ArticlePublisher preview available

A facile approach to enhance antigen response for personalized cancer vaccination

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Existing strategies to enhance peptide immunogenicity for cancer vaccination generally require direct peptide alteration, which, beyond practical issues, may impact peptide presentation and result in vaccine variability. Here, we report a simple adsorption approach using polyethyleneimine (PEI) in a mesoporous silica microrod (MSR) vaccine to enhance antigen immunogenicity. The MSR-PEI vaccine significantly enhanced host dendritic cell activation and T-cell response over the existing MSR vaccine and bolus vaccine formulations. Impressively, a single injection of the MSR-PEI vaccine using an E7 peptide completely eradicated large, established TC-1 tumours in about 80% of mice and generated immunological memory. When immunized with a pool of B16F10 or CT26 neoantigens, the MSR-PEI vaccine eradicated established lung metastases, controlled tumour growth and synergized with anti-CTLA4 therapy. Our findings from three independent tumour models suggest that the MSR-PEI vaccine approach may serve as a facile and powerful multi-antigen platform to enable robust personalized cancer vaccination.
MSR–PEI vaccine enhances DC activation and trafficking in situ a, Schematic representations of the MSR vaccine (V), boxed in black, and MSR–PEI vaccine (VP), boxed in red. b, Total cell number at the vaccine site explanted on day 3 after immunization with V or VP using B60K PEI (n = 4, two-tailed t-test). c–e, Total number of CD11c⁺ CD86⁺ activated DCs (n = 4, two-tailed t-test) (c), CD11c⁺ CCR7⁺ LN homing DCs (n = 4, two-tailed t-test) (d) and SIINFEKL-presenting DCs (n = 4, two-tailed t-test) (e) recruited to the vaccine site on day 3 after immunization with V or VP using B60K PEI. f–h, Total number of cells (n = 4 for day 3 and n = 5 for day 5, two-way ANOVA) (f), of CD11c⁺ CD86⁺ or CD11c⁺ MHC-II⁺ activated DCs (n = 4 for day 3 and n = 5 for day 5, two-way ANOVA) (g) and OVA⁺ DCs (n = 4 for day 3 and n = 5 for day 5, two-way ANOVA) (h) in the dLN on days 3 and 5 after immunization with V or VP using B60K PEI or left unimmunized (N). i, Schematic representations of the MSR–PEI trans vaccine (trans VP) and the MSR–PEI cis vaccine (cis VP). j, Total number of CD11c⁺ CD86⁺ activated DCs at the vaccine site on day 3 after immunization with the trans VP vaccine or the cis VP vaccine (n = 5, two-tailed t-test). k,l, Total number of CD11c⁺ CD86⁺ activated DCs (k) and CD11c⁺ OVA⁺ DCs (l) in the dLN on day 5 after immunization with the trans VP vaccine or the cis VP vaccine (n = 5, two-tailed t-test). Data depict mean ± s.d.
… 
MSR–PEI vaccine enhances CD8 cytotoxic T-cell response against OVA a, Percentage of IFN-γ⁺ CD8⁺ T cells isolated from peripheral blood on day 7 after immunization with V or VP using B60K PEI or left unimmunized, and stimulated with SIINFEKL (three primary fluorescence-activated cell sorting (FACS) plots on the left, quantifications from the FACS plots on the right) (n = 5, one-way ANOVA). b, Percentage of SIINFEKL-tetramer⁺ CD8⁺ T cells isolated from peripheral blood on day 7 after immunization with V or VP using B60K PEI or left unimmunized (N) (n = 5 for VP, n = 4 for N and V, one-way ANOVA). c, Ratio of CD8⁺ effector T cells (Teff) to CD4⁺ Foxp3⁺ regulatory T cells (Treg) at the MSR vaccine site on day 11 after immunization with V or VP using B60K PEI (n = 5, two-tailed t-test). d, Percentage of IFN-γ⁺ CD8⁺ T cells isolated from peripheral blood on day 7 after immunization with VP containing various doses of B60K PEI or left unimmunized (N) (n = 4, one-way ANOVA). e, Percentage of IFN-γ⁺ CD8⁺ T cells isolated from peripheral blood on day 7 after immunization with V or the MSR-PEI vaccine using L25K PEI (VP L25) or left unimmunized (N) (n = 4, one-way ANOVA). f, Percentage of IFN-γ⁺ CD8⁺ T cells isolated from peripheral blood on day 7 after immunization with V, the MSR-PEI trans vaccine (trans, VP) using B60K PEI or the MSR-PEI cis vaccine (cis, VP) using B60K PEI, or left unimmunized (N), and stimulated with SIINFEKL (n = 9, * between cis VP and trans VP, # between cis VP and V by one-way ANOVA). Data depict mean ± s.d.
… 
MSR–PEI vaccine enhances CD8 cytotoxic T-cell response against E7 and regresses established tumours a,b, Percentage of IFN-γ⁺ CD8⁺ T cells in response to RAHYNIVTF stimulation (a) and percentage of tetramer⁺ CD8⁺ T cells (b) in peripheral blood on day 7 after immunization with the MSR E7 vaccine (V) or the MSR–PEI E7 vaccine (VP) using 5 μg or 20 μg of B60K PEI, or left unimmunized (N) (n = 4, one-way ANOVA). c, ELISA analysis of TNF-α level in serum 24 h after vaccination with V or the MSR-PEI (B60K) vaccine (VP), or left unimmunized (N) (n = 4, compared with N by one-way ANOVA). d,e, Tumour growth (d) and overall survival (e) of mice bearing established E7-expressing TC-1 tumours (allowed to develop for 8 days) and treated with V or VP using L25K PEI, or left untreated (N), and subsequently rechallenged with TC-1 cells 6 months after the first inoculation (n = 10, compared with V by two-way ANOVA for d and by log-rank test for e). f, Overall survival of mice bearing established E7-expressing TC-1 tumours and treated with the MSR–PEI vaccine containing E7 (E7 VP) or the MSR–PEI vaccine containing SIINFEKL (SIINFEKL VP), or left untreated (Naive) (n = 8, compared with SIINFEKL VP by log-rank test). g, Flow cytometry analysis of blood T cells 3 days after treatment with a-CD8a monoclonal antibody (mab) or an isotype monoclonal antibody (representative data, repeated three times). h, Tumour growth of mice bearing established E7-expressing TC-1 tumours and treated with the MSR–PEI vaccine with either a-CD8a monoclonal antibody or an isotype monoclonal antibody (n = 8, compared with VP a-CD8 by two-way ANOVA). In a–c data depict mean ± s.d. and in d,h data depict mean ± s.e.m.
… 
MSR–PEI vaccine enhances melanoma TIL effector function and induces tumour control and synergy with anti-CTLA4 therapy using combined B16 neoantigens a, Number of CD44⁺ IFN-γ⁺, CD44⁺ TNF-α⁺ and CD44⁺ granzyme B⁺ TILs per 500,000 tumour cells on day 15 after inoculation. Mice bearing established B16F10 tumours (allowed to develop for 5 days) and treated with the MSR vaccine (V) or the MSR–PEI vaccine (VP) using L25K PEI and 50 μg of the B16 neoantigens, or left untreated (N) (n = 5 for VP, n = 3 for N and V, one-way ANOVA). b, Number of lung metastases formed on day 16 after inoculation in mice that received IV inoculation of B16F10 melanoma cells (allowed to develop for 1 day) and were treated with VP using L25K PEI and 50 μg of the B16 neoantigens, or left untreated (N). Primary representative photographs of excised lungs are shown (n = 6, two-tailed t-test). c, Tumour growth in mice bearing established B16F10 tumours (allowed to develop for 3 days) and treated with two injections of VP using L25K PEI and 50 μg of the B16 neoantigens on days 3 and 13, or left untreated (N) (n = 8, two-tailed t-test). d, Tumour volume change between days 13 and 17 after tumour inoculation (n = 8, two-tailed t-test). e, Tumour growth of mice bearing established B16F10 tumours (inoculated with 1 × 10⁵ cells) and treated with anti-CTLA4 antibody (a-CTLA4), anti-CTLA4 antibody in combination with the MSR–PEI vaccine (VP + a-CTLA4) using L25K PEI and 50 μg of the B16 neoantigens on day 5, or left untreated (N) (n = 8, *significant difference between VP + a-CTLA4 and a-CTLA4, ***significant difference between VP + a-CTLA4, ns between a-CTLA4 and N by one-way ANOVA). In a,b,d data depict mean ± s.d., in c,e data depict individual tumour growth.
… 
This content is subject to copyright. Terms and conditions apply.
Letters
https://doi.org/10.1038/s41563-018-0028-2
1John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA. 2Wyss Institute for Biologically Inspired
Engineering, Harvard University, Boston, MA, USA. 3Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.
4School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea. 5Department of Health Sciences and Technology, Samsung
Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Suwon, Republic of Korea. 6Biomedical Institute for Convergence
at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea. *e-mail: mooneyd@seas.harvard.edu
Existing strategies to enhance peptide immunogenicity for
cancer vaccination generally require direct peptide altera-
tion, which, beyond practical issues, may impact peptide pre-
sentation and result in vaccine variability. Here, we report a
simple adsorption approach using polyethyleneimine (PEI)
in a mesoporous silica microrod (MSR) vaccine to enhance
antigen immunogenicity. The MSR–PEI vaccine significantly
enhanced host dendritic cell activation and T-cell response
over the existing MSR vaccine and bolus vaccine formulations.
Impressively, a single injection of the MSR–PEI vaccine using
an E7 peptide completely eradicated large, established TC-1
tumours in about 80% of mice and generated immunological
memory. When immunized with a pool of B16F10 or CT26 neo-
antigens, the MSR–PEI vaccine eradicated established lung
metastases, controlled tumour growth and synergized with
anti-CTLA4 therapy. Our findings from three independent
tumour models suggest that the MSR-PEI vaccine approach
may serve as a facile and powerful multi-antigen platform to
enable robust personalized cancer vaccination.
Cancer vaccines targeting multiple tumour-specific antigens
can elicit broad immune responses and decrease tumour escape1,2,
and recent advances enable identification of tumour-specific muta-
tions (‘neoantigens’)3,4. Neoantigens are attractive vaccine targets
as they are not expressed in healthy tissues and are predicted to
have strong major histocompatibility complex (MHC)-binding
affinity5. Recent clinical data have shown that neoantigen vaccines
could generate T cells that specifically target heterogeneous tumour
clones6. However, neoantigen peptides exhibit rapid clearance and
low immunogenicity, which limits optimal presentation by antigen-
presenting cells to initiate strong T-cell responses7. Macro- and
nano-engineering strategies have been designed to overcome these
challenges811, but many approaches require chemical modification
or physical emulsification of the peptides, potentially altering their
presentation capacity. Moreover, since neoantigen vaccines typi-
cally require many peptides12, modification of individual peptides
is cumbersome for clinical translation and is likely to result in high
batch-to-batch variability.
We propose a facile strategy to enhance antigen immunogenicity
using polyethyleneimine (PEI) combined with a mesoporous silica
microrod (MSR) vaccine. The MSR vaccine can be injected using
standard needles, was shown to effectively concentrate and acti-
vate large populations of host antigen-presenting cells and induced
more potent humoral responses and prophylactic tumour protec-
tion than traditional vaccine formulations13. Moreover, the MSR
surface could potentially be modified to induce stronger responses.
Recent studies have shown that complexes based on PEI, a widely
used cationic polymer14,15, can stimulate pro-inflammatory cytokine
production16,17, and induce potent humoral responses when com-
plexed with glycoproteins18. Here, we explore the application of PEI
to co-present an antigen in a facile, layered adsorption manner in
the MSR vaccine.
MSRs were adsorbed with PEI (MSR–PEI) by simply mixing
with a PEI solution for 15 minutes; subsequently, an antigen pool
was directly adsorbed onto MSR–PEI particles (Fig. 1a). Both rela-
tive molecular mass 60,000 (60K) branched PEI (B60K) and 25K
linear PEI (L25K) absorbed to MSR with high efficiency (Fig. 1b),
with an incorporation capacity of about 20 μ g PEI per mg MSR.
Over 90% of B60K and L25K PEI polymers were adsorbed after
1 min of mixing (Fig. 1c). Zeta potential measurements confirmed
PEI incorporation (Fig. 1d). MSR–PEIs maintained the intrinsic
mesopores of MSRs (Supplementary Fig. 1a), pore structure and
bulk particle structure (Supplementary Fig. 1b–d), with reduced
surface area and pore volume as expected (Supplementary Table 1).
MSRs and MSR–PEIs showed high incorporation efficiency for
net positive and neutral example murine (Fig. 1e) and human
(Fig. 1f) peptides, but MSR–PEI enhanced the incorporation of net
negative peptides.
The underlying adjuvant effect of PEI19,20 on bone-marrow-
derived dendritic cells (BMDCs) was next examined. BMDCs take up
free PEI, reaching maximum uptake at 24 h (Supplementary Fig. 2a),
and showed a significant increase in CD86 and MHC-II expression
(Fig. 1f) and tumour necrosis factor α (TNF-α ) (Fig. 1g) and inter-
leukin-6 (IL-6) (Fig. 1h) production in a PEI dose-dependent man-
ner. BMDCs stimulated with MSR–PEI also showed significantly
increased CD86 expression (Supplementary Fig. 2b) and TNF-α
production (Supplementary Fig. 2c). MSR–PEI also triggered the
increased production of IL-1β , a key cytokine produced in response
to Nlrp3 inflammasome activation21 (Supplementary Fig. 3a). This
was probably a result of lysosomal rupture upon MSR–PEI uptake
(Supplementary Fig. 3b), leading to the release of phagosomal con-
tents into the cytosolic compartment. Interestingly, as TNF-α and
IL-6 have been shown to be Nlrp3-independent cytokines22, it is
possible that MSR–PEI particles can stimulate multiple damage-
associated molecular pattern (DAMP) receptors. The impact of PEI
A facile approach to enhance antigen response for
personalized cancer vaccination
Aileen Weiwei Li1,2, Miguel C. Sobral1,2, Soumya Badrinath3, Youngjin Choi4, Amanda Graveline2,
Alexander G. Stafford2, James C. Weaver2, Maxence O. Dellacherie1,2, Ting-Yu Shih1,2, Omar A. Ali2,
Jaeyun Kim4,5,6, Kai W. Wucherpfennig3 and David J. Mooney1,2*
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NATURE MATERIALS | VOL 17 | JUNE 2018 | 528–534 | www.nature.com/naturematerials
528
The Nature trademark is a registered trademark of Springer Nature Limited.
... Although subunit vaccines have great potential in cancer immunotherapy, the efficiency of antigen and adjuvants combined delivery to lymph nodes (LNs) is relatively low, 2,3 leading to weak immune stimulation and tolerance. In addition, in order to effectively induce CTLs mediated cellular immunity, antigens must enter APCs and then combine with major histocompatibility complex for synergistically activating APCs; 11,12 3) nanocarriers with small size increasing the aggregation of antigens in LNs, 13 further enhancing the immune response. ...
... This reaction mainly includes three important factors: 1) spatiotemporal control of antigen delivery to LNs; 25,34,35 2) cytoplasmic delivery and cross presentation of antigen in APCs; [36][37][38] 3) combined delivery of antigens and adjuvants. 11,12 The OVA@MMSNs@BM-Man nanovaccine meets the requirements of the above three factors. The prepared OVA@MMSNs@BM-Man nanovaccine can target DCs in LNs through the combination of mannose with CD205. ...
Article
Full-text available
Introduction Tumor vaccines can activate tumor-specific immune responses to inhibit tumor growth, recurrence, and metastasis. However, the efficiency of antigen and adjuvants combined delivery to lymph nodes (LNs) is relatively low, leading to weak immune stimulation and tolerance. In this study, a tumor nanovaccine was constructed for the targeted dendritic cells (DCs)-mediated immunotherapy. Methods Ovalbumin (OVA) antigen was first loaded into manganese-doped mesoporous silica nanoparticles (MMSNs) and coated with bacterial cytoplasmic membrane (BM), which was further inserted with mannose to prepare OVA@MMSNs@BM-Man nanovaccines. In vitro and in vivo experiments were conducted to assess their properties and function of the synthesized nanovaccines. Results The nanovaccine can effectively target DCs in LNs by the combination of mannose with mannose receptor (CD205). BM serves as an immune adjuvant and co-delivers with OVA antigen, effectively improving antigen presentation efficiency. In an acidic environment, the Mn²⁺ produced by the degradation of MMSNs can not only serve as an MR imaging agent but also activate the cGAS-STING pathway, followed by the release of IFN-β. The activated DCs further activate the body’s cytotoxic T cells (CTLs), thereby exerting anti-tumor effects. The conclusion This study will provide a new idea for the construction of tumor nanovaccines.
... The self-adjuvated nanofiber vaccines were fabricated by coating Salmonella-derived flagella with cationic polyethyleneimine (PEI) and the model antigen of OVA through a layer-by-layer selfassembly nanotechnology. [26] Flagella obtained from VNP20009 (termed as FLA) were isolated and purified from its culture medium by a set of procedures including acidification, salting out, and ultracentrifugation ( Figure S1, Supporting Information). By simply mixing with negatively-charged FLA, positivelycharged PEI could be deposited onto flagellum surface via electrostatic interaction. ...
Article
Full-text available
Bacteria‐based vaccines have received increasing attention given the ability to induce strong systemic immune responses. However, the application of bacteria as therapeutic agents inevitably suffers from infection‐associated side effects due to the living characteristics. Here, the use of bacteria‐derived flagella is described to construct self‐adjuvated nanofiber vaccines. With the help of charge‐reversal mediated by decoration with cationic polymers, the flagella can be coated with negatively charged antigens through electrostatic interaction. By virtue of the large aspect ratio, the resulting nanofiber vaccines show prolonged retention at the injection site and increased uptake by dendritic cells and macrophages. Thanks to the innate immunogenicity, self‐adjuvated flagella robustly promote dendritic cell maturation and macrophage polarization, resulting in the elicitation of antigen‐specific T‐cell and B‐cell immune responses. In ovalbumin‐overexpressing melanoma‐bearing mice, immunization with ovalbumin‐carried vaccines not only exhibits a favorable tolerance, but also displays superior inhibition efficacies on tumor growth and metastasis separately under the therapeutic and prophylactic settings. The flexibility of this approach is further demonstrated for vaccine fabrication by coating with the SARS‐CoV‐2 Spike protein S1 subunit. Bacterial flagella‐based self‐adjuvated nanofiber platform proposes a versatile strategy to develop various vaccines for disease prevention and treatment.
... Wild-type B16F10 cells were used to construct the tumour model. A mixture of mutation-derived neoantigen peptides (B16-M27, B16-M30, B16-M47 and B16-M48) were used to prepare various vaccines 54 . XMVs encapsulating B16 neoantigen peptides (XMV-B16), AUVs encapsulating B16 neoantigen peptides (AUV-B16), LIPs encapsulating B16 neoantigen peptides (LIP-B16) or free B16 neoantigen peptides (Free B16) were s.c. ...
Article
Full-text available
Targeting the delivery of vaccines to dendritic cells (DCs) is challenging. Here we show that, by mimicking the fast and strong antigen processing and presentation that occurs during the rejection of xenotransplanted tissue, xenogeneic cell membrane-derived vesicles exposing tissue-specific antibodies can be leveraged to deliver peptide antigens and mRNA-encoded antigens to DCs. In mice with murine melanoma and murine thymoma, xenogeneic vesicles encapsulating a tumour-derived antigenic peptide or coated on lipid nanoparticles encapsulating an mRNA coding for a tumour antigen elicited potent tumour-specific T-cell responses that inhibited tumour growth. Mice immunized with xenogeneic vesicle-coated lipid nanoparticles encapsulating an mRNA encoding for the spike protein of severe acute respiratory syndrome coronavirus 2 elicited titres of anti-spike receptor-binding domain immunoglobulin G and of neutralizing antibodies that were approximately 32-fold and 6-fold, respectively, those elicited by a commercialized mRNA–lipid nanoparticle vaccine. The advantages of mimicking the biological recognition between immunoglobulin G on xenogeneic vesicles and fragment crystallizable receptors on DCs may justify the assessment of the safety risks of using animal-derived biological products in humans.
... Various nanomaterials have been used as delivery systems to improve the stability of TSAs. Polyethyleneimine-coated silica microrods can augment the immunogenicity of HPV-16 E7 and several peptides carrying novel antigens, with remarkable efficacy in tumor treatment in murine models [26]. Furthermore, conventional nanovaccine delivery systems often rely on nanocarriers, which can impede the effective delivery of new antigens and potentially result in significant toxicity. ...
Research
Full-text available
Tumor vaccines have become a crucial strategy in cancer immunotherapy. Challenges of traditional tumor vaccines include inadequate immune activation and low efficacy of antigen delivery. Nanoparticles, with their tunable properties and versatile functionalities, have redefined the landscape of tumor vaccine design. In this review, we outline the multifaceted roles of nanoparticles in tumor vaccines, ranging from their capacity as delivery vehicles to their function as immunomodulatory adjuvants capable of stimulating anti-tumor immunity. We discuss how this innovative approach significantly boosts antigen presentation by leveraging tailored nanoparticles that facilitate efficient uptake by antigen-presenting cells. These nanoparticles have been meticulously designed to overcome biological barriers, ensuring optimal delivery to lymph nodes and effective interaction with the immune system. Overall, this review highlights the transformative power of nanotechnology in redefining the principles of tumor vaccines. The intent is to inform more efficacious and precise cancer im-munotherapies. The integration of these advanced nanotechnological strategies should unlock new frontiers in tumor vaccine development, enhancing their potential to elicit robust and durable anti-tumor immunity.
Article
Full-text available
Nanovaccines, as a new generation of vaccines, have garnered significant interest due to their exceptional potential in enhancing disease prevention and treatment. Their unique features, such as high stability, antigens protection, prolonged retention, and targeted delivery to lymph nodes, immune cells, and tumors, set them apart as promising candidates in the field of immunotherapy. Polymers, with their superior degradability, capacity to mimic pathogen characteristics, and surface functionality that facilitates modifications, serve as ideal carriers for vaccine components. Polymer‐based self‐adjuvanted nanovaccines have the remarkable ability to augment immune responses. The inherent adjuvant‐like properties of polymers themselves offer a pathway toward more efficient exploitation of nanomaterials and the optimization of nanovaccines. This review article aims to summarize the categorization of polymers and elucidate their mechanisms of action as adjuvants. Additionally, it delves into the advantages and limitations of polymer‐based self‐adjuvanted nanovaccines in disease management and prevention, providing valuable insights for their design and application. This comprehensive analysis could contribute to the development of more effective and tailored nanovaccines for a wide range of diseases.
Article
Cancer immunotherapy has shifted the paradigm for clinical cancer treatment in the past decade, especially with the success of checkpoint blockades and chimeric antigen receptor (CAR)-T cell therapy. However, the low patient response rate to checkpoint blockades, poor efficacy of CAR-T cell therapy against solid tumors, and severe side effects in both have limited the utility of cancer immunotherapy. These issues motivate the development of new immunotherapies that can induce persistent cytotoxic T lymphocyte response with minimal side effects. This often requires the targeted modulation of specific types of immune cells (e.g., dendritic cells and T cells) in lymphatic tissues or cancerous tissues, which is inevitably challenging. Immune cell homing materials, though, enable in situ recruitment and modulation of immune cells for the orchestration of systemic immune responses and overall antitumor efficacy. Here we introduce the design, synthesis, characterization, and immune analysis of dendritic cell-homing macroporous hydrogels for the development of cancer immunotherapy.
Chapter
A promising method for treating cancer, autoimmune disorders, and infectious diseases is immunotherapy. The creation of efficient biomaterials to deliver and boost the activity of immunotherapeutic drugs, however, is a key factor in the clinical success of immunotherapy. New chances to design immunotherapeutic platforms with increased efficacy and safety have been made possible by recent advancements in biomaterials. These biomaterials can control immune cell activity, exert enzyme-like action, neutralise cytokines, and cure inflammation, malignancies, autoimmune illnesses, etc. The utilisation of nanoparticle-based delivery systems designed immune cell treatments, and scaffold materials for immune tissue engineering are a few important advancements. The precise targeting of immune cells and the regulated delivery of immunotherapeutic drugs are both possible with nanoparticle engineering. Biomaterials are used in engineered immune cell therapies, such as CAR-T cells, to increase the persistence and effectiveness of immune cells. Scaffold materials can be utilised to create immunological tissues and support immune cell growth and function. These biomaterials can improve clinical outcomes of immunotherapeutic drugs by increasing their potency, specificity, and durability. Biomaterials for immunotherapeutic applications are developing quickly and show significant promise for the immunotherapy field’s future. This chapter discusses current developments in biomaterials for immunotherapeutic uses and their prospective effects on immunotherapy in the future.
Article
Full-text available
Effective anti-tumour immunity in humans has been associated with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumour-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumour immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumours, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) molecules. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoural balance in favour of enhanced tumour control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumour neoantigens. Vaccine-induced polyfunctional CD4(+) and CD8(+) T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, respectively. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumour. Of six vaccinated patients, four had no recurrence at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumour regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.
Article
Full-text available
Our evolving understandings of cell-material interactions provide insights for using polymers to modulate cell behaviour that may lead to therapeutic applications. It is known that in certain cancers, myeloid-derived suppressor cells (MDSCs) play vital roles in promoting tumour progression, chiefly because of their 'alternatively activated' (or M2) phenotype that orchestrates immunosuppression. In this study, we demonstrated that two cationic polymers - cationic dextran (C-dextran) and polyethyleneimine (PEI) - could directly remodel these cells into an anti-tumour, 'classically activated' (or M1) phenotype, thereby stimulating these cells to express tumouricidal cytokines, reactivating the T cell functions, and prolonging the lifespan of the mice model. Our investigations with knock-out mice further indicate that the functions of these cationic polymers require the involvement of toll-like receptor 4-mediated signalling. Taken together, our study suggests that these cationic polymers can effectively and directly re-polarize MDSCs from an immunosuppressive characteristic to an anti-tumour phenotype, leading to successful restoration of immune surveillance in the tumour microenvironment and elimination of tumour cells. Our findings may have immediate impact on further development of polymer-based therapeutics for cancer immunotherapy.
Article
Full-text available
Therapeutic vaccination with human papillomavirus type 16 synthetic long peptides (HPV16-SLPs) results in T cell-mediated regression of HPV16-induced premalignant lesions but fails to install clinically effective immunity in patients with HPV16-positive cervical cancer. We explored whether HPV16-SLP vaccination can be combined with standard carboplatin and paclitaxel chemotherapy to improve immunity and which time point would be optimal for vaccination. This was studied in the HPV16 E6/E7-positive TC-1 mouse tumor model and in patients with advanced cervical cancer. In mice and patients, the presence of a progressing tumor was associated with abnormal frequencies of circulating myeloid cells. Treatment of TC-1-bearing mice with chemotherapy and therapeutic vaccination resulted in superior survival and was directly related to a chemotherapymediated altered composition of the myeloid cell population in the blood and tumor. Chemotherapy had no effect on tumor-specific T cell responses. In advanced cervical cancer patients, carboplatin-paclitaxel also normalized the abnormal numbers of circulating myeloid cells, and this was associated with increased T cell reactivity to recall antigens. The effect was most pronounced starting 2 weeks after the second cycle of chemotherapy, providing an optimal immunological window for vaccination. This was validated with a single dose of HPV16-SLP vaccine given in this time window. The resulting proliferative HPV16-specific T cell responses were unusually strong and were retained after all cycles of chemotherapy. In conclusion, carboplatin-paclitaxel therapy fosters vigorous vaccineinduced T cell responses when vaccination is given after chemotherapy and has reset the tumor-induced abnormal myeloid cell composition to normal values.
Article
Full-text available
Therapeutic vaccines preferentially stimulate T cells against tumour-specific epitopes that are created by DNA mutations or oncogenic viruses. In the setting of premalignant disease, carcinoma in situ or minimal residual disease, therapeutic vaccination can be clinically successful as monotherapy; however, in established cancers, therapeutic vaccines will require co-treatments to overcome immune evasion and to become fully effective. In this Review, we discuss the progress that has been made in overcoming immune evasion controlled by tumour cell-intrinsic factors and the tumour microenvironment. We summarize how therapeutic benefit can be maximized in patients with established cancers by improving vaccine design and by using vaccines to increase the effects of standard chemotherapies, to establish and/or maintain tumour-specific T cells that are re-energized by checkpoint blockade and other therapies, and to sustain the antitumour response of adoptively transferred T cells.
Article
Full-text available
Significance Nearly all cervical, anal, vulvar, and penile cancer and up to half of oropharyngeal cancers are driven by the E6 and E7 oncoproteins of human papilloma virus (HPV). Therapeutic vaccination against these HPV proteins can slow disease progression in animal models and in patients, but is rarely curative. We demonstrate that coadministration of agonist antibodies targeting the T-cell costimulatory receptor 4-1BB and an intranasal HPV E6/E7 peptide vaccine promoted durable regression in 100% of animals bearing HPV ⁺ TC-1 tumors established in the female reproductive tract. The efficacy of 4-1BB in this system was unique among immune checkpoint antibodies and provides a paradigm for enhancement of therapeutic cancer vaccines with costimulatory agonist antibodies.
Article
Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte–associated protein (CTLA)-4 or programmed cell death 1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors in experimental settings typically viewed as intractable.
Article
Emerging clinical evidence on the role of the antitumor activity of the immune system has generated great interest in immunotherapy in all cancer types. Recent clinical data clearly demonstrated that human tumor cells express antigenic peptides (epitopes) that can be recognized by autologous tumor-specific T cells and that enhancement of such immune reactivity can potentially lead to cancer control and cancer regression in patients with advanced disease. However, in most cases, it is unclear which tumor antigens (Ags) mediated cancer regression. Mounting evidence indicates that numerous endogenous mutated cancer proteins, a hallmark of tumor cells, can be processed into peptides and presented on the surface of tumor cells, leading to their immune recognition in vivo as "non-self" or foreign. Massively parallel sequencing has now overcome the challenge of rapidly identifying the comprehensive mutational spectrum of individual tumors (i.e., the "mutanome") and current technologies, as well as computational tools, have emerged that allow the identification of private epitopes derived from their mutanome and called neoantigens (neoAgs). On this basis, both CD4⁺ and CD8⁺ neoantigen-specific T cells have been identified in multiple human cancers and shown to be associated with a favorable clinical outcome. Notably, emerging data also indicate that neoantigen recognition represents a major factor in the activity of clinical immunotherapies. In the post-genome era, the mutanome holds promise as a long-awaited 'gold mine' for the discovery of unique cancer cell targets, which are exclusively tumor-specific and unlikely to drive immune tolerance, hence offering the chance for highly promising clinical programs of cancer immunotherapy.
Article
Lymphoid organs, in which antigen presenting cells (APCs) are in close proximity to T cells, are the ideal microenvironment for efficient priming and amplification of T-cell responses. However, the systemic delivery of vaccine antigens into dendritic cells (DCs) is hampered by various technical challenges. Here we show that DCs can be targeted precisely and effectively in vivo using intravenously administered RNA-lipoplexes (RNA-LPX) based on well-known lipid carriers by optimally adjusting net charge, without the need for functionalization of particles with molecular ligands. The LPX protects RNA from extracellular ribonucleases and mediates its efficient uptake and expression of the encoded antigen by DC populations and macrophages in various lymphoid compartments. RNA-LPX triggers interferon-α (IFNα) release by plasmacytoid DCs and macrophages. Consequently, DC maturation in situ and inflammatory immune mechanisms reminiscent of those in the early systemic phase of viral infection are activated. We show that RNA-LPX encoding viral or mutant neo-antigens or endogenous self-antigens induce strong effector and memory T-cell responses, and mediate potent IFNα-dependent rejection of progressive tumours. A phase I dose-escalation trial testing RNA-LPX that encode shared tumour antigens is ongoing. In the first three melanoma patients treated at a low-dose level, IFNα and strong antigen-specific T-cell responses were induced, supporting the identified mode of action and potency. As any polypeptide-based antigen can be encoded as RNA, RNA-LPX represent a universally applicable vaccine class for systemic DC targeting and synchronized induction of both highly potent adaptive as well as type-I-IFN-mediated innate immune mechanisms for cancer immunotherapy.
Article
s: CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY Mutations are regarded as ideal targets for cancer immunotherapy. As neoepitopes with strict lack of expression in any healthy tissue, they are expected to be safe and could bypass the central tolerance mechanisms. Recent advances in nucleic acid sequencing technologies have revolutionized the field of genomics, allowing the readily targeting of mutated neoantigens for personalized cancer vaccination. We demonstrated in three independent murine tumor models that a considerable fraction of non-synonymous cancer mutations is immunogenic and that unexpectedly the immunogenic mutanome is pre-dominantly recognized by CD4+ T cells. RNA vaccination with such MHC class II restricted immunogenic mutations leads to infiltration of CD4+ and CD8+ T cells into the tumor, reduces intratumoral regulatory T cells and ultimately confers strong anti-tumor activity. Encouraged by these findings we set up a process comprising mutation detection by exome sequencing, selection of vaccine targets by solely bioinformatical prioritization of mutated epitopes predicted to be abundantly expressed and presented on MHC class II molecules. Synthetic mRNA vaccines encoding multiple of these prioritized mutated epitopes induce potent tumor control and complete rejection of established aggressively growing tumors in mice. Moreover, we demonstrate that CD4+ T cell neoepitope vaccination primes CTL responses against an independent immunodominant antigen in tumor bearing mice indicating orchestration of antigen spread. Our findings reveal that cancer mutation based MHC class II restricted epitopes are attractive vaccination targets and provide the preclinical proof of concept for an integrated process from tumor sample to a cancer vaccine customized to the unique repertoire of each patient`s tumor. Citation Format: Mathias Vormehr, Sebastian Kreiter, Niels van de Roemer, Mustafa Diken, Martin Lower, Fulvia Vascotto, Jan Diekmann, Sebastian Boegel, Barbara Schroers, Arbel D. Tadmor, Ozlem Tureci, Ugur Sahin. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A110.
Article
Biomaterial scaffold based vaccines show significant potential in generating potent antigen-specific immunity. However, the role of the scaffold surface chemistry in initiating and modulating the immune response is not well understood. In this study, a mesoporous silica micro-rod (MSR) scaffold was modified with PEG, PEG-RGD and PEG-RDG groups. PEG modification significantly enhanced BMDC activation marker up-regulation and IL-1β production in vitro, and innate immune cell infiltration in vivo. PEG-RGD MSRs and PEG-RDG MSRs displayed decreased inflammation compared to PEG MSRs, and the effect was not RGD specific. Finally, the Nlrp3 inflammasome was found to be necessary for MSR stimulated IL-1ß production in vitro and played a key role in regulating immune cell infiltration in vivo. These findings suggest that simply modulating the surface chemistry of a scaffold can regulate its immune cell infiltration profile and have implications for the design and development of new material based vaccines.