Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Angew Chem Int Ed Engl ; 63(13): e202316791, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38308859

RESUMO

Heparin and heparan sulfate (HS) are naturally occurring mammalian glycosaminoglycans, and their synthetic and semi-synthetic mimetics have attracted significant interest as potential therapeutics. However, understanding the mechanism of action by which HS, heparin, and HS mimetics have a biological effect is difficult due to their highly charged nature, broad protein interactomes, and variable structures. To address this, a library of novel single-entity dendritic mimetics conjugated to BODIPY, Fluorine-19 (19 F), and biotin was synthesized for imaging and localization studies. The novel dendritic scaffold allowed for the conjugation of labeling moieties without reducing the number of sulfated capping groups, thereby better mimicking the multivalent nature of HS-protein interactions. The 19 F labeled mimetics were assessed in phantom studies and were detected at concentrations as low as 5 mM. Flow cytometric studies using a fluorescently labeled mimetic showed that the compound associated with immune cells from tumors more readily than splenic counterparts and was directed to endosomal-lysosomal compartments within immune cells and cancer cells. Furthermore, the fluorescently labeled mimetic entered the central nervous system and was detectable in brain-infiltrating immune cells 24 hours after treatment. Here, we report the enabling methodology for rapidly preparing various labeled HS mimetics and molecular probes with diverse potential therapeutic applications.


Assuntos
Biotina , Compostos de Boro , Heparitina Sulfato , Animais , Heparitina Sulfato/química , Glicosaminoglicanos/metabolismo , Heparina/metabolismo , Mamíferos/metabolismo
3.
Front Immunol ; 14: 1152035, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153625

RESUMO

Anti-tumour T cell responses play a crucial role in controlling the progression of colorectal cancer (CRC), making this disease a promising candidate for immunotherapy. However, responses to immune-targeted therapies are currently limited to subpopulations of patients and specific types of cancer. Clinical studies have therefore focussed on identifying biomarkers that predict immunotherapy responses and elucidating the immunological landscapes of different cancers. Meanwhile, our understanding of how preclinical tumour models resemble human disease has fallen behind, despite their crucial role in immune-targeted drug development. A deeper understanding of these models is therefore needed to improve the development of immunotherapies and the translation of findings made in these systems. MC38 colon adenocarcinoma is a widely used preclinical model, yet how it recapitulates human colorectal cancer remains poorly defined. This study investigated the tumour-T cell immune landscape of MC38 tumours using histology, immunohistochemistry, and flow cytometry. We demonstrate that early-stage tumours exhibit a nascent TME, lacking important immune-resistance mechanisms of clinical interest, while late-stage tumours exhibit a mature TME resembling human tumours, with desmoplasia, T cell exhaustion, and T cell exclusion. Consequently, these findings clarify appropriate timepoint selection in the MC38 model when investigating both immunotherapies and mechanisms that contribute to immunotherapy resistance. Overall, this study provides a valuable resource that will enable appropriate application of the MC38 model and expedite the development and clinical translation of new immunotherapies.


Assuntos
Adenocarcinoma , Neoplasias do Colo , Humanos , Imunoterapia , Citometria de Fluxo
4.
J Immunol ; 209(9): 1635-1651, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36104111

RESUMO

The initiation of CD8+ T cell responses against dead cell-associated Ags is tightly regulated, facilitating adaptive immunity against pathogens and tumors while preventing autoimmunity. It is now well established that dying cells actively regulate the generation of CD8+ T cell responses via the release or exposure of damage-associated molecular patterns. However, it is unclear whether nonproteasomal proteases (activated in stressed and dying cells) can influence the availability of Ags for cross-presentation. Using a mouse model of immunogenic necrosis, we investigated the role of tumor-derived proteases in the priming of CD8+ T cells. We demonstrate that proteases released from necrotic tumor cells can degrade whole-protein Ag, generating proteolytic intermediates that are efficiently cross-presented by dendritic cells and enhance CD8+ T cell cross-priming. We identify a dominant role for calpain proteases, which are activated during necrotic cell death induced by severe heat shock. Mechanistically, proteolytic intermediates generated by tumor-derived proteases associate with necrotic tumor cell debris, which acts as a vehicle for Ag transfer that facilitates highly efficient cross-presentation in dendritic cells. Our results suggest that proteolytic systems activated in Ag donor cells during cell death may influence the availability of antigenic substrates for cross-presentation, thereby regulating the antigenicity of cell death.


Assuntos
Apresentação Cruzada , Neoplasias , Apresentação de Antígeno , Linfócitos T CD8-Positivos , Calpaína/metabolismo , Células Dendríticas , Humanos , Necrose/metabolismo , Neoplasias/metabolismo
5.
Chem Asian J ; 17(12): e202200228, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35427432

RESUMO

Heparan sulfate (HS) is a highly sulfated natural carbohydrate that plays crucial roles in cancer, inflammation, and angiogenesis. Heparanase (HPSE) is the sole HS degrading endoglycosidase that cleaves HS at structure-dependent sites along the polysaccharide chain. Overexpression of HPSE by cancer cells correlates with increased tumor size and enhanced metastasis. Previously we have shown that a tetramer HS mimetic is a potent HPSE inhibitor displaying remarkable anticancer activity in vivo. Building on that work, we report the synthesis and testing of a novel library of single entity trimer glycolipid mimetics that effectively inhibit HPSE at low nanomolar concentrations. A lipophilic arm was introduced to assess whether an improvement of pharmacokinetics and plasma residence time would offset the reduction in charge and multivalency. Preclinical tests in a mouse syngeneic model showed effective tumor growth inhibition by the tetramer but not the trimer glycomimetic.


Assuntos
Neoplasias Colorretais , Glicolipídeos , Animais , Neoplasias Colorretais/tratamento farmacológico , Glicolipídeos/farmacologia , Heparitina Sulfato/farmacologia , Camundongos , Neovascularização Patológica
6.
Vaccines (Basel) ; 9(5)2021 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-34066318

RESUMO

Breast cancer (BC) is the most frequently diagnosed cancer in women, with many patients experiencing recurrence following treatment. Antigens delivered on virus-like particles (VLPs) induce a targeted immune response and here we investigated whether the co-delivery of multiple antigens could induce a superior anti-cancer response for BC immunotherapy. VLPs were designed to recombinantly express murine survivin and conjugated with an aberrantly glycosylated mucin-1 (MUC1) peptide using an intracellular cleavable bis-arylhydrazone linker. Western blotting, electron microscopy and UV absorption confirmed survivin-VLP expression and MUC1 conjugation. To assess the therapeutic efficacy of VLPs, orthotopic BC tumours were established by injecting C57mg.MUC1 cells into the mammary fat pad of mice, which were then vaccinated with surv.VLP-SS-MUC1 or VLP controls. While wild-type mice vaccinated with surv.VLP-SS-MUC1 showed enhanced survival compared to VLPs delivering either antigen alone, MUC1 transgenic mice vaccinated with surv.VLP-SS-MUC1 showed no enhanced survival compared to controls. Hence, while co-delivery of two tumour antigens on VLPs can induce a superior anti-tumour immune response compared to the delivery of single antigens, additional strategies must be employed to break tolerance when targeted tumour antigens are expressed as endogenous self-proteins. Using VLPs for the delivery of multiple antigens represents a promising approach to improving BC immunotherapy, and has the potential to be an integral part of combination therapy in the future.

7.
Mol Ther ; 25(1): 62-70, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28129129

RESUMO

Conjugation of a vaccine adjuvant to an antigen enhances anti-tumor immune responses. Direct chemical conjugation, however, may limit their processing by the antigen-presenting cell for immune stimulation. To test this hypothesis, antigen-adjuvant conjugates were designed to be cleaved by an intracellular trigger to release antigen and adjuvant from each other. The different reductive environment inside and outside antigen-presenting cells was used as a trigger for targeted intracellular release. Two redox-responsive disulphide linkers were used to conjugate the model antigen ovalbumin to CpG. In vitro stability assays with the reductant glutathione showed that one conjugate (SS) was cleaved by glutathione concentrations of the extra- and intracellular compartments. A second conjugate (HYN-SS) was only cleaved at the higher intracellular glutathione concentration. In vitro cell culture studies showed that high T cell responses were generated by the HYN-SS and the stable conjugate HYN. The SS conjugate induced a lower T cell response similar to a mixture of CpG and ovalbumin. An in vivo therapeutic tumor trial demonstrated a superior survival rate of 9/10 for mice vaccinated with HYN-SS conjugate compared to HYN (6/10), SS (2/10), and the mixture (2/10). This intracellular cleavable conjugation strategy represents a promising approach to improve cancer immunotherapy of soluble vaccines.


Assuntos
Adjuvantes Imunológicos , Antígenos/imunologia , Vacinas Anticâncer/imunologia , Neoplasias/imunologia , Oligodesoxirribonucleotídeos , Adjuvantes Imunológicos/química , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Modelos Animais de Doenças , Melanoma Experimental , Camundongos , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Oligodesoxirribonucleotídeos/química , Ovalbumina/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA