Maxence O. Dellacherie’s research while affiliated with Harvard University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (13)


IL-2/anti-IL-2 antibody complexes augment immune responses to therapeutic cancer vaccines
  • Article

November 2024

·

19 Reads

·

1 Citation

Proceedings of the National Academy of Sciences

Miguel C. Sobral

·

Laura Cabizzosu

·

Shawn J. Kang

·

[...]

·

David J. Mooney

One driver of the high failure rates of clinical trials for therapeutic cancer vaccines is likely the inability to sufficiently engage conventional dendritic cells (cDCs), the antigen-presenting cell (APC) subset that is specialized in priming antitumor T cells. Here, we demonstrate that, relative to vaccination with an injectable mesoporous silica rod (MPS) vaccine alone (Vax), combining MPS vaccines with CD122-biased IL-2/anti-IL-2 antibody complexes (IL-2cx) drives ~3-fold expansion of cDCs at the vaccination sites, vaccine-draining lymph nodes, and spleens of treated mice. Furthermore, relative to Vax alone, Vax+IL-2cx led to a ~3-fold increase in the numbers of CD8 ⁺ T cells and ~15-fold increase in the numbers of NK cells at the vaccination site. Notably, with both the model protein antigen OVA as well as various peptide neoantigens, Vax+IL-2cx induced ~5 to 30-fold greater numbers of circulating antigen-specific CD8 ⁺ T cells relative to Vax alone. We further demonstrate that Vax+IL-2cx leads to significantly improved efficacy in the MC38 colon carcinoma model relative to either monotherapy alone, driving complete regressions in 50% of mice in a cDC-dependent manner. Relative to vaccine alone, Vax+IL-2cx led to comparable numbers of CD8 ⁺ T cells, but markedly greater numbers of NK cells and activated cDCs in the B16F10 melanoma tumor microenvironment post-therapy. Taken together, these findings suggest that the administration of factors that engage both the cDC-CD8 ⁺ T cell and cDC-NK cell axes can boost the potency of therapeutic cancer vaccines.


MPS vaccine characterization
a, Scanning electron microscopy imaging of MPS particles. b, MPS particle size distribution. c, Size and volume characterization of MPS particles. In vitro release curves of d CpG and e GM-CSF from MPS vaccines. n = 3 biologically independent replicates. For d-e, means depicted; error bars, s.d.
Source data
Comparing lymph-node expansion with MPS and alternative vaccine formulations
Mice were immunized with MPS vaccines (GM-CSF, CpG, and OVA protein), MPS vaccines without CpG (GM-CSF and OVA only), or MPS vaccines without GM-CSF (CpG and OVA only). Additional mice were immunized with the same quantities of GM-CSF, CpG, and OVA in alum, emulsion (MF59), or alginate cryogel-based vaccines. A final group was dosed with an MPS vaccine containing a log-fold lower dose each of CpG and OVA. Vaccine-draining LNs were longitudinally imaged using high frequency ultrasound. a, Quantification of vaccine draining LN fold expansion over time, comparing the full MPS vaccine with formulations lacking either CpG or GM-CSF. Statistical analysis was performed using a one-way ANOVA with Dunnett’s post hoc test. n = 4 (full MPS) or 5 (other groups) biologically independent animals per group. b, Vaccine draining LN fold expansion over time, comparing the full MPS vaccine with a low-dose MPS vaccine. n = 4 (full MPS) or 10 (low-dose MPS) biologically independent animals per group. The low-dose MPS group displays the combined results of two separate experiments. Statistical analysis was performed using a two-tailed t test. c, Vaccine draining LN fold expansion comparing MPS, alum, MF59 emulsion, and cryogel formulations. Statistical analysis was performed using a one-way ANOVA with Dunnett’s post hoc test (t = 7, 11, 21 days) or a Kruskal-Wallis test with Dunn’s post hoc test (t = 4 days). P values for the comparison between MPS and other vaccine systems are displayed on the plot. n = 4 (full MPS) or 5 (other groups) biologically independent animals per group. For a-c, values are normalized to the baseline volume for each individual LN; means depicted; error bars, s.d.
Source data
Comparing lymph-node expansion with MPS and vaccines lacking sustained antigen release
Mice were immunized with MPS vaccines (GM-CSF, CpG, and OVA protein), MPS vaccines without antigen (GM-CSF and CpG alone), or MPS vaccines with bolus antigen (GM-CSF and CpG loaded into MPS, with OVA injected separately as a bolus). Vaccine-draining LNs were longitudinally imaged using high frequency ultrasound. a, LN fold expansion over time, normalized to volume at the day 0 timepoint. n = 5 biologically independent animals per group. b, Vaccine draining LN fold expansion over time, comparing the full MPS vaccine (antigen loaded in MPS material) to an MPS vaccine with a separate bolus injection of antigen. n = 4 (full MPS) or 5 (bolus antigen) biologically independent animals per group. For a-b, values are normalized to the baseline volume for each individual LN; means depicted; error bars, s.d.
Source data
MPS vaccination induces potent germinal centre B cell responses in the LN
Mice were treated with MPS or bolus vaccines (containing GM-CSF, CpG, OVA), MPS vaccine without antigen (GM-CSF, CpG only), or PBS, and LNs were collected on days 7, 14, and 20 for cellular analysis. n = 5 biologically independent animals per group per timepoint. a, Representative flow cytometry plots depicting CD3⁻CD19⁺ B cells from LNs on day 20. GL7hi B cells (germinal centre B cells) are gated. b, Proportion GL7hi of B cells in LNs over time. Statistical analyses were performed using analysis of variance (ANOVA) with Tukey’s post hoc test (days 7 and 14) and Kruskal-Wallis test with Dunn’s post hoc test (day 20). c, Number of GL7hi B cells (that is germinal centre B cells) in LNs over time. Statistical analyses were performed using analysis of variance (ANOVA) with Tukey’s post hoc test (day 14) and Kruskal-Wallis test with Dunnett’s post hoc test (days 7 and 20). For b-c, means depicted; error bars, s.d. Differences present between any group and the PBS control group are depicted.
Source data
MPS vaccination minimally impacts splenic immune cells, with the exception of inflammatory monocytes
Mice were treated with MPS or bolus vaccines (containing GM-CSF, CpG, OVA), MPS vaccine without antigen (GM-CSF and CpG only), or PBS, and spleens and blood were collected on days 7, 14, and 20 for cellular analysis. a, Total cell counts in the spleen. b, Number of T cells (CD3⁺CD19⁻) in the spleen. Statistical analysis was performed using a one-way ANOVA with Tukey’s post hoc test. c, Number of B cells (CD3⁻CD19⁺) in the spleen. d, Numbers of dendritic cells (CD3⁻CD19⁻Ly6C⁻F4/80⁻CD11c⁺MHCII⁺) in the spleen. e, Number of macrophages (CD3⁻CD19⁻Ly6C⁻CD11b⁺F4/80⁺) in the spleen. Statistical analysis was performed using a one-way ANOVA with Tukey’s post hoc test. f, Proportion of inflammatory monocytes (CD3⁻CD19⁻Ly6ChiCD11b⁺) of total cells in the spleen. Statistical analysis was performed using a one-way ANOVA with Tukey’s post hoc test. g, Number of inflammatory monocytes in the spleen. Statistical analysis was performed using a one-way ANOVA with Tukey’s post hoc test. h, Proportion of inflammatory monocytes in the blood. Statistical analysis was performed using a one-way ANOVA with Tukey’s post hoc test. For a-h, n = 5 biologically independent animals per group per timepoint; means depicted; error bars, s.d.; statistical significance is shown between the MPS dLN group and all other groups.
Source data

+12

Durable lymph-node expansion is associated with the efficacy of therapeutic vaccination
  • Article
  • Full-text available

May 2024

·

55 Reads

·

9 Citations

Nature Biomedical Engineering

Following immunization, lymph nodes dynamically expand and contract. The mechanical and cellular changes enabling the early-stage expansion of lymph nodes have been characterized, yet the durability of such responses and their implications for adaptive immunity and vaccine efficacy are unknown. Here, by leveraging high-frequency ultrasound imaging of the lymph nodes of mice, we report more potent and persistent lymph-node expansion for animals immunized with a mesoporous silica vaccine incorporating a model antigen than for animals given bolus immunization or standard vaccine formulations such as alum, and that durable and robust lymph-node expansion was associated with vaccine efficacy and adaptive immunity for 100 days post-vaccination in a mouse model of melanoma. Immunization altered the mechanical and extracellular-matrix properties of the lymph nodes, drove antigen-dependent proliferation of immune and stromal cells, and altered the transcriptional features of dendritic cells and inflammatory monocytes. Strategies that robustly maintain lymph-node expansion may result in enhanced vaccination outcomes.

Download

Fine tuning of CpG spatial distribution with DNA origami for improved cancer vaccination

March 2024

·

251 Reads

·

49 Citations

Nature Nanotechnology

Multivalent presentation of ligands often enhances receptor activation and downstream signalling. DNA origami offers a precise nanoscale spacing of ligands, a potentially useful feature for therapeutic nanoparticles. Here we use a square-block DNA origami platform to explore the importance of the spacing of CpG oligonucleotides. CpG engages Toll-like receptors and therefore acts to activate dendritic cells. Through in vitro cell culture studies and in vivo tumour treatment models, we demonstrate that square blocks induce Th1 immune polarization when CpG is spaced at 3.5 nm. We observe that this DNA origami vaccine enhances DC activation, antigen cross-presentation, CD8 T-cell activation, Th1-polarized CD4 activation and natural-killer-cell activation. The vaccine also effectively synergizes with anti-PD-L1 for improved cancer immunotherapy in melanoma and lymphoma models and induces long-term T-cell memory. Our results suggest that DNA origami may serve as a platform for controlling adjuvant spacing and co-delivering antigens in vaccines.


Adoptive T cell transfer and host antigen-presenting cell recruitment with cryogel scaffolds promotes long-term protection against solid tumors

June 2023

·

127 Reads

·

27 Citations

Although adoptive T cell therapy provides the T cell pool needed for immediate tumor debulking, the infused T cells generally have a narrow repertoire for antigen recognition and limited ability for long-term protection. Here, we present a hydrogel that locally delivers adoptively transferred T cells to the tumor site while recruiting and activating host antigen-presenting cells with GMCSF or FLT3L and CpG, respectively. T cells alone loaded into these localized cell depots provided significantly better control of subcutaneous B16-F10 tumors than T cells delivered through direct peritumoral injection or intravenous infusion. T cell delivery combined with biomaterial-driven accumulation and activation of host immune cells prolonged the activation of the delivered T cells, minimized host T cell exhaustion, and enabled long-term tumor control. These findings highlight how this integrated approach provide both immediate tumor debulking and long-term protection against solid tumors, including against tumor antigen escape.


Lymph node expansion predicts magnitude of vaccine immune response

October 2022

·

60 Reads

·

2 Citations

Lymph nodes (LNs) dynamically expand in response to immunization, but the relationship between LN expansion and the accompanying adaptive immune response is unclear. Here, we first characterized the LN response across time and length scales to vaccines of distinct strengths. High-frequency ultrasound revealed that a bolus ‘weak’ vaccine induced a short-lived, 2-fold volume expansion, while a biomaterial-based ‘strong’ vaccine elicited an ∼7-fold LN expansion, which was maintained several weeks after vaccination. This latter expansion was associated with altered matrix and mechanical properties of the LN microarchitecture. Strong vaccination resulted in massive immune and stromal cell engagement, dependent on antigen presence in the vaccine, and conventional dendritic cells and inflammatory monocytes upregulated genes involved in antigen presentation and LN enlargement. The degree of LN expansion following therapeutic cancer vaccination strongly correlated with vaccine efficacy, even 100 days post-vaccination, and direct manipulation of LN expansion demonstrated a causative role in immunization outcomes.


Fig. 2 T cell depots alone enhance B16-F10 tumor control. a. Schematic of therapeutic study. Tumor volumes (b) and Kaplan-Meier survival curves (c), with sub-lethal irradiation preconditioning of mice left untreated, injected peritumorally with empty depots or treated with the following T cell conditions: intravenous T cell delivery, direct peritumoral T cell injection or T cell depots. p-values for c were determined by Log-rank (Mantel-Cox) test. Data are n=15-16 mice per condition, 2 independent studies (7-8 mice per study). d-e. Therapeutic study without preconditioning. Tumor growth (d) and Kaplan-Meier survival curves (e) of the indicated conditions. For the 'Contralateral Injection_T cell Depot' condition, T cell loaded depots were injected contralateral to the tumor location. p-values for e were determined by Log-rank (Mantel-Cox) test. Data are n=7 or 8 mice per condition.
Fig. 3 SIVETs elicit host T cell responses. a. Schematic of experiment. The following conditions were investigated: 1) no treatment controls (NT), 2) T cell only depots (TcellOnly_Depot), 3) SIVET with antigen-free FLT3L vaccine (SIVET_ FLT3L), 4) SIVET with antigen-free GMCSF vaccine (SIVET_ GMCSF), 5) antigen-free GMCSF vaccine-only depot (Vax_GMCSF) and 6) antigen-free FLT3L vaccine-only depot (Vax_ FLT3L). Proportions of IFNG expressing host CD8+ T cells isolated from lymph nodes (b) and spleens (c) after in vitro antigen re-stimulation. Proportions of IFNG expressing host CD4+ isolated from lymph nodes (d) and spleens (e) after in vitro antigen re-stimulation. p-values were determined by two-tailed one-way ANOVA with GeisserGreenhouse correction. Data are mean ± s.e.m from n=4 mice per condition.
Fig. 4 SIVETs enhance the relative levels of activated antigen-presenting cells in tumors. a. Schematic of experiment. b. Total numbers of tumor infiltrating CD45+ cells per mm^3 of tumor volume. p-value was determined by two-tailed one-way ANOVA with Geisser-Greenhouse correction. Data are mean ± s.e.m from n=3 mice per condition. c. Immunofluorescence imaging of myeloid cells expressing the indicated markers in NT,
Fig. 5 SIVETs minimize host T cell exhaustion in tumors. a. Schematic of experiment. b. Numbers of tumor infiltrating T cells per mm^3 of tumor volume. p-value was determined by two-tailed one-way ANOVA with Geisser-Greenhouse correction. Data are mean ± s.e.m from n=3 mice per condition. c-d. Immunofluorescence imaging of T cells present in NT and SIVET_GMCSF (c), as well as specific T cell subtypes in TcellOnly_Depot and SIVET_GMCSF tumors (d). e. Umap plots showing expression of the indicated markers. f. Umap plots of individual treatment conditions showing distinct localization of cells based on treatment group. g. Umap plot overlaid with Kmeans clusters of T cells in tumors. h. Heatmap plot showing the average expression of the indicated
Fig. 6 SIVETs enhance long-term tumor control. a. Schematic of experiment. b-c. Primary B16-F10 tumor studies. Tumor volumes (b) and Kaplan-Meier survival curves (c) comparing tumor growth and survival of mice treated with the indicated conditions. p-values for c were determined by Log-rank (Mantel-Cox) test. Data represent n=7-8 mice per condition for TcellOnly_Depot, Vax_FLT3L and Vax_GMCSF conditions and n=15-16 mice per condition, 2 independent studies (7-8 mice per study) for NT, SIVET_FLT3L and SIVET_GMCSF conditions. d-e. Contralateral tumor re-challenge studies for long-term surviving mice. d. Tumor growth (d) and Kaplan-Meier survival curves (e) of mice contralaterally re-challenged with 1e5 B16-F10 tumor cells after 120 days of primary tumor challenge. p-values for e were determined by Log-rank (Mantel-Cox) test. Data are n=10-13 mice per condition (2 independent studies). f-h. Antigen escape study. f. Schematic of therapeutic study for g-h. Tumor
Enhancing adoptive T cell therapy with synergistic host immune engagement promotes long-term protection against solid tumors

September 2022

·

66 Reads

·

1 Citation

Adoptive T cell therapy provides the T cell pool needed for immediate tumor debulking, but the infused T cells generally have a narrow repertoire for antigen recognition and limited ability for long-term protection. Here, we present a biomaterial platform that enhances adoptive T cell therapy by synergistically engaging the host immune system via in-situ antigen-free vaccination. T cells alone loaded into these localized cell depots provided significantly better control of subcutaneous B16-F10 tumors than T cells delivered through direct peritumoral injection or intravenous infusion. The anti-tumor response was significantly enhanced when T cell delivery was combined with biomaterial-driven accumulation and activation of host immune cells, as this prolonged the activation state of the delivered T cells, minimized host T cell exhaustion, and enabled long-term tumor control. This integrated approach provides both immediate tumor debulking and long-term protection against solid tumors, including against tumor antigen escape.


Efficacy of the MICA/B α3 domain cancer vaccine
a, Design of the MICA/B vaccine. pAbs, polyclonal serum IgG. b, MICB-specific serum antibody (Ab) titres quantified by flow cytometry (n = 4 mice per group) in MICB-transgenic mice immunized with Ctrl-vax (blue) or MICB-vax (red). MFI, median fluorescence intensity. c, MICB-specific CD4⁺ T cell responses following immunization with MICB-vax or Ctrl-vax; CFSE dilution of splenocytes stimulated with MICB or control protein (ovalbumin (OVA)); shown are representative flow cytometry plots (left) and quantification for three mice per group (right). d, Cell-surface levels of MICB on B16F10 (MICB) tumours from mice immunized with MICB-vax or Ctrl-vax (n = 4 mice per group); staining of tumour cells with isotype-control monoclonal antibody (grey) or anti-MICA/B monoclonal antibody (specific for the α1–α2 domains, monoclonal antibody not blocked by vaccine-induced antibodies). e, Therapeutic efficacy of MICB-vax (red) or Ctrl-vax (blue) in mice with established B16F10 (MICB) tumours immunized at the indicated time points (n = 7 mice per group). Vax, vaccination. *P = 0.0137, ****P < 0.0001; NS (not significant), P > 0.999. f, g, Vaccine efficacy in two models of spontaneous metastasis. Mice were immunized with Ctrl-vax (blue) or MICB-vax (red) following surgical removal of primary tumours using the B16-B6 melanoma (f; 10 mice per group) or 4T1 breast cancer (g; 13 mice per group) models. Shown are the size of primary tumours at the time of surgery (left), representative images of lung metastases (middle) and quantification of the total number of lung surface metastases (right). D, day; s.c., subcutaneous. h, i, Immunogenicity of the rhesus MICA/B α3 domain vaccine in the rhesus macaque model. h, Timeline of vaccination; blood was drawn 24 h before indicated immunization or boost. i, Serum titres of antibody to rhesus MICA/B for animal ID 9312. Representative data are shown from at least three (b) or two (c–g) independent experiments. Data from a single experiment with technical replicates for each time point are shown in i. Statistical significance was assessed by two-tailed unpaired Student’s t test (b), two-way ANOVA with Sidak’s multiple-comparison test (c), one-way ANOVA with Tukey’s multiple-comparison test (d), two-way ANOVA with Bonferroni’s post hoc test (left) and the log-rank (Mantel–Cox) test (right) (e), two-tailed Mann–Whitney test (f, g) and two-way ANOVA with Tukey’s multiple-comparison test (i). Data are depicted as the mean ± s.e.m. (b–g) or mean ± s.d. (i).
Source data
Vaccine induces T cell and NK cell recruitment into tumours
a, Strategy for characterization of tumour-infiltrating immune cells by flow cytometry (FACS) and scRNA-seq. Mice received two immunizations (days 0 and 14), B16F10 (MICB-dox) tumour cells were implanted (day 21), MICB expression was induced on tumour cells by doxycycline treatment (day 28) and tumour-infiltrating immune cells were analysed 7 d later. b–d, Tumour-infiltrating T cell populations following immunization with Ctrl-vax (blue) or MICB-vax (red) (Ctrl-vax, n = 9 mice per group; MICB-vax, n = 10 mice per group; n = 7 mice per group for the γδ T cell panel). Treg cells, regulatory T cells. e, Tumour-infiltrating NK cells (n = 8 mice per group). f, g, Quantification of IFNγ-positive CD4⁺ and CD8⁺ T cells (Ctrl-vax, n = 9 mice per group; MICB-vax, n = 10 mice per group). h, i, UMAP representation of all T cell clusters from scRNA-seq data (h) and the fraction of each T cell subpopulation among total CD45⁺ cells from the experimental group (MICB-vax + doxycycline; red) and three combined control groups (Ctrl-vax ± doxycycline and MICB-vax without doxycycline; blue) (i). DN, double negative. j, k, UMAP representation of NK and ILC1 cells from all experimental groups (j) and the fraction of these subpopulations among total CD45⁺ cells from experimental (red) and combined control (blue) groups (k). l, Fraction of T cells representing expanded clones based on TCR sequence analysis for the experimental group (red) and combined control groups (blue). m, Contribution of CD4⁺ T cells, NK cells and NKG2D receptor to vaccine efficacy. Mice were first immunized with MICB-vax (M) or Ctrl-vax (C) (days 0 and 14) and treated with isotype-control monoclonal antibody (iso), depleting monoclonal antibody (targeting CD4⁺ T cells or NK cells (αCD4 and αNK1.1, respectively)) or NKG2D receptor-blocking monoclonal antibody (αNKG2D) starting on day 21, followed by implantation of B16F10 (MICB) tumour cells (n = 7 mice per group). Representative data from three independent experiments are shown in b–g. scRNA-seq data from a single experiment with sorted CD45⁺ cells pooled from five mice per group are shown in h–l. Representative data from two independent experiments are shown in m. Statistical significance was assessed by two-tailed Mann–Whitney test (b–g) or log-rank (Mantel–Cox) test (m). Data are depicted as the mean ± s.e.m.
Source data
Vaccine retains efficacy against MHC-I-deficient tumours
a, b, Comparison of vaccine efficacy against B16F10 (MICB) WT tumours and tumours with resistance mutations in the B2m (a) or Ifngr1 (b) gene. Mice received MICB-vax or Ctrl-vax and were then challenged with tumours of the indicated genotype (n = 7 mice per group). KO, knockout. c, Effect of CD4⁺ T cell and NK cell depletion on immunity to B2m-knockout tumours. Mice were immunized with MICB-vax or Ctrl-vax; treatment with depleting or isotype-control monoclonal antibody was started 2 d before injection of B2m-knockout B16F10 (MICB) tumour cells (n = 7 mice per group). d, Contribution of vaccine-induced anti-MICB antibodies to NK cell-mediated cytotoxicity against B2m-knockout B16F10 (MICB) tumour cells. CFSE-labelled B2m-knockout B16F10 (MICB) tumour cells were pre-incubated with 10 µg per well of purified serum IgG from mice immunized with MICB-vax or Ctrl-vax before the addition of NK cells at different effector to target (E:T) ratios as indicated. The percentage of dead target cells was assessed by flow cytometry. e, Effect of CD4⁺ T cell depletion on vaccine-induced NK cell infiltration into tumours. Flow cytometry quantification of total NK cell numbers is shown in WT (left) and B2m-knockout (right) tumours for the following treatment groups: Ctrl-vax + isotype-control monoclonal antibody (blue), Ctrl-vax + anti-CD4 (orange), MICB-vax + isotype-control monoclonal antibody (red) and MICB-vax + anti-CD4 (green) (n = 7 mice per group). Representative data from two independent experiments are shown in a–e. Statistical significance was assessed by two-way ANOVA with Bonferroni’s post hoc test (left) and log-rank (Mantel–Cox) test (right) (a–c), two-way ANOVA with Sidak’s multiple-comparison test (d) and one-way ANOVA with Tukey’s multiple-comparison test (e). Data are depicted as the mean ± s.e.m.
Source data
Role of CD4⁺ T cells and cDC1 cells in NK cell recruitment to tumours
a, Effect of CD4⁺ T cells on migratory DC populations in the tdLNs of mice immunized with MICB-vax versus Ctrl-vax. Total migratory DCs as well as cDC1 and cDC2 cells were quantified 2 d after induction of MICB expression in tumour cells by doxycycline treatment (n = 7 mice per group, except n = 6 for Ctrl-vax without anti-CD4). b, Migratory DC subsets within the tdLN of MICB-vax-immunized mice treated following immunization (days 28 + 30) with isotype-control, CD4-depleting or CD40L-blocking monoclonal antibody (n = 7 mice per group). c, Quantification of DC populations within the tumours of mice immunized with Ctrl-vax (blue) or MICB-vax (red) on day 7 following induction of MICB expression with doxycycline (n = 7 mice per group). d, Effect of cDC1 depletion on MICB vaccine-induced T cell and NK cell accumulation within tumours in Xcr1DTR mice. Mice were treated with DT or left untreated starting on day 26 following immunization with Ctrl-vax or MICB-vax (days 0 + 14) and B16F10 (MICB-dox) tumour implantation (day 21). Immune cells were analysed in tumours 7 d after induction of MICB expression on tumours with doxycycline (day 37) (n = 7 mice per group). e, Contribution of vaccine-induced anti-MICB antibodies to DC-mediated cross-presentation of tumour antigens to CD8⁺ T cells. Bone marrow-derived DCs (BMDCs) were pre-incubated with B2m-knockout B16F10 (MICB-OVA) tumour cells in the presence of affinity-purified serum IgG from mice immunized with Ctrl-vax or MICB-vax at the indicated concentrations. DCs were co-cultured with CFSE-labelled OT-1 CD8⁺ T cells with T cell proliferation as the readout. The role of activating Fc receptor (FcR) was assessed using BMDCs from Fcer1g–/– mice (orange) or pre-incubation of BMDCs with FcR-blocking antibody (yellow) before tumour cell addition. Representative data from two independent experiments are shown in a–e. Statistical significance was assessed by one-way ANOVA with Tukey’s multiple-comparison test (a, b, d), two-tailed Mann–Whitney test (c) and two-way ANOVA with Tukey’s multiple-comparison test (e). Data are depicted as the mean ± s.e.m. (a–d) or mean ± s.d. (e).
Source data
A vaccine targeting resistant tumours by dual T cell plus NK cell attack

June 2022

·

1,010 Reads

·

107 Citations

Nature

Most cancer vaccines target peptide antigens, necessitating personalization owing to the vast inter-individual diversity in major histocompatibility complex (MHC) molecules that present peptides to T cells. Furthermore, tumours frequently escape T cell-mediated immunity through mechanisms that interfere with peptide presentation1. Here we report a cancer vaccine that induces a coordinated attack by diverse T cell and natural killer (NK) cell populations. The vaccine targets the MICA and MICB (MICA/B) stress proteins expressed by many human cancers as a result of DNA damage2. MICA/B serve as ligands for the activating NKG2D receptor on T cells and NK cells, but tumours evade immune recognition by proteolytic MICA/B cleavage3,4. Vaccine-induced antibodies increase the density of MICA/B proteins on the surface of tumour cells by inhibiting proteolytic shedding, enhance presentation of tumour antigens by dendritic cells to T cells and augment the cytotoxic function of NK cells. Notably, this vaccine maintains efficacy against MHC class I-deficient tumours resistant to cytotoxic T cells through the coordinated action of NK cells and CD4+ T cells. The vaccine is also efficacious in a clinically important setting: immunization following surgical removal of primary, highly metastatic tumours inhibits the later outgrowth of metastases. This vaccine design enables protective immunity even against tumours with common escape mutations. A vaccine targeting stress proteins expressed by many cancers blocks a tumour escape mechanism, enabling protective immunity mediated by diverse T cell and NK cell populations.


Optimizing CpG spatial distribution with DNA origami for Th1-polarized therapeutic vaccination

June 2022

·

69 Reads

·

12 Citations

Multivalent presentation of ligands often enhances receptor activation and downstream signaling. DNA origami offers precise nanoscale spacing of ligands, a potentially useful feature for therapeutic nanoparticles. Here we introduce a “square block” DNA origami platform to explore the importance of spacing of CpG oligonucleotides, which engage Toll-like receptors and thereby act as danger signals for dendritic cells. Through in vitro cell-culture studies and in vivo tumor-treatment models, we demonstrate that square blocks induce Th1 immune polarization when CpG is spaced at 3.5 nm. We observe that this DNA origami vaccine enhances DC activation, antigen cross-presentation, CD8 T cell activation, Th1-polarized CD4 activation and NK cell activation. The vaccine also synergizes effectively with anti-PD-L1 for improved cancer immunotherapy in melanoma and lymphoma models and induced long-term T cell memories. Our results suggest that DNA origami may serve as an advanced vaccine platform for controlling adjuvant spacing and co-delivering antigens. One Sentence Summary This study developed a DNA origami-based cancer vaccine (DoriVac) that co-delivers antigen and CpG immune adjuvant with an optimal spacing for Th1 immune polarization.


Tumor enzyme treatment expands immune cell populations in melanoma tumor-draining lymph nodes. a Image of untreated or enzyme-treated tdLNs; scale bar = 5 mm. b Total cell count in tdLNs. c Total cell count in spleens. No significant differences were found between groups. d CD8⁺ and CD4⁺ T cell numbers in tdLNs. e DC numbers (CD11c⁺MHCII⁺) in tdLNs. f Representative flow cytometry plots of myeloid cells within tdLNs, gated on CD45⁺CD11b⁺ live cells. g Numbers of neutrophils (CD11b⁺Gr-1⁺SSC-Ahi), granulocytes (CD11b⁺Gr-1⁻SSC-Ahi), and macrophages (CD11b⁺F4/80⁺Gr-1⁻) within tdLNs. In a and d–g, mice bearing B16-OVA tumors received intratumoral hyaluronidase (enzyme) or no injection (untreated) along with adoptive transfer of OT1 cells, and tdLNs were collected. In b, c, tdLNs and spleens were collected from mice bearing B16-mCherry tumors treated with ET or PBS, or naïve mice without tumors (non-tdLN). Data are depicted mean ± SD. For b, c, n = 5 (untreated) or 6 (PBS or enzyme-treated) biologically independent animals per group; statistical analysis was performed using analysis of variance (ANOVA) with Tukey’s post hoc test. For d, e and g, n = 3–4 biologically independent animals per group; statistical analyses were performed using two-tailed t tests
Enzyme treatment enhances tumor antigen presentation in secondary lymphoid organs. a Representative flow cytometry plots of CD103 expression on DCs (CD11c⁺MHCII⁺) in tdLNs. b Percentage of DCs expressing CD103 in tdLNs. c Percentage of DCs expressing CD103 in spleens. d Overview of experimental conditions and e timeline. f Number of CD11c⁺ DCs expressing CD103 in tdLNs. g Number of SIINFEKL-presenting CD11c⁺ DCs in tdLNs. In a–c, tdLNs and spleens were collected from mice bearing B16-mCherry tumors treated with ET or PBS, or naïve mice without tumors (non-tdLN). In d–g, B16-OVA tumor-bearing mice were injected with vaccine cryogels with or without OVA antigen, administered intratumoral enzyme or PBS treatment, and tdLNs and spleens collected. Data are depicted mean ± SD. For b, c, n = 5 (untreated) or 6 (PBS or enzyme-treated) biologically independent animals per group; statistical analysis was performed using ANOVA with Tukey’s post hoc test. For f, g, n = 6 biologically independent animals per group; statistical analysis was performed using a Kruskal–Wallis test with Dunn’s post hoc test (f) or ANOVA with Tukey’s post hoc test (g)
Enzyme treatment improves antigen-specific T cell responses in the tumor-draining lymph node. B16-OVA tumor-bearing mice were injected with vaccine cryogels with or without OVA antigen, administered intratumoral enzyme or PBS treatment, and tdLNs collected. a Number of CD8⁺ T cells in tdLNs. b Number of OVA-tetramer⁺ CD8⁺ T cells in tdLNs. c Pie charts depicting the percentage of CD8⁺ T cells expressing 0–3 of the cytokines IFN-γ, IL-2, or TNF-α in each group following restimulation with SIINFEKL peptide. Blue represents T cells with no detectable cytokine expression, orange represents cells expressing a single cytokine, gray represents two cytokines, and yellow represents cells expressing all three cytokines. Higher-magnification view of the cytokine positive cells shown to the right of each pie chart. d Quantification of T cell polyfunctionality through the percentage of CD8⁺ T cells expressing two or more of the cytokines IFN-γ, IL-2, or TNF-α. Data are depicted mean ± SD; n = 6 biologically independent animals per group. Statistical analysis was performed using a Kruskal–Wallis test with Dunn’s post hoc test (a, b) or ANOVA with Tukey’s post hoc test (d)
Enzyme treatment synergizes with cryogel vaccination to elicit systemic antigen-specific immune responses. B16-OVA tumor-bearing mice were injected with vaccine cryogels with or without OVA antigen, administered intratumoral enzyme or PBS treatment, and spleens collected. a Percentage of CD8⁺ T cells binding OVA tetramer in spleens. b Representative flow cytometry plots of IFN-γ (top) and IL-2 expression (bottom) in CD8⁺ T cells in spleens following OVA peptide restimulation. c Percentage of CD8⁺ T cells expressing IFN-γ in spleens. d Percentage of CD8⁺ T cells expressing IL-2 in spleens. e Pie charts depicting the percentage of CD8⁺ T cells expressing 0–3 of the cytokines IFN-γ, IL-2, or TNF-α in each group following OVA peptide restimulation. Blue represents T cells with no detectable cytokine expression, orange represents cells expressing a single cytokine, gray represents two cytokines, and yellow represents cells expressing all three cytokines. Higher-magnification view of the cytokine positive cells shown to the right of each pie chart. f Quantification of T cell polyfunctionality through the percentage of CD8⁺ T cells expressing two or more of the cytokines IFN-γ, IL-2, or TNF-α. g Percentage of CD8⁺ T cells expressing PD-1 in spleens. Data are depicted mean ± SD; n = 6 biologically independent animals per group. Statistical analysis was performed using a Kruskal–Wallis test with Dunn’s post hoc test (a, c, d, f) or ANOVA with Tukey’s post hoc test (g)
Targeting tumor extracellular matrix activates the tumor-draining lymph nodes

May 2022

·

82 Reads

·

10 Citations

Cancer Immunology and Immunotherapy

Disruption of the tumor extracellular matrix (ECM) may alter immune cell infiltration into the tumor and antitumor T cell priming in the tumor-draining lymph nodes (tdLNs). Here, we explore how intratumoral enzyme treatment (ET) of B16 melanoma tumors with ECM-depleting enzyme hyaluronidase alters adaptive and innate immune populations, including T cells, DCs, and macrophages, in the tumors and tdLNs. ET increased CD103⁺ DC abundance in the tdLNs, as well as antigen presentation of a model tumor antigen ovalbumin (OVA), eliciting local OVA-specific CD8⁺ T cell responses. Delivered in combination with a distant cryogel-based cancer vaccine, ET increased the systemic antigen-specific CD8⁺ T cell response. By enhancing activity within the tdLN, ET may broadly support immunotherapies in generating tumor-specific immunity.


PLG vaccines generate strong and persistent humoral immune responses against GnRH peptide, with antibody subclasses dependent on CpG formulation. In (a) and (b), C57BL/6 mice were immunized with PLG, MSR, or Cryogel vaccines containing OVA‐GnRH peptide and compared to PBS‐injected controls. Serum was collected up to 400 d after vaccination. a) IgG1 and b) IgG2c titers against GnRH. Data represent mean ± SD; n = 8 biologically independent animals per group, sampled longitudinally. IgG1 titers are not significantly different (ns) between the three scaffold vaccine groups. For IgG2c titers in (b), α indicates p < 0.05 between PLG and Cryogel vaccine groups, β indicates p < 0.05 between PLG and MSR vaccine groups. c) Photographs (above) and scanning electron microscope images (below) of PLG scaffolds containing PEI‐CpG or free CpG. d) In vitro release of CpG, incorporated in free form or with PEI condensation, from PLG scaffolds. Differences are statistically significant (1 h, p = 0.005; 6 h, p = 0.009; 24 and 72 h, p = 0.008; 168 and 336 h, p = 0.007). For (e)–(g), C57BL/6 mice were immunized with PLG vaccines containing PEI‐CpG or free CpG and compared to naïve controls. Serum was collected up to 336 d after vaccination. e) IgG1 and f) IgG2c titers. Data represent mean ± SD; n = 5 biologically independent animals per group, sampled longitudinally. * indicates p < 0.05 between PEI‐CpG PLG vaccine group and free CpG PLG vaccine group. g) Immunohistochemistry of B220, IgG1, and IgG2c in axillary lymph nodes six weeks after vaccination. Naïve, PLG PEI‐CpG, and PLG free CpG lymph nodes are depicted (left scale bar = 500 µm) and higher magnification of a free CpG lymph node (different section of the same node, right scale bar = 150 µm).
Free CpG PLG vaccines promote durable germinal center activation, Tfh response, and Th1‐type immunity. a) Experimental timeline and vaccine conditions. C57BL/6 mice were immunized with PLG (PEI‐CpG or free CpG) or bolus vaccines containing OVA‐GnRH on day 0, and compared to naïve controls. Draining lymph nodes and the PLG scaffold site were collected 7, 14, 21, and 42 d after vaccination. b) Immunohistochemistry of axillary lymph nodes to assess germinal centers six weeks after immunization. Scale bar = 1 mm. Flow cytometry analysis of c) germinal center B cells (B220⁺ GL7⁺ peanut agglutinin⁺) and d) T follicular helper cells (CD3⁺ CD4⁺ CXCR5⁺ PD‐1⁺) within pooled axillary and inguinal lymph nodes. e) Concentrations of cytokines IL‐5 (left) and IL‐10 (right) within tissues formed in PLG scaffolds. For (c)–(e), data represent mean ± SD; n = 4 biologically independent animals per group per timepoint. f,g) C57BL/6 mice were immunized with PLG scaffolds containing no adjuvant (Blank), PEI alone, or PEI + CpG in a free form (not condensed), and scaffolds, lymph nodes, and spleens were extracted after 7 d. f) Concentration of IL‐5 in scaffolds and g) ratio of Th1 (Tbet⁺) to Th2 (GATA3⁺) CD4⁺ T cells in lymph nodes (left) and spleens (right). Data represent mean ± SD; n = 6 biologically independent animals per group.
PLG vaccines elicit humoral responses against HER2. a) Design of the peptide antigen to target HER2. b) Schematic of the experimental timeline. BALB/c mice were immunized with PLG (PEI‐CpG or free CpG) or bolus vaccines containing MVF‐HER2 and compared to naïve controls. c) IgG1 and d) IgG2a antibody titers against HER2. e) Binding of serum from immunized or control mice to HER2⁺ breast cancer cells. Data represent mean ± SD; n = 10 biologically independent animals per group.
Prophylactic free CpG PLG vaccine protects against infectious disease. a) Schematic of the experimental timeline. BALB/c mice were immunized with PLG (PEI‐CpG or free CpG) or bolus vaccines containing microbead‐immobilized RS218 lysate and compared to naïve controls. Mice were challenged with RS218 E. coli 28 d after immunization. b) Total IgG antibody titer against RS218 E. coli. Data represent mean ± SD; n = 6 (vaccines) or 4 (naïve) biologically independent animals per group. c) Survival and d) endpoint organ bacterial burden of mice challenged with RS218 E. coli. Data represent mean ± SD; n = 8 (vaccines) or 6 (naïve) biologically independent animals per group.
Scaffold Vaccines for Generating Robust and Tunable Antibody Responses

January 2022

·

252 Reads

·

22 Citations

Traditional bolus vaccines often fail to sustain robust adaptive immune responses, typically requiring multiple booster shots for optimal efficacy. Additionally, these provide few opportunities to control the resulting subclasses of antibodies produced, which can mediate effector functions relevant to distinct disease settings. Here, it is found that three scaffold-based vaccines, fabricated from poly(lactide-co-glycolide) (PLG), mesoporous silica rods, and alginate cryogels, induce robust, long-term antibody responses to a model peptide antigen gonadotropin-releasing hormone with single-shot immunization. Compared to a bolus vaccine, PLG vaccines prolong germinal center formation and T follicular helper cell responses. Altering the presentation and release of the adjuvant (cytosine-guanosine oligodeoxynucleotide, CpG) tunes the resulting IgG subclasses. Further, PLG vaccines elicit strong humoral responses against disease-associated antigens HER2 peptide and pathogenic E. coli, protecting mice against E. coli challenge more effectively than a bolus vaccine. Scaffold-based vaccines may thus enable potent, durable and versatile humoral immune responses against disease.


Citations (11)


... [7][8][9] Yet, platforms that allow for controlled, sustained vaccine delivery offer opportunities to further tune vaccine immunogenicity through persistent activation of lymph node germinal centers by sustained antigen release. [10][11][12][13][14] Thus, a high-precision microfabrication method that can be easily chemically and spatially tuned could combine the advantages of injectable hollow particle platforms with the ability to engineer immunogenicity by tuning release rates. ...

Reference:

Nanoscale Biodegradable Printing for Designed Tuneability of Vaccine Delivery Kinetics
Durable lymph-node expansion is associated with the efficacy of therapeutic vaccination

Nature Biomedical Engineering

... Oligonucleotides containing unmethylated guanine cytosine dinucleotide (CpG ODN) can activate T and B lymphocytes, antigen presenting cells (APC), and induce immune cells to produce Th1-type inflammatory cytokines such as IFN-mediated [93,94]. CpG ODN has the ability to activate innate and adaptive immune responses, and has an excellent prospect in adjuvant therapy of head and neck squamous cell carcinoma vaccine. ...

Fine tuning of CpG spatial distribution with DNA origami for improved cancer vaccination

Nature Nanotechnology

... For example, 'bio-instructive materials' can be used to mimic natural ECM properties, and such materials can direct the healing and repair of damaged tissues 4,5 . Tailored biomaterials can also control engineered cell activities to modulate therapeutic T-cell responses in situ and to facilitate immune cell engineering in vivo [6][7][8][9] . Yet despite these advances, synthetic biology strategies for programming customized ECM-dependent signaling activities have remained relatively limited, especially in comparison to tools for sensing and programming responses to cell-cell interactions [10][11][12][13][14] . ...

Adoptive T cell transfer and host antigen-presenting cell recruitment with cryogel scaffolds promotes long-term protection against solid tumors

... While this method preserves the spatial organization of tissues, the analysis is limited to the sample surface and is sacrificial, making it less than ideal for studies aiming to look at both structure and function. Researchers have also used more specific methods, including tension nanoprobes to study lymph node elasticity 27 , tensiometers to study capsule rigidity 28 , 2photon microscopy to study lymph node fibrosis 17 , and nanoindentors to test lymph node viscoelasticity 9,29 . These methods have been helpful in elucidating lymph node biomechanics, but none of these are non-sacrificial nor provide biomechanical properties beyond the surface. ...

Lymph node expansion predicts magnitude of vaccine immune response
  • Citing Preprint
  • October 2022

... The SQB DNA origami nanoparticle used as a vaccine in study was repurposed from a study previously published by our lab 20 . The p8634 scaffold strand used for the SQB was produced in-house as previously described 21 , and the staple strands were purchased from IDT. ...

Optimizing CpG spatial distribution with DNA origami for Th1-polarized therapeutic vaccination
  • Citing Preprint
  • June 2022

... A significant advancement in this field emerged with the development of a cancer vaccine targeting MICA and MICB (MICA/B). This innovative approach induces a coordinated attack by both T cell and NK cell populations, maintaining effectiveness against MHC class I-deficient tumors through the combined action of NK cells and CD4+ T cells (160). ...

A vaccine targeting resistant tumours by dual T cell plus NK cell attack

Nature

... In addition, FUT8-mediated core fucosylation promotes tumor immune evasion by inhibiting the degradation of B7-H3 in TNBC. The combination of the fucose [140,141] Collagen receptor PRTH-101 mAb Preclinical [146] 1192 Glycosylation in the tumor immune response analogue 2F-Fuc [80] with an anti-PD-1 antibody synergistically promotes an antitumor immune response. Moreover, the concomitant administration of O-GlcNAc, which inhibits OSMI4, along with an anti-PD-L1 antibody further bolsters the antitumor immune response [74]. ...

Targeting tumor extracellular matrix activates the tumor-draining lymph nodes

Cancer Immunology and Immunotherapy

... In the past century, over one hundred vaccines have been licensed for clinical use, including therapeutic and preventive vaccines [13,14]. Cancer vaccines deliver tumor-associated antigens in various formulations, such as live-attenuated, inactivated, toxoid, protein subunit/conjugate, or nucleic acid [15]. Vaccine components are taken up by antigen-presenting cells (APCs) and transported to local draining lymph nodes (LNs), where B and T cells detect the antigen and initiate the adaptive immune response. ...

Scaffold Vaccines for Generating Robust and Tunable Antibody Responses

... Force was assessed by pressing the brush against a balance with the angle used in the behavioral test and was ranked into mild pressure in mice. 42 2. Repeat procedure 1 for 3 trials (one trial includes 100-s pressing and 5-min rest). ...

Skeletal muscle regeneration with robotic actuation-mediated clearance of neutrophils

Science Translational Medicine

... Biomaterial mesoporous silica rods loaded with SARS-CoV-2 viral protein, a growth factor (granulocyte-macrophage colony-stimulating factor) and MPLA showed an effective immune response by delayed releases after making subcutaneous scaffoldings. These biomaterials recruited the antigen-presenting cells on the local site and produced a strong adaptive immune response [98]. ...

A Modular Biomaterial Scaffold‐Based Vaccine Elicits Durable Adaptive Immunity to Subunit SARS‐CoV‐2 Antigens