Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep Med ; 4(8): 101145, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37552990

ABSTRACT

Immune checkpoint immunotherapy (ICI) can re-activate immune reactions against neoantigens, leading to remarkable remission in cancer patients. Nevertheless, only a minority of patients are responsive to ICI, and approaches for prediction of responsiveness are needed to improve the success of cancer treatments. While the tumor mutational burden (TMB) correlates positively with responsiveness and survival of patients undergoing ICI, the influence of the subcellular localizations of the neoantigens remains unclear. Here, we demonstrate in both a mouse melanoma model and human clinical datasets of 1,722 ICI-treated patients that a high proportion of membrane-localized neoantigens, particularly at the plasma membrane, correlate with responsiveness to ICI therapy and improved overall survival across multiple cancer types. We further show that combining membrane localization and TMB analyses can enhance the predictability of cancer patient response to ICI. Our results may have important implications for establishing future clinical guidelines to direct the choice of treatment toward ICI.


Subject(s)
Immunotherapy , Melanoma , Animals , Humans , Mice , Biomarkers, Tumor/metabolism , Immunotherapy/methods , Melanoma/therapy
2.
Sci Adv ; 7(13)2021 03.
Article in English | MEDLINE | ID: mdl-33762337

ABSTRACT

In melanoma, the induction of lymphatic growth (lymphangiogenesis) has long been correlated with metastasis and poor prognosis, but we recently showed it can synergistically enhance cancer immunotherapy and boost T cell immunity. Here, we develop a translational approach for exploiting this "lymphangiogenic potentiation" of immunotherapy in a cancer vaccine using lethally irradiated tumor cells overexpressing vascular endothelial growth factor C (VEGF-C) and topical adjuvants. Our "VEGFC vax" induced extensive local lymphangiogenesis and promoted stronger T cell activation in both the intradermal vaccine site and draining lymph nodes, resulting in higher frequencies of antigen-specific T cells present systemically than control vaccines. In mouse melanoma models, VEGFC vax elicited potent tumor-specific T cell immunity and provided effective tumor control and long-term immunological memory. Together, these data introduce the potential of lymphangiogenesis induction as a novel immunotherapeutic strategy to consider in cancer vaccine design.


Subject(s)
Cancer Vaccines , Melanoma , Animals , Lymphangiogenesis/physiology , Lymphatic Metastasis , Melanoma/pathology , Mice , T-Lymphocytes/metabolism , Vascular Endothelial Growth Factor C/metabolism
3.
Article in English | MEDLINE | ID: mdl-32117911

ABSTRACT

Therapeutic cancer vaccines constitute a valuable tool to educate the immune system to fight tumors and prevent cancer relapse. Nevertheless, the number of cancer vaccines in the clinic remains very limited to date, highlighting the need for further technology development. Recently, cancer vaccines have been improved by the use of materials, which can strongly enhance their intrinsic properties and biodistribution profile. Moreover, vaccine efficacy and safety can be substantially modulated through selection of the site at which they are delivered, which fosters the engineering of materials capable of targeting cancer vaccines to specific relevant sites, such as within the tumor or within lymphoid organs, to further optimize their immunotherapeutic effects. In this review, we aim to give the reader an overview of principles and current strategies to engineer therapeutic cancer vaccines, with a particular focus on the use of site-specific targeting materials. We will first recall the goal of therapeutic cancer vaccination and the type of immune responses sought upon vaccination, before detailing key components of cancer vaccines. We will then present how materials can be engineered to enhance the vaccine's pharmacokinetic and pharmacodynamic properties. Finally, we will discuss the rationale for site-specific targeting of cancer vaccines and provide examples of current targeting technologies.

4.
Cancer Immunol Res ; 6(11): 1301-1313, 2018 11.
Article in English | MEDLINE | ID: mdl-30131378

ABSTRACT

Treatment of patients bearing human papillomavirus (HPV)-related cancers with synthetic long-peptide (SLP) therapeutic vaccines has shown promising results in clinical trials against premalignant lesions, whereas responses against later stage carcinomas have remained elusive. We show that conjugation of a well-documented HPV-E7 SLP to ultra-small polymeric nanoparticles (NP) enhances the antitumor efficacy of therapeutic vaccination in different mouse models of HPV+ cancers. Immunization of TC-1 tumor-bearing mice with a single dose of NP-conjugated E7LP (NP-E7LP) generated a larger pool of E7-specific CD8+ T cells with increased effector functions than unconjugated free E7LP. At the tumor site, NP-E7LP prompted a robust infiltration of CD8+ T cells that was not accompanied by concomitant accumulation of regulatory T cells (Tregs), resulting in a higher CD8+ T-cell to Treg ratio. Consequently, the amplified immune response elicited by the NP-E7LP formulation led to increased regression of large, well-established tumors, resulting in a significant percentage of complete responses that were not achievable by immunizing with the non-NP-conjugated long-peptide. The partial responses were characterized by distinct phases of regression, stable disease, and relapse to progressive growth, establishing a platform to investigate adaptive resistance mechanisms. The efficacy of NP-E7LP could be further improved by therapeutic activation of the costimulatory receptor 4-1BB. This NP-E7LP formulation illustrates a "solid-phase" antigen delivery strategy that is more effective than a conventional free-peptide ("liquid") vaccine, further highlighting the potential of using such formulations for therapeutic vaccination against solid tumors. Cancer Immunol Res; 6(11); 1301-13. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Nanoparticles/chemistry , Papillomavirus E7 Proteins/chemistry , Animals , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/chemistry , Female , Lung Neoplasms/secondary , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Recurrence, Local , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/therapy , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Vaginal Neoplasms/immunology , Vaginal Neoplasms/pathology , Vaginal Neoplasms/prevention & control
5.
Sci Transl Med ; 9(407)2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28904226

ABSTRACT

In melanoma, vascular endothelial growth factor-C (VEGF-C) expression and consequent lymphangiogenesis correlate with metastasis and poor prognosis. VEGF-C also promotes tumor immunosuppression, suggesting that lymphangiogenesis inhibitors may be clinically useful in combination with immunotherapy. We addressed this concept in mouse melanoma models with VEGF receptor-3 (VEGFR-3)-blocking antibodies and unexpectedly found that VEGF-C signaling enhanced rather than suppressed the response to immunotherapy. We further found that this effect was mediated by VEGF-C-induced CCL21 and tumor infiltration of naïve T cells before immunotherapy because CCR7 blockade reversed the potentiating effects of VEGF-C. In human metastatic melanoma, gene expression of VEGF-C strongly correlated with CCL21 and T cell inflammation, and serum VEGF-C concentrations associated with both T cell activation and expansion after peptide vaccination and clinical response to checkpoint blockade. We propose that VEGF-C potentiates immunotherapy by attracting naïve T cells, which are locally activated upon immunotherapy-induced tumor cell killing, and that serum VEGF-C may serve as a predictive biomarker for immunotherapy response.


Subject(s)
Immunotherapy , Lymphangiogenesis , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , T-Lymphocytes/pathology , Animals , Cell Proliferation , Chemokine CCL21/metabolism , Disease-Free Survival , Epitopes/immunology , Humans , Melanoma, Experimental/pathology , Mice , Neoplasm Metastasis , Receptors, CCR7/metabolism , Signal Transduction , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
6.
Biomaterials ; 132: 48-58, 2017 07.
Article in English | MEDLINE | ID: mdl-28407494

ABSTRACT

Nanoparticle delivery systems are known to enhance the immune response to soluble antigens (Ags) and are thus a promising tool for the development of new vaccines. Our laboratory has engineered two different nanoparticulate systems in which Ag is either encapsulated within the core of polymersomes (PSs) or decorated onto the surface of nanoparticles (NPs). Previous studies showed that PSs are better at enhancing CD4 T cells and antibody titers, while NPs preferentially augment cytotoxic CD8 T cells. Herein, we demonstrate that the differential activation of T cell immunity reflects differences in the modes of intracellular trafficking and distinct biodistribution of the Ag in lymphoid organs, which are both driven by the properties of each nanocarrier. Furthermore, we found that Ags within PSs promoted better CD4 T cell activation and induced a higher frequency of CD4 T follicular helper (Tfh) cells. These differences correlated with changes in the frequency of germinal center B cells and plasma cell formation, which reflects the previously observed antibody titers. Our results show that PSs are a promising vector for the delivery of Ags for B cell vaccine development. This study demonstrates that nanocarrier design has a large impact on the quality of the induced adaptive immune response.


Subject(s)
Antigens/administration & dosage , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Nanocapsules/chemistry , Vaccines/administration & dosage , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chick Embryo , Cytoplasm/metabolism , Dendritic Cells/metabolism , Drug Delivery Systems , Female , Germinal Center/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nanocomposites/chemistry , Ovalbumin/administration & dosage , Ovalbumin/immunology , Polymers/chemistry , Silver/chemistry , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Tissue Distribution , Vaccines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...