Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 22(11)2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34206051

RESUMO

Tumors arising in the context of Lynch Syndrome or constitutional mismatch repair deficiency are hypermutated and have a good response towards immune-checkpoint inhibitors (ICIs), including α-PD-L1 antibodies. However, in most cases, resistance mechanisms evolve. To improve outcomes and prevent resistance development, combination approaches are warranted. Herein, we applied a combined regimen with an α-PD-L1 antibody and gemcitabine in a preclinical tumor model to activate endogenous antitumor immune responses. Mlh1-/- mice with established gastrointestinal tumors received the α-PD-L1 antibody (clone 6E11; 2.5 mg/kg bw, i.v., q2wx3) and gemcitabine (100 mg/kg bw, i.p., q4wx3) in mono- or combination therapy. Survival and tumor growth were recorded. Immunological changes in the blood were routinely examined via multi-color flow cytometry and complemented by ex vivo frameshift mutation analysis to identify alterations in Mlh1-/--tumor-associated target genes. The combined therapy of α-PD-L1 and gemcitabine prolonged median overall survival of Mlh1-/- mice from four weeks in the untreated control group to 12 weeks, accompanied by therapy-induced tumor growth inhibition, as measured by [18F]-FDG PET/CT. Plasma cytokine levels of IL13, TNFα, and MIP1ß were increased and also higher than in mice receiving either monotherapy. Circulating splenic and intratumoral myeloid-derived suppressor cells (MDSCs), as well as M2 macrophages, were markedly reduced. Besides, residual tumor specimens from combi-treated mice had increased numbers of infiltrating cytotoxic T-cells. Frameshift mutations in APC, Tmem60, and Casc3 were no longer detectable upon treatment, likely because of the successful eradication of single mutated cell clones. By contrast, novel mutations appeared. Collectively, we herein confirm the safe application of combined chemo-immunotherapy by long-term tumor growth control to prevent the development of resistance mechanisms.


Assuntos
Antígeno B7-H1/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Colorretais Hereditárias sem Polipose/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Proteína 1 Homóloga a MutL/genética , Síndromes Neoplásicas Hereditárias/tratamento farmacológico , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Quimiocina CCL4/sangue , Neoplasias Colorretais/sangue , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais Hereditárias sem Polipose/sangue , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/imunologia , Reparo de Erro de Pareamento de DNA/genética , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Interleucina-13/sangue , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Camundongos , Células Supressoras Mieloides , Síndromes Neoplásicas Hereditárias/sangue , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia , Fator de Necrose Tumoral alfa/sangue , Gencitabina
2.
Immunol Invest ; 50(4): 338-355, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32397769

RESUMO

Background: The role of T cell Ig and ITIM domain (TIGIT) and programmed cell death-1 (PD-1) in colorectal cancer (CRC) with mismatch repair deficiency is unknown.Methods: This was a study of 60 CRC patients with mismatch repair deficiency and 30 healthy controls between June 2015 and October 2015.Results: The expression of Foxp3, PD-1, and TIGIT was higher in cancer tissues compared with adjacent mucosa (all P < .05). Patients with advanced TNM stage had a significantly higher expression of TIGIT (P = .025) and PD-1 (P = .020) than patients with early-stage CRC. The disease-free survival (DFS) of patients with high TIGIT (HR = 3.96, 95%CI: 1.34-11.69, P = .013) or PD-1 (HR = 214.8, 95%CI: 49.88-925.2, P < .001) expression were better. The overall survival (OS) of the patients with CRC and high expression of PD-1 was worse than those with low expression (HR = 4.01, 95%CI:1.26-12.69, P = .019).Conclusion: TIGIT and PD-1 are upregulated in CRC with mismatch repair deficiency and associated with TNM stage and DFS.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Colorretais/imunologia , Síndromes Neoplásicas Hereditárias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Receptores Imunológicos/imunologia , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Estudos de Casos e Controles , Neoplasias Colorretais/sangue , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Citocinas/sangue , Fatores de Transcrição Forkhead/genética , Humanos , Estimativa de Kaplan-Meier , Síndromes Neoplásicas Hereditárias/sangue , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/mortalidade , Receptor de Morte Celular Programada 1/genética , Receptores Imunológicos/genética , Linfócitos T/imunologia , Regulação para Cima
4.
Br J Cancer ; 120(8): 815-818, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30862951

RESUMO

In colorectal cancer (CRC), T-cell checkpoint blockade is only effective in patients diagnosed with mismatch repair-deficient (MMR-d) cancers. However, defects in Human Leukocyte Antigen (HLA) class I expression were reported to occur in most MMR-d CRCs, which would preclude antigen presentation in these tumours, considered essential for the clinical activity of this immunotherapeutic modality. We revisited this paradox by characterising HLA class I expression in two independent cohorts of CRC. We determined that loss of HLA class I expression occurred in the majority (73-78%) of MMR-d cases. This phenotype was rare in CRC liver metastases, irrespective of MMR status, whereas weak, inducible expression of HLA class I molecules was frequent in liver lesions. We propose that HLA class I is an important determinant of metastatic homing in CRCs. This observation is paramount to understand CRC carcinogenesis and for the application of immunotherapies in the metastatic setting.


Assuntos
Neoplasias Colorretais/terapia , Genes MHC Classe I/genética , Imunoterapia , Linfócitos T/imunologia , Apresentação de Antígeno/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Carcinogênese/genética , Carcinogênese/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Genes cdc/efeitos dos fármacos , Genes cdc/imunologia , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/imunologia
5.
Rev Neurol ; 68(3): 132-133, 2019 Feb 01.
Artigo em Espanhol | MEDLINE | ID: mdl-30687923
6.
Mod Pathol ; 32(4): 576-584, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30401949

RESUMO

Mismatch repair-deficient endometrial cancers have a high somatic mutation burden, suggesting that patients with these tumors may benefit from immunotherapy. Elucidating the immune suppressive mechanisms of mismatch repair-deficient endometrial cancers is fundamental to developing future immune-based interventions. This study aimed to determine the immune cell populations associated with mismatch repair-deficient endometrial cancers, especially focusing on targetable regulatory pathways of the immune response. A total of 76 endometrial cancer hysterectomy specimens were evaluated for tumor-infiltrating immune cells by immunohistochemistry. Immune specific markers were used to evaluate each specimen for the number of CD8 + cytotoxic T lymphocytes, forkhead-box P3 (FoxP3) + regulatory T cells, CD68 + tumor-associated macrophages, as well as programmed death-1 (PD-1) + immune cells, and the percentage of programmed death ligand-1 (PD-L1) + immune cells. Mismatch repair-deficient tumors exhibited a significantly higher number of CD8 + cytotoxic T lymphocytes (p = 0.0006), FoxP3 + regulatory T cells (p = 0.0003), PD-1 + immune cells (p = 0.0069), and a higher percentage of PD-L1 + immune cells (p = 0.0007) occupying the tumor compared to mismatch repair-proficient endometrial cancers. There was no significant difference in CD68 + tumor-associated macrophages infiltration between the two groups. Endometrial cancers with tumor PD-L1 expression also showed significantly increased infiltration of CD8 + cytotoxic T lymphocytes (p = 0.0002), FoxP3 + regulatory T cells (p = 0.0003), PD-1 + immune cells (p < 0.0001), and PD-L1 + immune cells (p < 0.0001). Endometrial cancers showing mismatch repair-deficiency and PD-L1 expression in tumor cells exhibit a prominent T cell-inflamed phenotype. More importantly, the increased number of FoxP3 + regulatory T cells in mismatch repair-deficient endometrial cancers suggests that combination therapy by targeting both regulatory T cells and immune checkpoints may be warranted to improve clinical efficacy.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Colorretais/imunologia , Neoplasias do Endométrio/imunologia , Linfócitos do Interstício Tumoral/imunologia , Síndromes Neoplásicas Hereditárias/imunologia , Subpopulações de Linfócitos T/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/patologia , Neoplasias Colorretais/patologia , Neoplasias do Endométrio/patologia , Feminino , Fatores de Transcrição Forkhead/imunologia , Humanos , Inflamação/imunologia , Linfócitos do Interstício Tumoral/patologia , Pessoa de Meia-Idade , Síndromes Neoplásicas Hereditárias/patologia , Fenótipo , Subpopulações de Linfócitos T/patologia
7.
Gynecol Oncol ; 149(1): 146-154, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29496294

RESUMO

OBJECTIVE: The role of mismatch repair (MMR) deficiency in ovarian cancer (OC) pathogenesis and its association with other clinicopathologic features, such as microsatellite instability (MSI) and expression of checkpoint proteins, remain largely elusive. METHODS: We performed Immunohistochemistry (IHC) for MLH1, MSH2, MSH6 and PMS2 on full-section slides from 419 OCs to assess the MMR status. The clinical relevance of MMR deficiency was analyzed in combination with clinical data. The MSI status (by MSI assay) and expression of CD3, CD8, PD-1 and PD-L1 (by IHC) were compared in OCs with different MMR status. RESULTS: We found that 2.6% OCs were MMR-negative, 4.3% OCs were MMR-low, and 63.6% of MMR-negative OCs were of endometrioid subtype. A significantly higher proportion of MMR-negative OCs were diagnosed at stage I or II compared to MMR-proficient OCs (p=0.0041). MSI was observed in all tested MMR-negative OCs, 14.3% of tested MMR-low OCs and 3.2% of tested MMR-proficient OCs. In addition, MMR-negative OCs had better progression free survival compared to MMR-proficient and MMR-low OCs (p=0.0046). Furthermore, the majority of OCs were PD-1-positive in intratumoral lymphocytes regardless of MMR status; while MMR-negative OCs exhibited significantly increased CD3+ and CD8+ tumor-infiltrating lymphocytes, and PD-L1+ intratumoral immune cells compared to MMR-proficient OCs. CONCLUSION: Our data suggests that MMR deficient OC is a unique molecular subgroup, characterized by early stage of diagnosis, MSI phenotype, and increased tumor-infiltrating lymphocytes. These patients may be good candidates for anti-PD-1/PD-L1 therapy.


Assuntos
Antígeno B7-H1/biossíntese , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Instabilidade de Microssatélites , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/imunologia , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/imunologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Adulto , Idoso , Antígeno B7-H1/imunologia , Carcinoma Epitelial do Ovário , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia
8.
Drugs ; 78(2): 155-162, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29350327

RESUMO

Microsatellite instability-high/DNA mismatch repair deficient tumors are found across the cancer spectrum and often harbor markedly increased numbers of mutations when compared to microsatellite stable/DNA mismatch repair proficient tumors. As a result of this high mutational load, tumor-infiltrating lymphocyte density is increased and more immunogenic neoepitopes are expressed, leading to upregulation of immune checkpoints in these tumors. Checkpoint inhibitors such as pembrolizumab and nivolumab, both immunoglobulin G4 (IgG4) monoclonal antibodies that block interactions between the programmed cell death receptor-1 and its ligands, have significant activity in this tumor class. This review will focus on hypermutated tumors and immuno-oncology drug development for this biologically unique tumor type, with an emphasis on FDA-approved immunotherapies for these cancers, as well as a short discussion of the many therapeutic and scientific challenges ahead in order to optimize the uses of this new class of drug.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Neoplasias Encefálicas/terapia , Neoplasias Colorretais/terapia , Síndromes Neoplásicas Hereditárias/terapia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoterapia , Instabilidade de Microssatélites , Mutação , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/imunologia , Nivolumabe/administração & dosagem , Nivolumabe/efeitos adversos , Nivolumabe/farmacologia , Nivolumabe/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia
9.
Cancer Lett ; 403: 159-164, 2017 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-28645564

RESUMO

Monoallelic germline mutations in one of the DNA mismatch repair (MMR) genes cause Lynch syndrome, with a high lifetime risks of colorectal and endometrial cancer at adult age. Less well known, is the constitutional mismatch repair deficiency (CMMRD) syndrome caused by biallelic germline mutations in MMR genes. This syndrome is characterized by the development of childhood cancer. Patients with CMMRD are at extremely high risk of developing multiple cancers including hematological, brain and intestinal tumors. Mutations in MMR genes impair DNA repair and therefore most tumors of patients with CMMRD are hypermutated. These mutations lead to changes in the translational reading frame, which consequently result in neoantigen formation. Neoantigens are recognized as foreign by the immune system and can induce specific immune responses. The growing evidence on the clinical efficacy of immunotherapies, such as immune checkpoint inhibitors, offers the prospect for treatment of patients with CMMRD. Combining neoantigen-based vaccination strategies and immune checkpoint inhibitors could be an effective way to conquer CMMRD-related tumors. Neoantigen-based vaccines might also be a preventive treatment option in healthy biallelic MMR mutation carriers. Future studies need to reveal the safety and efficacy of immunotherapies for patients with CMMRD.


Assuntos
Anticorpos/uso terapêutico , Antígenos de Neoplasias , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Neoplasias Colorretais/terapia , Reparo de Erro de Pareamento de DNA , Enzimas Reparadoras do DNA , Imunoterapia/métodos , Mutação , Síndromes Neoplásicas Hereditárias/terapia , Animais , Anticorpos/efeitos adversos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Vacinas Anticâncer/efeitos adversos , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/mortalidade , Reparo de Erro de Pareamento de DNA/genética , Reparo de Erro de Pareamento de DNA/imunologia , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/imunologia , Predisposição Genética para Doença , Humanos , Imunoterapia/efeitos adversos , Imunoterapia/mortalidade , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/imunologia , Síndromes Neoplásicas Hereditárias/mortalidade , Fenótipo
10.
Science ; 357(6349): 409-413, 2017 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-28596308

RESUMO

The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Biomarcadores Tumorais/antagonistas & inibidores , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Síndromes Neoplásicas Hereditárias/imunologia , Síndromes Neoplásicas Hereditárias/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Reparo de Erro de Pareamento de DNA , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/mortalidade , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Adulto Jovem
12.
Blood ; 113(25): 6288-95, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19351959

RESUMO

Wiskott-Aldrich syndrome (WAS) is a severe X-linked immunodeficiency caused by mutations in the gene encoding for WASP, a key regulator of signaling and cytoskeletal reorganization in hematopoietic cells. Mutations in WASP result in a wide spectrum of clinical manifestations ranging from the relatively mild X-linked thrombocytopenia to the classic full-blown WAS phenotype characterized by thrombocytopenia, immunodeficiency, eczema, and high susceptibility to developing tumors and autoimmune manifestations. The life expectancy of patients affected by severe WAS is reduced, unless they are successfully cured by bone marrow transplantation from related identical or matched unrelated donors. Because many patients lack a compatible bone marrow donor, the administration of WAS gene-corrected autologous hematopoietic stem cells could represent an alternative therapeutic approach. In the present review, we focus on recent progress in understanding the molecular and cellular mechanisms contributing to the pathophysiology of WAS. Although molecular and cellular studies have extensively analyzed the mechanisms leading to defects in T, B, and dendritic cells, the basis of autoimmunity and thrombocytopenia still remains poorly understood. A full understanding of these mechanisms is still needed to further implement new therapeutic strategies for this peculiar immunodeficiency.


Assuntos
Síndrome de Wiskott-Aldrich/fisiopatologia , Adolescente , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Criança , Células Dendríticas/imunologia , Eczema/genética , Eczema/imunologia , Predisposição Genética para Doença , Neoplasias Hematológicas/etiologia , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Humanos , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/patologia , Masculino , Camundongos , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/imunologia , Síndromes Neoplásicas Hereditárias/fisiopatologia , Síndromes Neoplásicas Hereditárias/terapia , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/imunologia , Síndrome de Wiskott-Aldrich/terapia , Proteína da Síndrome de Wiskott-Aldrich/deficiência , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Adulto Jovem
13.
Blood ; 113(25): 6382-5, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19234138

RESUMO

CD1d-restricted T cells are considered to play a host protective effect in tumor immunity, yet the evidence for a role of natural killer T (NKT) cells in tumor immune surveillance has been weak and data from several tumor models has suggested that some (type II) CD1d-restricted T cells may also suppress some types of antitumor immune response. To substantiate an important role for CD1d-restricted T cells in host response to cancer, we have evaluated tumor development in p53(+/-) mice lacking either type I NKT cells (TCR Jalpha18(-/-)) or all CD1d-restricted T cells (CD1d(-/-)). Our findings support a key role for type I NKT cells in suppressing the onset of sarcomas and hematopoietic cancers caused by p53 loss but do not suggest that other CD1d-restricted T cells are critical in regulating the same tumor development.


Assuntos
Deleção de Genes , Genes p53 , Vigilância Imunológica , Células T Matadoras Naturais/imunologia , Neoplasias Experimentais/imunologia , Síndromes Neoplásicas Hereditárias/imunologia , Subpopulações de Linfócitos T/imunologia , Envelhecimento/imunologia , Animais , Antígenos CD1d/genética , Cruzamentos Genéticos , Feminino , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/classificação , Neoplasias Experimentais/genética , Síndromes Neoplásicas Hereditárias/genética , Organismos Livres de Patógenos Específicos , Subpopulações de Linfócitos T/classificação , Carga Tumoral
14.
J Pathol ; 206(2): 220-3, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15880615

RESUMO

The aim of this study was to investigate closely the nature of premalignant lesions that occur in prophylactically removed breast tissue from patients at hereditary high risk of breast cancer. Breast tissues obtained from 41 patients who underwent prophylactic mastectomy (pM) because of a hereditary high risk of breast cancer and control tissues from 82 age-matched healthy controls who underwent breast reduction surgery were screened for premalignant lesions. Premalignant and malignant lesions were more frequent (p = 0.0016) in pM samples (5/41) than in controls (1/82). Interestingly, lobulitis, defined as more than 100 lymphocytes and/or plasma cells per lobule in more than one section in morphologically normal lobules, was encountered in 21 of 41 (51%) pM patients, in contrast to only 8 of 82 (10%) controls (p < 0.0001). Preliminary observations indicate a predominance of T-cells in these infiltrates, in agreement with the already known frequent presence of lymphocytic infiltration in hereditary ductal in situ and infiltrating ductal/medullary carcinomas. This novel finding implies an immune reaction to an as yet unidentified antigen frequently present in women at hereditary high risk of breast cancer, possibly as part of an early carcinogenic event.


Assuntos
Neoplasias da Mama/prevenção & controle , Síndromes Neoplásicas Hereditárias/prevenção & controle , Lesões Pré-Cancerosas/patologia , Adulto , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Feminino , Genes BRCA2 , Predisposição Genética para Doença , Humanos , Linfócitos do Interstício Tumoral/patologia , Mastectomia , Pessoa de Meia-Idade , Mutação , Síndromes Neoplásicas Hereditárias/imunologia , Síndromes Neoplásicas Hereditárias/patologia , Plasmócitos/patologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/imunologia , Linfócitos T/patologia
15.
Cell Mol Biol (Noisy-le-grand) ; 45(7): 1119-29, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10644016

RESUMO

Applying selective breeding for 10 years we established the MeLiM (Melanoblastoma-bearing Libechov Minipigs) strain. Melanoblastoma (MB) in this strain shows a hereditary occurrence. Cutaneous tumours are usually nodular, multiple and distributed on various parts of body. They appear in darkly pigmented animals already at the birth or during two months thereafter (57% of all animals). Numerous organ metastases mainly into the spleen, lymph nodes and lungs are regularly ascertained in animals with cutaneous MB. Tumour cells were surprisingly found also in the inner organs of phenotypically healthy minipigs in which no cutaneous MBs were observed visually (27% of all animals). About 34% of all affected piglets die during the first 2 months of age. These features document a malignancy of this tumour in the MeLiM strain. Original surgical technique was applied in more than 40 affected minipigs at 1-2 months of age. It consists in a devitalization (ischemization) of one of cutaneous tumours by the mattress sutures conducted around the tumour base without any excision of tumour tissue. This simple procedure causes a total destruction of MB cells in all cutaneous tumours as well as in all organ metastases during 4-6 months. Animals treated by this technique were fully healed of tumour cells and no relapses were observed. This technique could bring similar positive results also in therapy of human MB.


Assuntos
Melanoma Experimental/genética , Síndromes Neoplásicas Hereditárias/genética , Nevo Pigmentado/genética , Neoplasias Cutâneas/genética , Porco Miniatura/genética , Animais , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/imunologia , Cruzamento , Feminino , Genes Dominantes , Genes Recessivos , Cor de Cabelo/genética , Ligadura , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/congênito , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Complexo Principal de Histocompatibilidade/genética , Masculino , Melanócitos/imunologia , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/congênito , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/cirurgia , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Síndromes Neoplásicas Hereditárias/imunologia , Síndromes Neoplásicas Hereditárias/patologia , Nevo Pigmentado/congênito , Nevo Pigmentado/patologia , Fenótipo , Remissão Espontânea , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/congênito , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/cirurgia , Pigmentação da Pele/genética , Neoplasias Esplênicas/irrigação sanguínea , Neoplasias Esplênicas/congênito , Neoplasias Esplênicas/genética , Neoplasias Esplênicas/patologia , Neoplasias Esplênicas/cirurgia , Técnicas de Sutura , Suínos , Vísceras/patologia
16.
Thymus ; 16(2): 67-87, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2256125

RESUMO

The developmental status of T cell lineages at prethymic, intrathymic and postthymic stages in spontaneous thymoma Buffalo/Mna (BUF/Mna) rats was characterized on the basis of surface phenotypes and some immune responses. The proportion of bone marrow cell populations was shown to be normal in thymoma rats by immunofluorescence flow cytometry (FACS). In spite of a cortex-predominant appearance of thymomas, the proportion of thymocyte populations was phenotypically normal as assessed by FACS. Double immunohistochemical stainings revealed that a substantial number of single-possible (CD4+CD8- or CD4-CD8+) thymocytes existed not only in the narrowed medullary areas but also in the enlarged cortical areas of the thymomas. In lymph nodes, the proportion of T cells increased with age, reaching 88% after 20 months of age. This increase was due mainly to an increase in the CD8+ population but not the CD4+ population, resulting in low CD4/CD8 ratios. An abnormal increase of Thy1+ immature T cells was also observed in the lymph nodes. However, these phenotypic changes in the T cell lineages in the thymoma rats were not so influential as to alter their immunological reactivities, such as the primary antibody response to a T-dependent antigen, the graft-versus-host reaction and the mixed lymphocyte reaction to allo-antigens. These results suggest the possible presence of some altered differentiation pathways for intrathymic and postthymic T cell development in BUF/Mna rats.


Assuntos
Linfócitos T/patologia , Timoma/patologia , Neoplasias do Timo/patologia , Animais , Antígenos de Diferenciação de Linfócitos T/análise , Antígenos de Neoplasias/análise , Diferenciação Celular , Separação Celular , Citometria de Fluxo , Imunidade Celular , Síndromes Neoplásicas Hereditárias/imunologia , Síndromes Neoplásicas Hereditárias/patologia , Ratos , Ratos Endogâmicos BUF/imunologia , Subpopulações de Linfócitos T , Linfócitos T/imunologia , Timoma/imunologia , Neoplasias do Timo/imunologia
17.
Laryngorhinootologie ; 69(1): 21-3, 1990 Jan.
Artigo em Alemão | MEDLINE | ID: mdl-2310457

RESUMO

Although the number of carcinomas of the upper aerodigestive tract is increasing, there are not many reports in the literature about familial occurrence. Epidemiological studies have shown that the major causative factors are tobacco and alcohol abuse. But the possibility of an endogenous susceptibility is also discussed. A possible explanation is failure of the immune defense system. The author was able to examine immunologic patterns in three siblings with cancer of the upper aerodigestive tract. The results obtained so far indicate normal numbers of lymphocyte subpopulations in the peripheral blood, but with impaired function.


Assuntos
Epiglote , Imunidade Celular/genética , Neoplasias Laríngeas/genética , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Faríngeas/genética , Epiglote/imunologia , Feminino , Humanos , Tolerância Imunológica/genética , Células Matadoras Naturais/imunologia , Neoplasias Laríngeas/imunologia , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Síndromes Neoplásicas Hereditárias/imunologia , Neoplasias Faríngeas/imunologia
18.
J Natl Cancer Inst ; 78(3): 455-8, 1987 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-3469459

RESUMO

Cancer incidence and mortality were analyzed in 1,181 blood relatives and 558 spouse controls in 24 families of severe combined immune deficiency (SCID) patients, to test the hypothesis that heterozygous carriers of a gene for an autosomal recessive form of SCID are predisposed to cancer. Since at least 1 patient in each family was female and there were no cases outside the probands' sibships, the pattern of occurrence of SCID within the families was compatible with autosomal recessive inheritance. The observed numbers of cancer cases and deaths did not exceed the expected numbers derived from population-based rates; and there was no cancer excess when incidence rates in blood relatives were compared directly to those in spouse controls, since the rate ratios were 1.2 and 1.0 for males and females, respectively. In addition, cancer rate ratios were not significantly elevated when calculated separately for the 9 families of adenosine deaminase (ADA)-deficient SCID patients and for the 15 families without evidence of ADA deficiency.


Assuntos
Síndromes de Imunodeficiência/genética , Neoplasias/genética , Síndromes Neoplásicas Hereditárias , Adenosina Desaminase/deficiência , Adulto , Pré-Escolar , Suscetibilidade a Doenças , Feminino , Genes Recessivos , Heterozigoto , Humanos , Síndromes de Imunodeficiência/complicações , Lactente , Masculino , Neoplasias/etiologia , Neoplasias/imunologia , Síndromes Neoplásicas Hereditárias/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...