Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Front Immunol ; 14: 1057292, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37251410

RESUMO

Introduction: Characterization of the tumour immune infiltrate (notably CD8+ T-cells) has strong predictive survival value for cancer patients. Quantification of CD8 T-cells alone cannot determine antigenic experience, as not all infiltrating T-cells recognize tumour antigens. Activated tumour-specific tissue resident memory CD8 T-cells (TRM) can be defined by the co-express of CD103, CD39 and CD8. We investigated the hypothesis that the abundance and localization of TRM provides a higher-resolution route to patient stratification. Methods: A comprehensive series of 1000 colorectal cancer (CRC) were arrayed on a tissue microarray, with representative cores from three tumour locations and the adjacent normal mucosa. Using multiplex immunohistochemistry we quantified and determined the localization of TRM. Results: Across all patients, activated TRM were an independent predictor of survival, and superior to CD8 alone. Patients with the best survival had immune-hot tumours heavily infiltrated throughout with activated TRM. Interestingly, differences between right- and left-sided tumours were apparent. In left-sided CRC, only the presence of activated TRM (and not CD8 alone) was prognostically significant. Patients with low numbers of activated TRM cells had a poor prognosis even with high CD8 T-cell infiltration. In contrast, in right-sided CRC, high CD8 T-cell infiltration with low numbers of activated TRM was a good prognosis. Conclusion: The presence of high intra-tumoural CD8 T-cells alone is not a predictor of survival in left-sided CRC and potentially risks under treatment of patients. Measuring both high tumour-associated TRM and total CD8 T-cells in left-sided disease has the potential to minimize current under-treatment of patients. The challenge will be to design immunotherapies, for left-sided CRC patients with high CD8 T-cells and low activate TRM,that result in effective immune responses and thereby improve patient survival.


Assuntos
Neoplasias Colorretais , Células T de Memória , Humanos , Memória Imunológica , Linfócitos T CD8-Positivos
2.
J Pathol ; 258(2): 199-209, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35851954

RESUMO

High-level expression of decay-accelerating factor, CD55, has previously been found in human gastric cancer (GC) and intestinal metaplasia (IM) tissues. Therapeutic effects of CD55 inhibition in cancer have been reported. However, the role of Helicobacter pylori infection and virulence factors in the induction of CD55 and its association with histological changes of the human gastric mucosa remain incompletely understood. We hypothesised that CD55 would be increased during infection with more virulent strains of H. pylori, and with more marked gastric mucosal pathology. RT-qPCR and immunohistochemical analyses of gastric biopsy samples from 42 H. pylori-infected and 42 uninfected patients revealed that CD55 mRNA and protein were significantly higher in the gastric antrum of H. pylori-infected patients, and this was associated with the presence of IM, but not atrophy, or inflammation. Increased gastric CD55 and IM were both linked with colonisation by vacA i1-type strains independently of cagA status, and in vitro studies using isogenic mutants of vacA confirmed the ability of VacA to induce CD55 and sCD55 in gastric epithelial cell lines. siRNA experiments to investigate the function of H. pylori-induced CD55 showed that CD55 knockdown in gastric epithelial cells partially reduced IL-8 secretion in response to H. pylori, but this was not due to modulation of bacterial adhesion or cytotoxicity. Finally, plasma samples taken from the same patients were analysed for the soluble form of CD55 (sCD55) by ELISA. sCD55 levels were not influenced by IM and did not correlate with gastric CD55 mRNA levels. These results suggest a new link between active vacA i1-type H. pylori, IM, and CD55, and identify CD55 as a molecule of potential interest in the management of IM as well as GC treatment. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Antígenos CD55/genética , Antígenos CD55/metabolismo , Citotoxinas/metabolismo , Mucosa Gástrica/patologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Humanos , Metaplasia/patologia , RNA Mensageiro/metabolismo , Neoplasias Gástricas/patologia
3.
Biochem J ; 477(17): 3219-3235, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32789497

RESUMO

Immunotherapy has been successful in treating many tumour types. The development of additional tumour-antigen binding monoclonal antibodies (mAbs) will help expand the range of immunotherapeutic targets. Lewis histo-blood group and related glycans are overexpressed on many carcinomas, including those of the colon, lung, breast, prostate and ovary, and can therefore be selectively targeted by mAbs. Here we examine the molecular and structural basis for recognition of extended Lea and Lex containing glycans by a chimeric mAb. Both the murine (FG88.2) IgG3 and a chimeric (ch88.2) IgG1 mAb variants showed reactivity to colorectal cancer cells leading to significantly reduced cell viability. We determined the X-ray structure of the unliganded ch88.2 fragment antigen-binding (Fab) containing two Fabs in the unit cell. A combination of molecular docking, glycan grafting and molecular dynamics simulations predicts two distinct subsites for recognition of Lea and Lex trisaccharides. While light chain residues were exclusively used for Lea binding, recognition of Lex involved both light and heavy chain residues. An extended groove is predicted to accommodate the Lea-Lex hexasaccharide with adjoining subsites for each trisaccharide. The molecular and structural details of the ch88.2 mAb presented here provide insight into its cross-reactivity for various Lea and Lex containing glycans. Furthermore, the predicted interactions with extended epitopes likely explains the selectivity of this antibody for targeting Lewis-positive tumours.


Assuntos
Anticorpos Monoclonais Murinos , Antineoplásicos Imunológicos , Fragmentos Fab das Imunoglobulinas , Antígenos do Grupo Sanguíneo de Lewis , Antígenos CD15 , Simulação de Acoplamento Molecular , Neoplasias , Oligossacarídeos , Animais , Anticorpos Monoclonais Murinos/química , Anticorpos Monoclonais Murinos/imunologia , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/imunologia , Linhagem Celular Tumoral , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Antígenos do Grupo Sanguíneo de Lewis/química , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Antígenos CD15/química , Antígenos CD15/imunologia , Camundongos , Neoplasias/química , Neoplasias/imunologia , Oligossacarídeos/química , Oligossacarídeos/imunologia
4.
Biomedicines ; 8(7)2020 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-32708498

RESUMO

Granulocyte macrophage colony stimulating factor (GM-CSF) is a pro-inflammatory cytokine produced by immune cells. Recent evidence suggests that GM-CSF plays an important role in multiple sclerosis (MS) pathogenesis. We investigated the expression and regulation of GM-CSF in different immune cells in MS. We also investigated the differentiation and frequency of GM-CSF-producing Th cells that do not co-express interferon (IFN)-γ or interleukin-17 (IL-17) (Th-GM cells) in MS. We found a significant increase in the percentage of GM-CSF-expressing Th cells, Th1 cells, Th-GM cells, cytotoxic T (Tc) cells, monocytes, natural killer (NK) cells, and B cells in PBMC from MS patients stimulated with T cell stimuli. Stimulated PBMC culture supernatants from MS patients contained significantly higher levels of IL-2, IL-12, IL-1ß, and GM-CSF and significantly lower levels of transforming growth factor (TGF-)ß. Blocking IL-2 reduced the frequency of Th-GM cells in PBMC from MS patients. The frequency of Th-GM cells differentiated in vitro from naïve CD4+ T cells was significantly higher in MS patients and was further increased in MS with IL-2 stimulation. These findings suggest that all main immune cell subsets produce more GM-CSF in MS after in vitro stimulation, which is associated with defective TGF-ß and increased IL-2 and IL-12 production. Th-GM cells are increased in MS. GM-CSF may be a potential therapeutic target in MS.

5.
Cancer Res ; 80(16): 3399-3412, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32532823

RESUMO

Murine IgG3 glycan-targeting mAb often induces direct cell killing in the absence of immune effector cells or complement via a proinflammatory mechanism resembling oncotic necrosis. This cancer cell killing is due to noncovalent association between Fc regions of neighboring antibodies, resulting in enhanced avidity. Human isotypes do not contain the residues underlying this cooperative binding mode; consequently, the direct cell killing of mouse IgG3 mAb is lost upon chimerization or humanization. Using the Lewisa/c/x -targeting 88mAb, we identified the murine IgG3 residues underlying the direct cell killing and increased avidity via a series of constant region shuffling and subdomain swapping approaches to create improved ("i") chimeric mAb with enhanced tumor killing in vitro and in vivo. Constant region shuffling identified a major CH3 and a minor CH2 contribution, which was further mapped to discontinuous regions among residues 286-306 and 339-378 that, when introduced in 88hIgG1, recapitulated the direct cell killing and avidity of 88mIgG3. Of greater interest was the creation of a sialyl-di-Lewisa-targeting i129G1 mAb via introduction of these selected residues into 129hIgG1, converting it into a direct cell killing mAb with enhanced avidity and significant in vivo tumor control. The human iG1 mAb, termed Avidimabs, retained effector functions, paving the way for the proinflammatory direct cell killing to promote antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity through relief of immunosuppression. Ultimately, Fc engineering of human glycan-targeting IgG1 mAb confers proinflammatory direct cell killing and enhanced avidity, an approach that could be used to improve the avidity of other mAb with therapeutic potential. SIGNIFICANCE: Fc engineering enhances avidity and direct cell killing of cancer-targeting anti-glycan antibodies to create superior clinical candidates for cancer immunotherapy.


Assuntos
Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Morte Celular/imunologia , Neoplasias Colorretais/terapia , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Polissacarídeos/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Proteínas do Sistema Complemento , Feminino , Engenharia Genética , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Distribuição Aleatória
6.
J Biol Chem ; 295(4): 1009-1020, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31831622

RESUMO

Cancer remains a leading cause of morbidity and mortality worldwide, requiring ongoing development of targeted therapeutics such as monoclonal antibodies. Carbohydrates on embryonic cells are often highly expressed in cancer and are therefore attractive targets for antibodies. Stage-specific embryonic antigen-4 (SSEA-4) is one such glycolipid target expressed in many cancers, including breast and ovarian carcinomas. Here, we defined the structural basis for recognition of SSEA-4 by a novel monospecific chimeric antibody (ch28/11). Five X-ray structures of ch28/11 Fab complexes with the SSEA-4 glycan headgroup, determined at 1.5-2.7 Å resolutions, displayed highly similar three-dimensional structures indicating a stable binding mode. The structures also revealed that by adopting a horseshoe-shaped conformation in a deep groove, the glycan headgroup likely sits flat against the membrane to allow the antibody to interact with SSEA-4 on cancer cells. Moreover, we found that the terminal sialic acid of SSEA-4 plays a dominant role in dictating the exquisite specificity of the ch28/11 antibody. This observation was further supported by molecular dynamics simulations of the ch28/11-glycan complex, which show that SSEA-4 is stabilized by its terminal sialic acid, unlike SSEA-3, which lacks this sialic acid modification. These high-resolution views of how a glycolipid interacts with an antibody may help to advance a new class of cancer-targeting immunotherapy.


Assuntos
Anticorpos Antineoplásicos/imunologia , Ácido N-Acetilneuramínico/metabolismo , Neoplasias/imunologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Anticorpos Antineoplásicos/química , Especificidade de Anticorpos/imunologia , Configuração de Carboidratos , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Ligantes , Simulação de Dinâmica Molecular , Polissacarídeos/química , Polissacarídeos/metabolismo , Antígenos Embrionários Estágio-Específicos/química
7.
Breast Cancer Res Treat ; 174(1): 93-102, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30474779

RESUMO

PURPOSE: Gap junctions are specialized membrane structures that form channels between adjacent cells allowing cell communication. Gap junctions and specifically Connexin 43 (Cx43) are down-regulated in cancer; however, there are contrasting reports on how this effects breast cancer patient survival. This paper is the first large-scale tissue microarray analysis of Cx43 expression in breast cancer patients with an associated clinical long-term follow-up. METHODS: Using a validated TMA of 1118 primary breast cancers, coupled to a comprehensive database of clinicopathological variables, the expression levels and subcellular localisation of Cx43 was assessed by immunohistochemistry. Its impact in terms of survival, distant metastasis-free survival, and clinicopathological variables was determined. RESULTS: Patients whose tumors expressed high levels of Cx43 had significantly better survival (p < 0.001) than patients with low levels. High Cx43 expression within tumors was associated with an 18-month survival advantage. Loss of Cx43 expression was associated with markers of poor prognosis, namely large tumor size, high grade, high proliferation status, high pleomorphism, high mitosis, poor Nottingham Prognostic Index (NPI), and triple negative tumors. Cx43 expression was independent of tumor size, grade, stage and ER-status in predicting poor survival on multivariate analysis (p = 0.004). CONCLUSION: Connexin 43 (Cx43) is an independent predictor of breast cancer survival and distant metastasis-free survival. High expression of Cx43 was seen in only 13% of tumors, suggesting that drugs to increase Cx43 expression may result in prolonged patients survival.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/patologia , Conexina 43/biossíntese , Adulto , Idoso , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Conexina 43/análise , Feminino , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos
8.
EMBO Mol Med ; 10(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29930174

RESUMO

The adaptive cellular response to low oxygen tensions is mediated by the hypoxia-inducible factors (HIFs), a family of heterodimeric transcription factors composed of HIF-α and HIF-ß subunits. Prolonged HIF expression is a key contributor to cellular transformation, tumorigenesis and metastasis. As such, HIF degradation under hypoxic conditions is an essential homeostatic and tumour-suppressive mechanism. LIMD1 complexes with PHD2 and VHL in physiological oxygen levels (normoxia) to facilitate proteasomal degradation of the HIF-α subunit. Here, we identify LIMD1 as a HIF-1 target gene, which mediates a previously uncharacterised, negative regulatory feedback mechanism for hypoxic HIF-α degradation by modulating PHD2-LIMD1-VHL complex formation. Hypoxic induction of LIMD1 expression results in increased HIF-α protein degradation, inhibiting HIF-1 target gene expression, tumour growth and vascularisation. Furthermore, we report that copy number variation at the LIMD1 locus occurs in 47.1% of lung adenocarcinoma patients, correlates with enhanced expression of a HIF target gene signature and is a negative prognostic indicator. Taken together, our data open a new field of research into the aetiology, diagnosis and prognosis of LIMD1-negative lung cancers.


Assuntos
Adenocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Neoplasias Pulmonares/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/genética , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Masculino , Camundongos , Pessoa de Meia-Idade , Prognóstico , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/genética
9.
Oncotarget ; 8(60): 101215-101223, 2017 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-29254158

RESUMO

High-mobility group protein B1 (HMGB1) has been implicated in numerous tumour types where expression regulates tumour cell growth and survival. We hypothesised that high HMGB1 expression in ovarian tumours would predict poor patient survival. Using tissue microarrays of primary ovarian cancers combined with a comprehensive database of clinicopathological variables, the expression of HMGB1 was assessed by immunohistochemistry in two independent cohorts (n=194 and n=360) using a monoclonal antibody specific for HMGB1. Kaplan-Meier analysis showed an association of HMGB1 expression with progression free survival in the primary cohort (p=0.023). In the validation cohort, expression was associated with overall survival (p=0.002). Low expression of HMGB1 was protective and in a multivariate model HMGB1 expression was shown to be an independent predictor of poor survival in ovarian cancer (p=0.006). The role of HMGB1 in cancer is complex. As high levels of HMGB1 expression are likely to render ovarian cancer cells resistant to chemotherapy, therapies targeting the HMGB1 axis may be appropriate in the treatment of ovarian cancer patients.

10.
Br J Nutr ; 116(5): 798-804, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27523186

RESUMO

Gut microbes have a substantial influence on systemic immune function and allergic sensitisation. Manipulation of the gut microbiome through prebiotics may provide a potential strategy to influence the immunopathology of asthma. This study investigated the effects of prebiotic Bimuno-galactooligosaccharide (B-GOS) supplementation on hyperpnoea-induced bronchoconstriction (HIB), a surrogate for exercise-induced bronchoconstriction, and airway inflammation. A total of ten adults with asthma and HIB and eight controls without asthma were randomised to receive 5·5 g/d of either B-GOS or placebo for 3 weeks separated by a 2-week washout period. The peak fall in forced expiratory volume in 1 s (FEV1) following eucapnic voluntary hyperpnoea (EVH) defined HIB severity. Markers of airway inflammation were measured at baseline and after EVH. Pulmonary function remained unchanged in the control group. In the HIB group, the peak post-EVH fall in FEV1 at day 0 (-880 (sd 480) ml) was unchanged after placebo, but was attenuated by 40 % (-940 (sd 460) v. -570 (sd 310) ml, P=0·004) after B-GOS. In the HIB group, B-GOS reduced baseline chemokine CC ligand 17 (399 (sd 140) v. 323 (sd 144) pg/ml, P=0·005) and TNF-α (2·68 (sd 0·98) v. 2·18 (sd 0·59) pg/ml, P=0·040) and abolished the EVH-induced 29 % increase in TNF-α. Baseline C-reactive protein was reduced following B-GOS in HIB (2·46 (sd 1·14) v. 1·44 (sd 0·41) mg/l, P=0·015) and control (2·16 (sd 1·02) v. 1·47 (sd 0·33) mg/l, P=0·050) groups. Chemokine CC ligand 11 and fraction of exhaled nitric oxide remained unchanged. B-GOS supplementation attenuated airway hyper-responsiveness with concomitant reductions in markers of airway inflammation associated with HIB.


Assuntos
Broncoconstrição , Inflamação/prevenção & controle , Oligossacarídeos/farmacologia , Prebióticos/administração & dosagem , Doenças Respiratórias/prevenção & controle , Adulto , Biomarcadores , Proteína C-Reativa/genética , Proteína C-Reativa/metabolismo , Estudos de Casos e Controles , Quimiocinas/sangue , Quimiocinas/genética , Quimiocinas/metabolismo , Estudos Cross-Over , Método Duplo-Cego , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/genética , Imunoglobulina E/metabolismo , Inflamação/etiologia , Masculino , Oligossacarídeos/administração & dosagem , Doenças Respiratórias/etiologia , Fator de Necrose Tumoral alfa/sangue , Adulto Jovem
11.
J Leukoc Biol ; 100(2): 381-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27049023

RESUMO

Dendritic cell function is modulated by stromal cells, including fibroblasts. Although poorly understood, the signals delivered through this crosstalk substantially alter dendritic cell biology. This is well illustrated with release of TNF-α/IL-1ß from activated dendritic cells, promoting PGE2 secretion from stromal fibroblasts. This instructs dendritic cells to up-regulate IL-23, a key Th17-polarizing cytokine. We previously showed that ionizing radiation inhibited IL-23 production by human dendritic cells in vitro. In the present study, we investigated the hypothesis that dendritic cell-fibroblast crosstalk overcomes the suppressive effect of ionizing radiation to support appropriately polarized Th17 responses. Radiation (1-6 Gy) markedly suppressed IL-23 secretion by activated dendritic cells (P < 0.0001) without adversely impacting their viability and consequently, inhibited the generation of Th17 responses. Cytokine suppression by ionizing radiation was selective, as there was no effect on IL-1ß, -6, -10, and -27 or TNF-α and only a modest (11%) decrease in IL-12p70 secretion. Coculture with fibroblasts augmented IL-23 secretion by irradiated dendritic cells and increased Th17 responses. Importantly, in contrast to dendritic cells, irradiated fibroblasts maintained their capacity to respond to TNF-α/IL-1ß and produce PGE2, thus providing the key intermediary signals for successful dendritic cell-fibroblasts crosstalk. In summary, stromal fibroblasts support Th17-polarizing cytokine production by dendritic cells that would otherwise be suppressed in an irradiated microenvironment. This has potential ramifications for understanding the immune response to local radiotherapy. These findings underscore the need to account for the impact of microenvironmental factors, including stromal cells, in understanding the control of immunity.


Assuntos
Células Dendríticas/imunologia , Fibroblastos/imunologia , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Células Estromais/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Interleucina-1beta/metabolismo , Radiação Ionizante , Células Estromais/metabolismo , Células Estromais/efeitos da radiação , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos da radiação , Células Th17
12.
Mol Immunol ; 67(2 Pt A): 75-88, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25757815

RESUMO

Monoclonal antibodies represent the most successful class of biopharmaceuticals for the treatment of cancer. Mechanisms of action of therapeutic antibodies are very diverse and reflect their ability to engage in antibody-dependent effector mechanisms, internalize to deliver cytotoxic payloads, and display direct effects on cells by lysis or by modulating the biological pathways of their target antigens. Importantly, one of the universal changes in cancer is glycosylation and carbohydrate-binding antibodies can be produced to selectively recognize tumor cells over normal tissues. A promising group of cell surface antibody targets consists of carbohydrates presented as glycolipids or glycoproteins. In this review, we outline the basic principles of antibody-based targeting of carbohydrate antigens in cancer. We also present a detailed structural view of antibody recognition and the conformational properties of a series of related tissue-blood group (Lewis) carbohydrates that are being pursued as potential targets of cancer immunotherapy.


Assuntos
Anticorpos/uso terapêutico , Carboidratos/imunologia , Epitopos/imunologia , Imunoterapia/métodos , Neoplasias/terapia , Anticorpos/imunologia , Anticorpos/metabolismo , Configuração de Carboidratos , Sequência de Carboidratos , Carboidratos/química , Epitopos/química , Epitopos/metabolismo , Humanos , Antígenos do Grupo Sanguíneo de Lewis/química , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Dados de Sequência Molecular , Neoplasias/imunologia
13.
Clin Cancer Res ; 21(13): 2963-74, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25779947

RESUMO

PURPOSE: To produce antitumor monoclonal antibodies (mAbs) targeting glycans as they are aberrantly expressed in tumors and are coaccessory molecules for key survival pathways. EXPERIMENTAL DESIGN: Two mAbs (FG88.2 and FG88.7) recognizing novel tumor-associated Lewis (Le) glycans were produced by immunizations with plasma membrane lipid extracts of the COLO205 cell line. RESULTS: Glycan array analysis showed that both mAbs bound Le(c)Le(x), di-Le(a), and Le(a)Le(x), as well as Le(a)-containing glycans. These glycans are expressed on both lipids and proteins. Both mAbs showed strong tumor reactivity, binding to 71% (147 of 208) of colorectal, 81% (155 of 192) of pancreatic, 54% (52 of 96) of gastric, 23% (62 of 274) of non-small cell lung, and 31% (66 of 217) of ovarian tumor tissue in combination with a restricted normal tissue distribution. In colorectal cancer, high FG88 glyco-epitope expression was significantly associated with poor survival. The mAbs demonstrated excellent antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), in addition to direct tumor cell killing via a caspase-independent mechanism. Scanning electron microscopy revealed antibody-induced pore formation. In addition, the mAbs internalized, colocalized with lysosomes, and delivered saporin that killed cells with subnanomolar potency. In vivo, the mAbs demonstrated potent antitumor efficacy in a metastatic colorectal tumor model, leading to significant long-term survival. CONCLUSIONS: The mAbs direct and immune-assisted tumor cell killing, pan-tumor reactivity, and potent in vivo antitumor efficacy indicate their potential as therapeutic agents for the treatment of multiple solid tumors. In addition, internalization of saporin conjugates and associated tumor cell killing suggests their potential as antibody drug carriers.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos/farmacologia , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Animais , Anticorpos Monoclonais Murinos/metabolismo , Antineoplásicos/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Humanos , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Polissacarídeos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Immunol ; 191(12): 5895-903, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24198281

RESUMO

Unlike other helper T cells, the costimulatory ligands responsible for T regulatory type 1 (Tr1) cell differentiation remain undefined. Understanding the molecular interactions driving peripheral Tr1 differentiation is important because Tr1s potently regulate immune responses by IL-10 production. In this study, we show that costimulation of human naive CD4(+) cells through CD97/CD55 interaction drives Tr1 activation, expansion, and function. T cell activation and expansion was equipotent with CD55 or CD28 costimulation; however, CD55 costimulation resulted in two IL-10-secreting populations. Most IL-10 was secreted by the minor Tr1 population (IL-10(high)IFN-γ(-)IL-4(-), <5% cells) that expresses Tr1 markers CD49b, LAG-3, and CD226. This Tr1 phenotype was not restimulated by CD28. However, on CD55 restimulation, Tr1s proliferated and maintained their differentiated IL-10(high) phenotype. The Tr1s significantly suppressed effector T cell function in an IL-10-dependent manner. The remaining (>95%) cells adopted a Th1-like IFN-γ(+) phenotype. However, in contrast to CD28-derived Th1s, CD55-derived Th1s demonstrated increased plasticity with the ability to coexpress IL-10 when restimulated through CD55 or CD28. These data identify CD55 as a novel costimulator of human Tr1s and support a role for alternative costimulatory pathways in determining the fate of the growing number of T helper populations. This study demonstrates that CD55 acts as a potent costimulator and activator of human naive CD4(+) cells, resulting in the differentiation of a discrete Tr1 population that inhibits T cell function in an IL-10-dependent manner and maintains the Tr1 phenotype upon restimulation.


Assuntos
Antígenos CD55/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/análise , Antígenos CD28/imunologia , Divisão Celular , Células Cultivadas , Humanos , Imunofenotipagem , Interferon gama/análise , Interleucina-10/metabolismo , Interleucina-2/biossíntese , Interleucina-2/genética , Ativação Linfocitária , Linfopoese , Receptores Acoplados a Proteínas G , Subpopulações de Linfócitos T/química , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/química , Linfócitos T Reguladores/classificação , Linfócitos T Reguladores/metabolismo , Células Th1/química , Células Th1/imunologia , Células Th1/metabolismo
15.
J Immunol ; 190(7): 3246-55, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23460736

RESUMO

Little is known of the regulation of IL-23 secretion in dendritic cells (DC) despite its importance for human Th17 responses. In this study, we show for first time, to our knowledge, that the ataxia telangiectasia mutated (ATM) pathway, involved in DNA damage sensing, acts as an IL-23 repressor. Inhibition of ATM with the highly selective antagonist KU55933 markedly increased IL-23 secretion in human monocyte-derived DC and freshly isolated myeloid DC. In contrast, inhibiting the closely related mammalian target of rapamycin had no effect on IL-23. Priming naive CD4(+) T cells with ATM-inhibited DC increased Th17 responses over and above those obtained with mature DC. Although ATM blockade increased the abundance of p19, p35, and p40 mRNA, IL-12p70 secretion was unaffected. To further examine a role for ATM in IL-23 regulation, we exposed DC to low doses of ionizing radiation. Exposure of DC to x-rays resulted in ATM phosphorylation and a corresponding depression of IL-23. Importantly, ATM inhibition with KU55933 prevented radiation-induced ATM phosphorylation and abrogated the capacity of x-rays to suppress IL-23. To explore how ATM repressed IL-23, we examined a role for endoplasmic reticulum stress responses by measuring generation of the spliced form of X-box protein-1, a key endoplasmic reticulum stress transcription factor. Inhibition of ATM increased the abundance of X-box protein-1 mRNA, and this was followed 3 h later by increased peak p19 transcription and IL-23 release. In summary, ATM activation or inhibition, respectively, inhibited or augmented IL-23 release. This novel role of the ATM pathway represents a new therapeutic target in autoimmunity and vaccine development.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Interleucina-23/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Células Dendríticas/imunologia , Estresse do Retículo Endoplasmático , Ativação Enzimática/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Interleucina-23/metabolismo , Ativação Linfocitária/imunologia , Fatores de Transcrição de Fator Regulador X , Transdução de Sinais/efeitos da radiação , Células Th17/imunologia , Células Th17/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
16.
PLoS One ; 8(2): e54892, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23408949

RESUMO

BACKGROUND: Several monoclonal antibodies (mAbs) recognising Lewis(y), such as BR96, have reached the clinic but have failed to show good anti-tumour responses with an acceptable level of toxicity. No Lewis(b) mAbs have been trialled in patients. In this study we compare the specificity of three mAbs; BR96 (Lewis(y)), 2-25 LE (Lewis(b)) and 692/29 that recognises a unique facet of both Lewis(y) and Lewis(b). We then assessed the in vivo therapeutic effect of 692/29 using xenograft models. METHODOLOGY/PRINCIPAL FINDINGS: Using a glycan array, each mAb was shown to display a different binding pattern with only 692/29 binding to both Lewis(y) and Lewis(b). 692/29 was able to kill tumour cells over-expressing Lewis(y/b) directly, as well as by antibody and complement mediated cytotoxicity (ADCC/CDC), but failed to kill cells expressing low levels of these haptens. In contrast, BR96, directly killed cells expressing either high or low levels of Lewis(y) perhaps explaining its toxicity in patients. 2-25 LE failed to cause any direct killing but did mediate ADCC/CDC. Both 692/29 and BR96 bound to >80% of a panel of over 400 colorectal tumours whereas 2-25 LE showed lower reactivity (52%). 692/29 demonstrated more restricted normal tissue reactivity than both BR96 and 2-25 LE. 692/29 anti-Lewis(y/b) mAb also showed good in vivo killing in xenograft models. CONCLUSIONS/SIGNIFICANCE: MAbs targeting both Lewis(y) and Lewis(b) may have a therapeutic advantage over mAbs targeting just one hapten. 692/29 has a more restricted normal tissue distribution and a higher antigen threshold for killing which should reduce its toxicity compared to a Lewis(y) specific mAb. 692/29 has an ability to directly kill tumours whereas the anti-Lewis(b) mAb does not. This suggests that Lewis(y) but not Lewis(b) are functional glycans. 692/29 showed good anti-tumour responses in vivo and is a strong therapeutic candidate.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Morte Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Humanos
17.
Expert Rev Vaccines ; 10(7): 1093-106, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21806402

RESUMO

Monoclonal antibodies (mAbs) have an established role in current cancer therapy with seven approved for the treatment of a wide variety of tumors. The approved mAbs directly target tumor cells; however, it is becoming increasingly clear that as well as their direct effects, these mAbs can present antigens to the immune system. This stimulates long-lasting T-cell immunity, which may correlate with long-term survival. A more direct approach is to use mAbs to target antigens directly to antigen-presenting cells. One approach, ImmunoBody, which has just entered the clinic, stimulates antitumor immunity using mAbs genetically engineered to express tumor-specific T-cell epitopes. T cells not only respond via their T-cell receptors recognizing T-cell epitopes presented on MHC but are also influenced by stimulation of a wide variety of costimulatory molecules. mAbs targeting these molecules can also influence antitumor immunity. The main protagonist in this class of mAbs is ipilimumab, which has recently been shown to improve survival at 2 years in 23% of advanced melanoma patients. Combinations of mAbs targeting tumor antigens to activated antigen-presenting cells and mAbs targeting costimulatory receptors may provide effective therapy for a broad range of tumors.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunidade Celular/fisiologia , Imunoterapia/métodos , Neoplasias/terapia , Anticorpos Monoclonais/imunologia , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/imunologia , Humanos , Imunidade Celular/imunologia , Neoplasias/imunologia , Linfócitos T/imunologia
18.
Expert Opin Biol Ther ; 10(5): 735-48, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20384522

RESUMO

IMPORTANCE OF THE FIELD: Considerable progress has been made in identifying the antigens recognised by the immune system. This has led to the success of monoclonal antibody therapy and the recent approval of prophylactic vaccines that give excellent protection against cervical cancer. Provenge will shortly be the first therapeutic vaccine to be approved. AREAS COVERED IN THIS REVIEW: Our aim is to discuss the recent success with prophylactic cancer vaccines for prevention of cancer and the progress with therapeutic vaccines design to eradicate established tumours. Therapeutic vaccines need to stimulate high-avidity T cell responses that can recognise and kill tumours. How this can be achieved in cancer patients is discussed. The immunosuppressive tumour environment also needs to be modified to allow extravasation and efficacy of the vaccine induced T cells. WHAT THE READER WILL GAIN: An insight into the limitations of present cancer vaccine approaches and how they can be manipulated to give more effective anti-tumour responses. TAKE HOME MESSAGE: A combination of more effective vaccines that stimulate high-avidity T cells, in combination with drugs or monoclonal antibodies that neutralize immunosuppressive factors within the tumour environment are needed to improve the efficacy of immunotherapy of cancer.


Assuntos
Vacinas Anticâncer/uso terapêutico , Tolerância Imunológica , Linfócitos do Interstício Tumoral/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Evasão Tumoral , Animais , Humanos , Neoplasias/imunologia , Neoplasias/prevenção & controle , Resultado do Tratamento
19.
Adv Exp Med Biol ; 706: 138-48, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21618833

RESUMO

CD97 was identified as an early activation marker on T cells, having low expression on naive T cells. This is a common feature of molecules that have a role in T-cell function. It was subsequently identified as a ligand for CD55, which has been previously identified as an innate regulator of complement. The interaction of this receptor-ligand pair has been shown to provide a potent costimulatory signal to human T cells, despite their modest affinity. Though both CD97 and CD55 are expressed on T cells as well as antigen presenting cells (APCs), their interaction is significant when CD97 on APCs interacts with CD55 on T cells. The converse interaction is poorly defined and may be less significant. A unique aspect of the interaction of CD97 with CD55 is the stimulation of naive T cells, leading to the induction of IL-10 producing cells that behave like Trl regulatory cells. This raises a number of questions regarding the dual functions of CD55; regulating complement and stimulating T cells via CD97 interaction and any potential overlap in the consequences of these dual roles.


Assuntos
Imunidade Adaptativa , Antígenos CD/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD/química , Biomarcadores/metabolismo , Antígenos CD55/imunologia , Proteínas do Sistema Complemento/imunologia , Humanos , Ativação Linfocitária , Subpopulações de Linfócitos/imunologia , Modelos Moleculares , Conformação Proteica , Receptores Acoplados a Proteínas G , Transdução de Sinais/fisiologia
20.
Int J Gynecol Pathol ; 29(1): 8-18, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19952944

RESUMO

Dysregulation of the cell cycle is an important prerequisite for cancer development. p27 has an established role in cell cycle control and hence may be disrupted during carcinogenesis. The influence of p27 expression, including its subcellular location, on tumor behavior in ovarian cancer has been controversial. The purpose of this study was to evaluate the expression of p27 in a large population of patients with ovarian cancer and correlate this to clinicopathologic variables including overall survival. Using a tissue microarray of 339 primary ovarian cancers, the expression of p27 was assessed immunohistochemically. Coupled to a comprehensive database of clinicopathologic variables, its effect on these factors and survival was studied. Cytoplasmic p27 showed a progressively negative impact on overall survival (P=0.004). Tumors displaying nuclear p27 also had poorer prognosis (P=0.014). Factors shown to predict prognosis independently of each other were age, stage, and the absence of macroscopic disease after surgery. Cytoplasmic p27 expression, but not nuclear, was independently predictive of prognosis on multivariate analysis (P=0.042). Both subcellular locations of p27 expression were more frequently observed in serous compared with mucinous subtypes. Cytoplasmic p27 independently predicts poorer prognosis in ovarian carcinoma. These results seem counterintuitive, when considering the antiproliferative role of p27, but may reflect a more complex function of p27 within cell cycle regulation. These data support a novel role for p27 within the cytoplasm, possibly through effects on apoptosis, cellular motility, and drug resistance.


Assuntos
Biomarcadores Tumorais/análise , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimioterapia Adjuvante , Terapia Combinada , Citoplasma/metabolismo , Feminino , Procedimentos Cirúrgicos em Ginecologia , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Prognóstico , Análise Serial de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA