 96-well plate for staining.
First, the peptide–major histocompatibility complex tetramer tagged with PE
Splenocyte cytokine and chemokine secretion from immunized mice. A (H-2Db-restricted EGSRNQDWL PE-labelled tetramer (Quimigen S.L.)) was
membrane-based sandwich immunoassay was performed according to the added to the single-cell suspension, including FcR blocking, following the supplier
manufacturer protocol of Proteome Profiler Mouse Cytokine Array Panel A Kit instructions. After 30 min of incubation at 4 °C, the cells were washed to remove
(catalogue no. R&D-ARY0068, R&D Systems Inc.). Splenocyte’s culture media unbound tetramers and centrifuged at 1,300 r.p.m. for 10 min at 10 °C. The cells were
from each group (Supplementary Table 3) were pooled (n = 6) and concentrated resuspended in an ice-cold sorter buffer and plated in 96-well plates. After adding a
in concentration tubes (Amicon). Each sample was applied on a nitrocellulose mix of the antibodies CD3-APC-Cy7 and CD8α-PE-Cy7, the cells were incubated
membrane containing capture antibodies that bind to the specific target protein. for 15 min at 4 °C, protected from light. The cells were washed, centrifuged and
After overnight incubation at 4 °C, streptavidin–horseradish peroxidase and resuspended in 200 µl of ice-cold sorter buffer to determine the percentages of
chemiluminescent reagents were added, and incubation steps were performed tumour antigen-specific CD3+ CD8α+ T cells by fluorescence-activated cell sorting.
according to the protocol provided by the analysis kit. Membranes were exposed
to X-rays for 10 min. The results were detected by a transmission-mode scanner Statistical methods. Data are presented as mean ± s.d. for in vitro assays and as
proportionally to the quantity of analyte and determined by protein array mean ± s.e.m. for ex vivo and in vivo assays. Statistical analyses were performed
analyser software. with the Student’s t-test, one-way and two-way analysis of variance, followed by
a Bonferroni post hoc test or Tukey test for comparison of multiple groups with
Ex vivo immune cell killing assay. Immune cell cytotoxic activity was assessed IBM SPSS Statistics (Version 21, Microsoft). The statistical significance in overall
using an IncuCyte ZOOM live-cell instrument. RMS cells (6 × 103 cells per well) survival was determined with a log-rank test using SigmaPlot software (Systat
were seeded in 96-well plates in complete RMPI without phenol red on the day Software Inc.) and GraphPad Prism 5 or 7 (GraphPad Software, Inc.). P < 0.05 was
before the addition of splenocytes. Prestimulated splenocytes were added to the considered statistically significant.
RMS cells in a 1:100 ratio. IncuCyt Caspase-3/7 Apoptosis Assay Reagent was
Reporting summary. Further information on research design is available in the
diluted in RPMI without phenol red and added to each well to a final concentration
Nature Research Reporting Summary linked to this article.
of 5 µM. Data were collected every 2 h for 12 h. Triton X-100 0.5% (v/v) and cell
culture medium were used as the positive and negative controls, respectively.
Data availability
Tumour inoculation, combination therapies and tumour volume measurements. The authors declare that the data supporting the findings of this study are available
Male C57BL/6J mice (8 weeks old) were randomized into different groups within the paper and its Supplementary Information. Additional data and source
(N = 4–5) according to Supplementary Table 4 for the prophylactic combination files are available from the corresponding authors upon reasonable request.
study. Different schedules were used for the prophylactic (Fig. 3a) and intervention
combinatorial studies without or with the addition of ibrutinib (Figs. 4a and 5a, References
respectively). For the prophylactic study, on day 0, 50 µl of cell suspension that 51. Schwartz, H. et al. Incipient melanoma brain metastases instigate astrogliosis
contained 4.5 × 105 murine RMS cells mixed with growth-factor reduced matrigel and neuroinflammation. Cancer Res. 76, 4359–4371 (2016).

Nature Nanotechnology | www.nature.com/naturenanotechnology


nature research | reporting summary
Corresponding author(s): Ronit Satchi-Fainaro and Helena F. Florindo

Last updated by author(s): Jun 3, 2019

Reporting Summary
Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Research policies, see Authors & Referees and the Editorial Policy Checklist.

Statistics
For all statistical analyses, confirm that the following items are present in the figure legend, table legend, main text, or Methods section.
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
A statement on whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.

A description of all covariates tested


A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistical parameters including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient)
AND variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)

For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.

For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated
Our web collection on statistics for biologists contains articles on many of the points above.

Software and code


Policy information about availability of computer code
Data collection FACS Diva was used to collect data and data was analyzed using FlowJo v10 software; NMR spectra were processed using Bruker NMR
Suite 3.5 and MestReNova 10.0. IncuCyte S3 software for acquiring, viewing and analyzing images of living cells during ex vivo immune
cell killing assay. SEM data was collected using FEI Quanta 200 FEG ESEM Phillips 500 scanning electron microscope at 5 kV accelerating
voltage.TEM data was collected using Philips CM 120 Bio-Twin TEM. ImmunoSpot S6 ULTRA-V Analyzer was used to acquire the ELISPOT
images during the functional assessment of T cells. A fluorescence and white light microscope (Evos FL Auto, life technologies) was used
to acquire images of stained tumor tissues.

Data analysis Analysis of flow cytometry data was performed with FlowJo v10 software. Graph Pad Prism 5.0a and 7 were used for individual tumor
volume at different days; Statistical significance in overall survival was determined with log-rank test using SigmaPlot software (Systat
Software Inc.) and GraphPad Prism 7. Zetasizer Nano software v3.30 for analyses of nano-vaccine size and surface charge. ImageJ
software for confocal analysis and to determine staining intensity of the different markers used in immunohistochemistry. IncuCyte S3
software for analyzing images of living cells during ex vivo immune cell killing assay. Leica Application Suite-Advanced Fluorescence (LAS-
AF) software for the analysis of three-dimensional (3D) projection images obtained by confocal microscopy.
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors/reviewers.
We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information.
October 2018

1
Data

nature research | reporting summary


Policy information about availability of data
All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
- A list of figures that have associated raw data
- A description of any restrictions on data availability

The authors declare that the data supporting the findings of this study are available within the paper and its Supplementary Information. Additional data and source
files are available from the corresponding author upon reasonable request.

Field-specific reporting
Please select the one below that is the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/documents/nr-reporting-summary-flat.pdf

Life sciences study design


All studies must disclose on these points even when the disclosure is negative.
Sample size The number of animals in each group was determined according to previous studies cited in our manuscript. The size of each sample is in
close agreement with those studies already published and with the need for statistical analysis to discuss the degree of differences and
measure the variability of these in vivo data.

Data exclusions No data were excluded from the analyses.

Replication Each group of animals used in the in vivo studies described in this manuscript included between 6-15 animals, depending on the purpose of
the study. Data from the main in vivo studies are presented as mean ± SEM of 3 independent replicates of the same experiment. All replicates
were successful.
FACS analysis of cell suspension prepared from tumor tissues collected from control, immunized or treated animals were performed as mean
± SD of 3 animals/group.
Histology sections of tumor tissues collected from all control and treatment groups are representative of at least 5 independent successful
replicates.

Randomization Prophylactic study: animals were distributed randomly into the control and immunization groups. Each animal received different doses
following established schedules for nano-vaccine, immune checkpoint antibodies and ibrutinib.
Therapeutic studies: animals were inoculated with tumor cell suspensions and after randomly distributed into the control and experimental
groups. Each specific treatment was administered to animals according to established schedules and regimens.

Blinding Investigators were double-blinded during Immunohistochemical staining, data collection and analysis of tumor sections.
During the in vivo experiments designed to evaluate anti-tumor efficacy, animals were inoculated with tumor cells and randomly divided into
control and treatment groups. The schedules and regimens established for each group encompassed prime and booster doses for the nano-
vaccine and several doses of ibrutinib and immune checkpoint antibodies. The investigators were not blinded during these pre-clinical proof-
of-concept studies based on combinatorial schemes.

Reporting for specific materials, systems and methods


We require information from authors about some types of materials, experimental systems and methods used in many studies. Here, indicate whether each material,
system or method listed is relevant to your study. If you are not sure if a list item applies to your research, read the appropriate section before selecting a response.

Materials & experimental systems Methods


n/a Involved in the study n/a Involved in the study
Antibodies ChIP-seq
October 2018

Eukaryotic cell lines Flow cytometry


Palaeontology MRI-based neuroimaging
Animals and other organisms
Human research participants
Clinical data

2
Antibodies

nature research | reporting summary


Antibodies used Antibodies used for flow cytometry:
Lymphocyte panel: CD3ε-APC (Miltenyi Biotec, Cat.# 130-109-240, mouse, clone: REA606, lot: 5190401141 – dilution: 1:10),
CD4-FITC (Miltenyi Biotec, Cat.# 130-109-419, mouse, clone: REA604, lot: 5190401266 – dilution: 1:10), CD8a-PE (Miltenyi
Biotec, Cat.# 130-109-247, mouse, clone: REA60, lot: 5171219075 – dilution: 1:10).

Treg panel: CD3ε-APC (Miltenyi Biotec, Cat.# 130-109-240, mouse, clone: REA606, lot: 5190401141 – dilution: 1:10), CD4-FITC
(Miltenyi Biotec, Cat.# 130-109-419, mouse, clone: REA604, lot: 5190401266 – dilution: 1:10), CD25-PE (Miltenyi Biotec, Cat.#
130-109-051, mouse, clone: REA568, lot: 5180925049 – dilution: 1:10).

Intracellular staining: FoxP3 -Vio515 (FITC), (Miltenyi Biotec, Cat.# 130-111-681, mouse, clone: REA788, lot: 5190401271 –
dilution: 1:50).

Myeloid Panel: CD11c-FITC (Miltenyi Biotec, Cat.# 130-102-466, mouse, clone: N418, lot: 5171219060 – dilution: 1:10), CD11b-
APC-Cy7 (Miltenyi Biotec, Cat.# 130-109-288, mouse, clone: REA592, lot: 5161121319 – 1:10), MHC Class II-Pac. Blue (Miltenyi
Biotec, Cat.# 130-102-145, mouse, clone: M5/114.15.2, lot: 5180925030 – 1:10) Gr-1-APC (Miltenyi Biotec Cat.# 130-112-307,
mouse, clone: REA810, lot: 5190110365 – dilution: 1:50).

Cytokine panel: CD3-PerCP-Cy5.5 (Miltenyi Biotec, Cat.# 130-109-841, mouse, clone: REA641, lot: 5180711267 – 1:10), CD8a-
APC-Cy7 (Miltenyi Biotec, Cat.# 130-109-328, mouse, clone: REA601, lot: 5180808300 – dilution: 1:10); and intracellular staining
of IFN-γ-FITC (Miltenyi Biotec, Cat.# 130-109-768, mouse, clone: REA638, lot: 5171219082 – dilution: 1:10).

Antibodies used for histology:


Rabbit anti-mouse PD-1 (Proteintech, cat. # 18106-1-ap, mouse, polyclonal, lot: 00068675, Dilution: 1:100). Validation data is
available on the manufacturer's website. References: Qiu, J., et al. (2018). "Estradiol Attenuates the Severity of Primary
Toxoplasma gondii Infection-Induced Adverse Pregnancy Outcomes Through the Regulation of Tregs in a Dose-Dependent
Manner." Front Immunol 9: 1102.

Rat anti-mouse PD-L1 (Biolegend, cat. # 124303, mouse, clone: 10F.9G2, lot: B248154, dilution: 1:100). Validation data is
available on the manufacturer's website. References: Grabie N, et al. (2007). Endothelial programmed death-1 ligand 1 (PD-L1)
regulates CD8+ T-cell mediated injury in the heart. Circulation 116:2062.

Rat anti-mouse OX40/TNFRSF4 (Novus, cat. # NBP1-06681, mouse, clone: AP-MAB0833, lot: 2017100203, dilution: 1:50).
Validation data is available on the manufacturer's website.

Rat anti-mouse CD8 (Invitrogen, cat. # 14-0808, mouse, clone: 4SM15, lot: 2003225, dilution: 1:50). Validation data is available
on the manufacturer's website. References: Lu, J., et al. (2017). "Nano-enabled pancreas cancer immunotherapy using
immunogenic cell death and reversing immunosuppression." 8(1): 1811.

Rat anti-mouse CD4 (Invitrogen, cat.# 14-9766, mouse, clone: 4SM95, lot: 4342629, dilution: 1:100). Validation data is available
on the manufacturer's website. References: Jaini, R., et al. (2017). "Immunotherapeutic target expression on breast tumors can
be amplified by hormone receptor antagonism: a novel strategy for enhancing efficacy of targeted immunotherapy." Oncotarget
8(20): 32536-32549.

Rabbit anti-mouse FOXP3 (Novus, cat.# NB600-245, mouse, polyclonal, lot: D-1, dilution: 1:50). Validation data is available on
the manufacturer's website. References: Ochando, JC et al. (2005) Lymph node occupancy is required for the peripheral
development of alloantigen-specific Foxp3+ regulatory T cells. J Immunol;174(11):6993-7005.

Alexa Fluor 488 anti-mouse Ly-6G/Ly-6C (Gr1) (Biolegend, cat.# 108417, mouse, clone: RB6-8C5, lot: B236641, dilution: 1:50).
Validation data is available on the manufacturer's website. References: Brown, C. R., et al. (2004). "Treatment of mice with the
neutrophil-depleting antibody RB6-8C5 results in early development of experimental lyme arthritis via the recruitment of Gr-1-
polymorphonuclear leukocyte-like cells." Infect Immun 72(9): 4956-4965.

APC/Cy7 anti-mouse/human CD11b (Biolegend, cat.# 101225, mouse, clone: M1/70, lot: B249242, dilution: 1:100). Validation
data is available on the manufacturer's website. References: Flotte, T. J., et al. (1983). "Dendritic cell and macrophage staining by
monoclonal antibodies in tissue sections and epidermal sheets." Am J Pathol 111(1): 112-124.

Rabbit anti-mouse cleaved caspase-3 (Cell Signaling, cat.# CST-9664L, mouse, clone: 5A1E, lot: 21 , dilution: 1:50). Validation
data is available on the manufacturer's website. References: Wu, H., et al. (2019). "MiR-208a-3p functions as an oncogene in
colorectal cancer by targeting PDCD4." Bioscience reports 39(4): BSR20181598.

Other antibodies:
Anti-mouse anti-PD-1 (RMP1-14 Cat.#BE0146) was validated by SDS-PAGE and all the relevant data are available on the
October 2018

manufacturer's website. References: Moynihan, K. D., et al. (2016). "Eradication of large established tumors in mice by
combination immunotherapy that engages innate and adaptive immune responses." Nat Med. doi: 10.1038/nm.4200.
Evans, E. E., et al. (2015). "Antibody Blockade of Semaphorin 4D Promotes Immune Infiltration into Tumor and Enhances
Response to Other Immunomodulatory Therapies." Cancer Immunol Res 3(6): 689-701.
Twyman-Saint Victor, C., et al. (2015). "Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in
cancer." Nature 520(7547): 373-377.

Anti-mouse anti-OX40 (OX-86 Cat.#BE0031) was validated by SDS-PAGE and all the relevant data are available on the
manufacturer's website. References: Bartkowiak, T., et al. (2015). "Unique potential of 4-1BB agonist antibody to promote

3
durable regression of HPV+ tumors when combined with an E6/E7 peptide vaccine." Proc Natl Acad Sci U S A 112(38):
E5290-5299.

nature research | reporting summary


Makkouk, A., et al. (2015). "Three steps to breaking immune tolerance to lymphoma: a microparticle approach." Cancer Immunol
Res 3(4): 389-398.
Zander, R. A., et al. (2015). "PD-1 Co-inhibitory and OX40 Co-stimulatory Crosstalk Regulates Helper T Cell Differentiation and
Anti-Plasmodium Humoral Immunity." Cell Host Microbe 17(5): 628-641.

Validation All primary anti-mouse antibodies used for flow cytometry were validated by the manufacturer and the results are available on
the manufacturer's website. All antibodies used for flow cytometry (besides CD11c-FITC (Miltenyi Biotec, Cat.# 130-102-466,
mouse and MHC Class II-Pac. Blue (Miltenyi Biotec, Cat.# 130-102-145, mouse)) were REAfinity, which are recombinant
antibodies that provide superior lot-to-lot consistency and purity compared to mouse or rat hybridoma-derived, monoclonal
antibodies. They have been engineered for high specificity and require no FcR blocking step.
All the clones were tested against different known clones, in a blocking experiment to identify whether they recognize
completely overlapping, partially overlapping, or completely different epitopes. Every lot was tested by mass spectrometry and
other biochemical and cell-based methods, as described in the manufacturer’s website.

Eukaryotic cell lines


Policy information about cell lines
Cell line source(s) Murine bone marrow dendritic cells (DC) JAWSII cell line (ATCC#CRL-11904).
Ret-Melanoma cells were obtained from spontaneously occurring skin tumor in Ret transgenic mice. We received those cells
from Umansky (PMID: 23560814) and further engineered them to express mCherry reporter gene (PMID: 19381291).
B16-F10 cell line (ATCC® CRL-6475) was obtained from American Type Culture Collection (ATCC, Manassas, VA, USA).

Authentication JAWSII and B16-F10 cell line were authenticated by ATCC using morphology, karyotyping, PCR-based techniques, and
Cytocrome oxidase I assay, following manufacturer validated procedures. Ret-Melanoma cells were authenticated by Victor
Umansky. Ret-Melanoma cells and mCherry-labeled Ret murine melanoma cells were not authenticated by the authors.

Mycoplasma contamination All cells lines were negative for mycoplasma

Commonly misidentified lines No commonly misidentified cell lines were used.


(See ICLAC register)

Animals and other organisms


Policy information about studies involving animals; ARRIVE guidelines recommended for reporting animal research
Laboratory animals Male C57BL/6J mice 8 weeks old

Wild animals No wild animals were used in these studies

Field-collected samples The study did not involve samples collected from the field.

Ethics oversight All animal procedures were performed in compliance with Tel Aviv University, Sackler Faculty of Medicine guidelines and with
the Portuguese competent authority for animal protection, Direcção Geral de Alimentação e Veterinária, Lisbon, Portugal. The
protocols were approved by the institutional animal care and use committee (IACUC) and performed in accordance with NIH
guidelines.
Note that full information on the approval of the study protocol must also be provided in the manuscript.

Flow Cytometry
Plots
Confirm that:
The axis labels state the marker and fluorochrome used (e.g. CD4-FITC).
The axis scales are clearly visible. Include numbers along axes only for bottom left plot of group (a 'group' is an analysis of identical markers).
All plots are contour plots with outliers or pseudocolor plots.
October 2018

A numerical value for number of cells or percentage (with statistics) is provided.

Methodology
Sample preparation Tumors were isolated from the animals directly after euthanasia. Tumor single-cell suspensions were obtained by mechanical
disruption of the tissues and enzymatic digestion for 1 hour at 37ºC. Enzymatic digestion solution was prepared in RMPI medium
with 0.5% BSA, 0.1% collagenase type II, 0.1% dispase, and DNase. After digestion, the suspension was filtered through a 70
micrometer filter (BD Biosciences) to remove the debris. The obtained single-cell suspension was then stained with

4
fluorochrome labeled antibodies and analyzed. Intracellular staining was performed with the Inside stain kit (Miltenyi Biotec
Biotec, Cat.# 130-090-477), according to the manufacturer’s protocol.

nature research | reporting summary


The inguinal lymph nodes were harvested 10 days after the administration of the second dose of the nano-vaccine, homogenized
in a single-cell suspension, and plated in 96-well plate for the tetramer staining assay.
The peptide-MHC tetramer tagged with PE (H-2Db – restricted EGSRNQDWL PE-labeled tetramer (Quimigen S.L., Madrid, Spain))
was added to this single cell suspension, including FcR blocking, following supplier instructions. Cells were washed to remove
unbound tetramers, centrifuged, resuspended in ice-cold sorter buffer and plated in 96-well plates. The antibodies CD3-APC-Cy7
and CD8α-PE-Cy7 were added and protected from light during incubation. The cells were washed, centrifuged and resuspended
in ice-cold sorter buffer to determine the percentages of tumor antigen-specific CD3+ CD8α+ T cells by FACS.

Instrument BD Biosciences BD LSR FORTESSA.

Software Facs Diva was used to collect data. This data was analyzed using FlowJo v10 software.

Cell population abundance These data are available in the Supplementary Information.

Gating strategy These data are detailed in the Supplementary Information for each flow cytometry panel.

Tick this box to confirm that a figure exemplifying the gating strategy is provided in the Supplementary Information.

October 2018

Вам также может понравиться