Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 354
Filtrar
2.
Histochem Cell Biol ; 156(2): 165-182, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34003355

RESUMEN

Diagnosis and prognosis of breast cancer is based on disease staging identified through histopathological and molecular biology techniques. Animal models are used to gain mechanistic insights into the development of breast cancer. C(3)1-TAg is a genetically engineered mouse model that develops mammary cancer. However, carcinogenesis caused by this transgene was characterized in the Friend Virus B (FVB) background. As most genetic studies are done in mice with C57BL/6 J background, we aimed to define the histological alterations in C3(1)-TAg C57BL/6 J animals. Our results showed that C3(1)-TAg animals with C57BL/6 J background develop solid-basaloid adenoid cystic carcinomas with increased fibrosis, decreased area of adipocytes, and a high proliferative index, which are triple-negative for progesterone, estrogen, and human epidermal growth factor receptor 2 (HER2) receptors. Our results also revealed that tumor development is slower in the C57BL/6 J background when compared with the FVB strain, providing a better model to study the different stages in breast cancer progression.


Asunto(s)
Antígenos Virales de Tumores/genética , Neoplasias de la Mama/genética , Carcinoma Adenoide Quístico/genética , Modelos Genéticos , Animales , Antígenos Virales de Tumores/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinoma Adenoide Quístico/inmunología , Carcinoma Adenoide Quístico/patología , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
3.
mBio ; 12(2)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33849980

RESUMEN

JC polyomavirus (JCV), a DNA virus that leads to persistent infection in humans, is the causative agent of progressive multifocal leukoencephalopathy, a lethal brain disease that affects immunocompromised individuals. Almost nothing is currently known about how JCV infection is controlled by the innate immune response and, further, whether JCV has evolved mechanisms to antagonize antiviral immunity. Here, we show that the innate immune sensors retinoic acid-inducible gene I (RIG-I) and cGMP-AMP synthase (cGAS) control JCV replication in human astrocytes. We further identify that the small t antigen (tAg) of JCV functions as an interferon (IFN) antagonist by suppressing RIG-I-mediated signal transduction. JCV tAg interacts with the E3 ubiquitin ligase TRIM25, thereby preventing its ability to bind RNA and to induce the K63-linked ubiquitination of RIG-I, which is known to facilitate RIG-I-mediated cytokine responses. Antagonism of RIG-I K63-linked ubiquitination and antiviral signaling is also conserved in the tAg of the related polyomavirus BK virus (BKV). These findings highlight how JCV and BKV manipulate a key innate surveillance pathway, which may stimulate research into designing novel therapies.IMPORTANCE The innate immune response is the first line of defense against viral pathogens, and in turn, many viruses have evolved strategies to evade detection by the host's innate immune surveillance machinery. Investigation of the interplay between viruses and the innate immune response provides valuable insight into potential therapeutic targets against viral infectious diseases. JC polyomavirus (JCV) is associated with a lifelong, persistent infection that can cause a rare neurodegenerative disease, called progressive multifocal leukoencephalopathy, in individuals that are immunosuppressed. The molecular mechanisms of JCV infection and persistence are not well understood, and very little is currently known about the relevance of innate immunity for the control of JCV replication. Here, we define the intracellular innate immune sensors responsible for controlling JCV infection and also demonstrate a novel mechanism by which a JCV-encoded protein acts as an antagonist of the type I interferon-mediated innate immune response.


Asunto(s)
Antígenos Virales de Tumores/inmunología , Proteína 58 DEAD Box/inmunología , Inmunidad Innata , Virus JC/inmunología , Proteínas de Unión al ARN/antagonistas & inhibidores , ARN/metabolismo , Receptores Inmunológicos/inmunología , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Antígenos Virales de Tumores/genética , Astrocitos/virología , Células Cultivadas , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Células HEK293 , Humanos , Virus JC/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética
4.
Cell Reprogram ; 23(2): 108-116, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33861637

RESUMEN

In our previous study, we constructed Schwann cells (SCs) that stably express Simian virus 40 T antigen (SV40T-SCs). SV40T-SCs functions and markers are similar to those of neural crest cells. There we used bone morphogenetic protein 9 (BMP9) to induce SV40T-SCs differentiation in vitro and in vivo and study possible related mechanism. SV40T-SCs differentiation was induced by BMP9 conditioned medium. The lipogenic differentiation of SV40T-SCs was assessed by Oil Red O staining. Alizarin red and Alcian blue staining, and alkaline phosphatase (ALP) assays were used to evaluate the SV40T-SCs osteogenic differentiation. The expression of adipocyte differentiation (c/EBPα and c/EBPß) and osteoblast differentiation markers (OSX and RUNX2) were detected by quantitative polymerase chain reaction (qPCR). To study possible mechanism related to SV40T-SCs differentiation, the P53 and E2F1 activity were assessed by luciferase reporter plasmid, and Slug and E-cadherin expression by qPCR. In vivo, SV40T-SCs infected by Ad-BMP9 or Ad-GFP were injected under the skin of nude mice. After 4-6 W, the mice were euthanized and subcutaneously mass formed at injecting sites was collected for pathological analysis. After SV40T-SCs were cultured in BMP9 conditioned medium, lipid droplets were formed in the cytoplasm of these cells. Alizarin red and Alcian blue staining were positive, and ALP activity of SV40T-SCs increased significantly. The expression of adipocyte differentiation (c/EBPα and c/EBPß) and osteoblast differentiation markers (OSX and RUNX2) in SV40T-SCs was upregulated by BMP9. SV40T significantly increased Slug expression and decreased E-cadherin expression. SV40T-SCs infected with Ad-BMP9 were able to differentiate into adipose tissue and form a small bone matrix under the nude mice skin. SV40T-SCs have the ability to differentiate into adipocytes and osteoblasts in vivo and in vitro. SV40T can upregulate the Slug expression and downregulate the E-cadherin expression to produce endothelial-to-mesenchymal transition (EMT). The multidirectional differentiation ability of SV40T-SCs may be related to EMT.


Asunto(s)
Adipocitos/citología , Antígenos Virales de Tumores/inmunología , Factor 2 de Diferenciación de Crecimiento/metabolismo , Osteoblastos/citología , Osteogénesis , Células de Schwann/citología , Virus 40 de los Simios/inmunología , Adipocitos/inmunología , Adipocitos/metabolismo , Animales , Antígenos Virales de Tumores/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Desnudos , Osteoblastos/inmunología , Osteoblastos/metabolismo , Células de Schwann/inmunología , Células de Schwann/metabolismo , Virus 40 de los Simios/metabolismo
5.
Pituitary ; 24(2): 159-169, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33044631

RESUMEN

BACKGROUND: Craniopharyngioma represents a troublesome tumor of the intracranial sellar region. There are currently no available well-characterized craniopharyngioma cell lines. This lack of reliable, immortal cell lines is a major reason for the slow progress in fundamental research related to craniopharyngioma. METHODS: We describe the development of an immortal papillary craniopharyngioma (PCP) cell line by transfecting primary PCP cells with the pLenti-simian virus 40 large T antigen(SV40LT). RESULTS: Three clones have been cultured for more than 14 months so far, while non-transfected cells ceased proliferation within three months of isolation. The established immortal PCP cell lines were identified to have BRAFV600E mutations, while no mutations in tumor suppressor genes were found in primary cells or immortal cells. Immortal cells had higher proliferation rates and formed tumors when implanted in the bran of nude mice. BRAF inhibition in immortal PCP cells altered cell morphology, inhibited cell proliferation and promoted apoptosis. CONCLUSION: We successfully developed PCP cell lines by SV40LT-mediated immortalization. These cell lines represent a powerful tool for fundamental and therapeutical studies on craniopharyngioma.


Asunto(s)
Antígenos Virales de Tumores/inmunología , Craneofaringioma/inmunología , Virus 40 de los Simios/inmunología , Animales , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteínas de Unión al GTP rho/genética
6.
Viruses ; 12(12)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371492

RESUMEN

Failing BK polyomavirus (BKPyV)-specific immune control is underlying onset and duration of BKPyV-replication and disease. We focused on BKPyV-specific CD8 T-cells as key effectors and characterized immunodominant 9mer epitopes in the viral large tumor-antigen (LTag). We investigated the variation of LTag-epitopes and their predicted effects on HLA-class 1 binding and T-cell activation. Available BKPyV sequences in the NCBI-nucleotide (N = 3263), and the NCBI protein database (N = 4189) were extracted (1368 sequences) and analyzed for non-synonymous aa-exchanges in LTag. Variant 9mer-epitopes were assessed for predicted changes in HLA-A and HLA-B-binding compared to immunodominant 9mer reference. We identified 159 non-synonymous aa-exchanges in immunodominant LTag-9mer T-cell epitopes reflecting different BKPyV-genotypes as well as genotype-independent variants altering HLA-A/HLA-B-binding scores. Decreased binding scores for HLA-A/HLA-B were found in 27/159 (17%). This included the immunodominant LPLMRKAYL affecting HLA-B*07:02-, HLA-B*08:01- and HLA-B*51:01-presentation. In two healthy BKPyV-seropositive HLA-B*07:02 blood donors, variant LSLMRKAYL showed reduced CD8 T-cell responses compared to LPLMRKAYL. Thus, despite LTag being highly conserved, aa-exchanges occur in immunodominant CD8 T-cell epitopes of BKPyV-genotypes as well as of genotypes -independent variants, which may contribute to genotype-dependent and genotype-independent failure of cellular immune control over BKPyV-replication. The data warrant epidemiological and immunological investigations in carefully designed clinical studies.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos Virales de Tumores/inmunología , Virus BK/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígenos HLA/inmunología , Epítopos Inmunodominantes/inmunología , Infecciones por Polyomavirus/inmunología , Variación Antigénica , Virus BK/genética , Biología Computacional/métodos , Mapeo Epitopo , Epítopos de Linfocito T/química , Humanos , Epítopos Inmunodominantes/química , Infecciones por Polyomavirus/virología
7.
Viruses ; 12(11)2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153187

RESUMEN

Progressive Multifocal Leukoencephalopathy (PML) is a fatal demyelinating disease of the CNS, resulting from the lytic infection of oligodendrocytes by the human neurotropic polyomavirus JC (JCPyV), typically associated with severe immunocompromised states and, in recent years, with the use of immunotherapies. Apoptosis is a homeostatic mechanism to dispose of senescent or damaged cells, including virally infected cells, triggered in the vast majority of viral infections of the brain. Previously, we showed upregulation of the normally dormant anti-apoptotic protein Survivin in cases of PML, which-in vitro-resulted in protection from apoptosis in JCPyV-infected primary cultures of astrocytes and oligodendrocytes. In the present study, we first demonstrate the absence of apoptotic DNA fragmentation and the lack of caspase activity in 16 cases of PML. We also identified the viral protein large T-Antigen as being responsible for the activation of the Survivin promoter. Chromatin Immunoprecipitation assay shows a direct binding between T-Antigen and the Survivin promoter DNA. Finally, we have identified the specific region of T-Antigen, spanning from amino acids 266 and 688, which binds to Survivin and translocates it to the nucleus, providing evidence of a mechanism that results in the efficient replication of JCPyV and a potential target for novel therapies.


Asunto(s)
Antígenos Virales de Tumores/genética , Apoptosis , Virus JC/genética , Regiones Promotoras Genéticas , Survivin/genética , Adulto , Anciano , Animales , Antígenos Virales de Tumores/inmunología , Astrocitos/virología , Caspasas/inmunología , Línea Celular Tumoral , Células Cultivadas , Niño , Fragmentación del ADN , Femenino , Humanos , Virus JC/inmunología , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva , Masculino , Ratones , Persona de Mediana Edad , Oligodendroglía/virología , Adhesión en Parafina , Survivin/inmunología
8.
PLoS One ; 15(11): e0242851, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33253272

RESUMEN

Mucin-type O-glycans are involved in cancer initiation and progression, although details of their biological and clinicopathological roles remain unclear. The aim of this study was to investigate the clinicopathological significance of ß1,3-N-acetylglucosaminyltransferase 6 (ß3Gn-T6), an essential enzyme for the synthesis of core 3 O-glycan and several other O-glycans in pancreatic ductal adenocarcinoma (PDAC). We performed immunohistochemical and lectin-histochemical analyses to detect the expression of ß3Gn-T6 and several O-glycans in 156 cases of PDAC with pancreatic intraepithelial neoplasias (PanINs) and corresponding normal tissue samples. The T antigen, Tn antigen, sialyl Lewis X (sLeX) antigen, and sLeX on core 2 O-glycan were more highly expressed in PDAC cells than in normal pancreatic duct epithelial cells (NPDEs). Conversely, the expression of 6-sulfo N-acetyllactosamine on extended core 1 O-glycan was found in NPDEs and was low in PDAC cells. These glycan expression levels were not associated with patient outcomes. ß3Gn-T6 was expressed in ~20% of PDAC cases and 30-40% of PanINs but not in NPDEs. Higher expression of ß3Gn-T6 was found in PDAC cells in more differentiated adenocarcinoma cases showing significantly longer disease-free survival in both univariate and multivariate analyses. In addition, the expression of ß3Gn-T6 in PDAC cells and PanINs significantly correlated with the expression of MUC5AC in these cells, suggesting that ß3Gn-T6 expression is related to cellular differentiation status of the gastric foveolar phenotype. Thus, it is likely that ß3Gn-T6 expression in PDAC cells is a favorable prognostic factor in PDAC patients, and that the expression of ß3Gn-T6 correlates with the gastric foveolar phenotype in pancreatic carcinogenesis.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , N-Acetilglucosaminiltransferasas/genética , Polisacáridos/genética , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Carbohidratos Asociados a Tumores/genética , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/inmunología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Polisacáridos/inmunología , Antígeno Sialil Lewis X/genética , Antígeno Sialil Lewis X/inmunología
9.
Cells ; 9(9)2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32942747

RESUMEN

Head and neck squamous cell carcinomas (HNSCC) are a heterogeneous group of malignant tumours typically caused by alcohol and tobacco consumption, although an increasing number of HNSCC arise due to persistent infection with high-risk human papilloma virus (HPV). The treatment of HNSCC remains challenging, and the first-line setting is focused on surgery and chemoradiotherapy. A substantial proportion of HNSCC patients die from their disease, especially those with recurrent and metastatic disease. Among factors linked with good outcome, immune cell infiltration appears to have a major role. HPV-driven HNSCC are often T-cell rich, reflecting the presence of HPV antigens that are immunogenic. Tumour-associated antigens that are shared between patients or that are unique to an individual person may also induce varying degrees of immune response; studying these is important for the understanding of the interaction between the host immune system and the cancer. The resulting knowledge is critical for the design of better immunotherapies. Key questions are: Which antigens lead to an adaptive immune response in the tumour? Which of these are exploitable for immunotherapy? Here, we review the current thinking regarding tumour antigens in HNSCC and what has been learned from early phase clinical trials.


Asunto(s)
Antígenos Virales de Tumores/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/terapia , Inmunoterapia/métodos , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/virología , Linfocitos T/inmunología
10.
Cancer Immunol Res ; 8(5): 648-659, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32179557

RESUMEN

Merkel cell carcinoma (MCC) is often caused by persistent expression of Merkel cell polyomavirus (MCPyV) T-antigen (T-Ag). These non-self proteins comprise about 400 amino acids (AA). Clinical responses to immune checkpoint inhibitors, seen in about half of patients, may relate to T-Ag-specific T cells. Strategies to increase CD8+ T-cell number, breadth, or function could augment checkpoint inhibition, but vaccines to augment immunity must avoid delivery of oncogenic T-antigen domains. We probed MCC tumor-infiltrating lymphocytes (TIL) with an artificial antigen-presenting cell (aAPC) system and confirmed T-Ag recognition with synthetic peptides, HLA-peptide tetramers, and dendritic cells (DC). TILs from 9 of 12 (75%) subjects contained CD8+ T cells recognizing 1-8 MCPyV epitopes per person. Analysis of 16 MCPyV CD8+ TIL epitopes and prior TIL data indicated that 97% of patients with MCPyV+ MCC had HLA alleles with the genetic potential that restrict CD8+ T-cell responses to MCPyV T-Ag. The LT AA 70-110 region was epitope rich, whereas the oncogenic domains of T-Ag were not commonly recognized. Specific recognition of T-Ag-expressing DCs was documented. Recovery of MCPyV oncoprotein-specific CD8+ TILs from most tumors indicated that antigen indifference was unlikely to be a major cause of checkpoint inhibition failure. The myriad of epitopes restricted by diverse HLA alleles indicates that vaccination can be a rational component of immunotherapy if tumor immune suppression can be overcome, and the oncogenic regions of T-Ag can be modified without impacting immunogenicity.


Asunto(s)
Antígenos Virales de Tumores/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células de Merkel/inmunología , Epítopos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Poliomavirus de Células de Merkel/inmunología , Neoplasias Cutáneas/inmunología , Adulto , Anciano , Antígenos Virales de Tumores/metabolismo , Carcinogénesis/inmunología , Carcinoma de Células de Merkel/terapia , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Persona de Mediana Edad , Neoplasias Cutáneas/terapia , Adulto Joven
11.
Mol Carcinog ; 59(7): 807-821, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32219902

RESUMEN

Great strides have been made in cancer immunotherapy including the breakthrough successes of anti-PD-(L)1 checkpoint inhibitors. In Merkel cell carcinoma (MCC), a rare and aggressive skin cancer, PD-(L)1 blockade is highly effective. Yet, ~50% of patients either do not respond to therapy or develop PD-(L)1 refractory disease and, thus, do not experience long-term benefit. For these patients, additional or combination therapies are needed to augment immune responses that target and eliminate cancer cells. Therapeutic vaccines targeting tumor-associated antigens, mutated self-antigens, or immunogenic viral oncoproteins are currently being developed to augment T-cell responses. Approximately 80% of MCC cases in the United States are driven by the ongoing expression of viral T-antigen (T-Ag) oncoproteins from genomically integrated Merkel cell polyomavirus (MCPyV). Since T-Ag elicits specific B- and T-cell immune responses in most persons with virus-positive MCC (VP-MCC), and ongoing T-Ag expression is required to drive VP-MCC cell proliferation, therapeutic vaccination with T-Ag is a rational potential component of immunotherapy. Failure of the endogenous T-cell response to clear VP-MCC (allowing clinically evident tumors to arise) implies that therapeutic vaccination will need to be potent ansd synergize with other mechanisms to enhance T-cell activity against tumor cells. Here, we review the relevant underlying biology of VP-MCC, potentially applicable therapeutic vaccine platforms, and antigen delivery formats. We also describe early successes in the field of therapeutic cancer vaccines and address several clinical scenarios in which VP-MCC patients could potentially benefit from a therapeutic vaccine.


Asunto(s)
Carcinoma de Células de Merkel/inmunología , Poliomavirus de Células de Merkel/inmunología , Neoplasias Cutáneas/inmunología , Vacunas/inmunología , Animales , Antígenos Virales de Tumores/inmunología , Carcinoma de Células de Merkel/terapia , Carcinoma de Células de Merkel/virología , Humanos , Inmunoterapia/métodos , Neoplasias Cutáneas/terapia , Neoplasias Cutáneas/virología , Linfocitos T/inmunología
12.
Sci Rep ; 9(1): 16641, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31719620

RESUMEN

Anti-mucin1 (MUC1) antibodies have long been used clinically in cancer diagnosis and therapy and specific bindings of some of them are known to be dependent on the differential glycosylation of MUC1. However, a systematic comparison of the binding specificities of anti-MUC1 antibodies was not previously conducted. Here, a total of 20 glycopeptides including the tandem repeat unit of MUC1, APPAHGVTSAPDTRPAPGSTAPPAHGV with GalNAc (Tn-antigen), Galß1-3GalNAc (T-antigen), NeuAcα2-3Galß1-3GalNAc (sialyl-T-antigen), or NeuAcα2-6GalNAc (sialyl-Tn-antigen) at each threonine or serine residue were prepared by a combination of chemical glycopeptide synthesis and enzymatic extension of carbohydrate chains. These glycopeptides were tested by the enzyme-linked immunosorbent assay (ELISA) for their capacity to bind 13 monoclonal antibodies (mAbs) known to be specific for MUC1. The results indicated that anti-MUC1 mAbs have diverse specificities but can be classified into a few characteristic groups based on their binding pattern toward glycopeptides in some cases having a specific glycan at unique glycosylation sites. Because the clinical significance of some of these antibodies was already established, the structural features identified by these antibodies as revealed in the present study should provide useful information relevant to their further clinical use and the biological understanding of MUC1.


Asunto(s)
Anticuerpos/inmunología , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antígenos Virales de Tumores/inmunología , Mucina-1/inmunología , Mucinas/inmunología , Secuencias Repetidas en Tándem , Anticuerpos/genética , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/genética , Especificidad de Anticuerpos/inmunología , Antígenos de Carbohidratos Asociados a Tumores/genética , Antígenos Virales de Tumores/genética , Ensayo de Inmunoadsorción Enzimática , Glicopéptidos/síntesis química , Glicopéptidos/inmunología , Humanos , Mucina-1/genética , Mucinas/síntesis química , Mucinas/genética , Secuencias Repetidas en Tándem/genética
13.
Proc Natl Acad Sci U S A ; 116(40): 20104-20114, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31527246

RESUMEN

Viral cancers show oncogene addiction to viral oncoproteins, which are required for survival and proliferation of the dedifferentiated cancer cell. Human Merkel cell carcinomas (MCCs) that harbor a clonally integrated Merkel cell polyomavirus (MCV) genome have low mutation burden and require viral T antigen expression for tumor growth. Here, we showed that MCV+ MCC cells cocultured with keratinocytes undergo neuron-like differentiation with neurite outgrowth, secretory vesicle accumulation, and the generation of sodium-dependent action potentials, hallmarks of a neuronal cell lineage. Cocultured keratinocytes are essential for induction of the neuronal phenotype. Keratinocyte-conditioned medium was insufficient to induce this phenotype. Single-cell RNA sequencing revealed that T antigen knockdown inhibited cell cycle gene expression and reduced expression of key Merkel cell lineage/MCC marker genes, including HES6, SOX2, ATOH1, and KRT20 Of these, T antigen knockdown directly inhibited Sox2 and Atoh1 expression. MCV large T up-regulated Sox2 through its retinoblastoma protein-inhibition domain, which in turn activated Atoh1 expression. The knockdown of Sox2 in MCV+ MCCs mimicked T antigen knockdown by inducing MCC cell growth arrest and neuron-like differentiation. These results show Sox2-dependent conversion of an undifferentiated, aggressive cancer cell to a differentiated neuron-like phenotype and suggest that the ontology of MCC arises from a neuronal cell precursor.


Asunto(s)
Antígenos Virales de Tumores/genética , Carcinoma de Células de Merkel/etiología , Carcinoma de Células de Merkel/metabolismo , Poliomavirus de Células de Merkel/genética , Fenotipo , Infecciones por Polyomavirus/complicaciones , Factores de Transcripción SOXB1/genética , Antígenos Virales de Tumores/inmunología , Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/patología , Ciclo Celular/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Transformación Celular Viral , Técnicas de Silenciamiento del Gen , Humanos , Queratinocitos , Células de Merkel/metabolismo , Poliomavirus de Células de Merkel/inmunología , Neuritas/metabolismo , Neuronas/metabolismo , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/virología , Factores de Transcripción SOXB1/metabolismo , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
14.
Int J Mol Sci ; 20(16)2019 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-31408949

RESUMEN

Viruses are intracellular parasites that require a permissive host cell to express the viral genome and to produce new progeny virus particles. However, not all viral infections are productive and some viruses can induce carcinogenesis. Irrespective of the type of infection (productive or neoplastic), viruses hijack the host cell machinery to permit optimal viral replication or to transform the infected cell into a tumor cell. One mechanism viruses employ to reprogram the host cell is through interference with signaling pathways. Polyomaviruses are naked, double-stranded DNA viruses whose genome encodes the regulatory proteins large T-antigen and small t-antigen, and structural proteins that form the capsid. The large T-antigens and small t-antigens can interfere with several host signaling pathways. In this case, we review the interplay between the large T-antigens and small t-antigens with host signaling pathways and the biological consequences of these interactions.


Asunto(s)
Antígenos Virales de Tumores/inmunología , Infecciones por Polyomavirus/inmunología , Poliomavirus/inmunología , Transducción de Señal , Animales , Interacciones Huésped-Patógeno , Humanos , Poliomavirus/fisiología
15.
J Vis Exp ; (147)2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31107454

RESUMEN

Carboxyfluorescein succinimidyl ester (CFSE)-based in vivo cytotoxicity assays enable sensitive and accurate quantitation of CD8+ cytolytic T lymphocyte (CTL) responses elicited against tumor- and pathogen-derived peptides. They offer several advantages over traditional killing assays. First, they permit the monitoring of CTL-mediated cytotoxicity within architecturally intact secondary lymphoid organs, typically in the spleen. Second, they allow for mechanistic studies during the priming, effector and recall phases of CTL responses. Third, they provide useful platforms for vaccine/drug efficacy testing in a truly in vivo setting. Here, we provide an optimized protocol for the examination of concomitant CTL responses against more than one peptide epitope of a model tumor antigen (Ag), namely, simian virus 40 (SV40)-encoded large T Ag (T Ag). Like most other clinically relevant tumor proteins, T Ag harbors many potentially immunogenic peptides. However, only four such peptides induce detectable CTL responses in C57BL/6 mice. These responses are consistently arranged in a hierarchical order based on their magnitude, which forms the basis for TCD8 "immunodominance" in this powerful system. Accordingly, the bulk of the T Ag-specific TCD8 response is focused against a single immunodominant epitope while the other three epitopes are recognized and responded to only weakly. Immunodominance compromises the breadth of antitumor TCD8 responses and is, as such, considered by many as an impediment to successful vaccination against cancer. Therefore, it is important to understand the cellular and molecular factors and mechanisms that dictate or shape TCD8 immunodominance. The protocol we describe here is tailored to the investigation of this phenomenon in the T Ag immunization model, but can be readily modified and extended to similar studies in other tumor models. We provide examples of how the impact of experimental immunotherapeutic interventions can be measured using in vivo cytotoxicity assays.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Inmunoensayo/métodos , Epítopos Inmunodominantes/inmunología , Neoplasias/inmunología , Animales , Antígenos Virales de Tumores/inmunología , Epítopos de Linfocito T/inmunología , Ratones Endogámicos C57BL , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología
16.
Clin Cancer Res ; 25(1): 110-124, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30242022

RESUMEN

PURPOSE: Clinical responses with programmed death (PD-1) receptor-directed antibodies occur in about 20% of patients with advanced head and neck squamous cell cancer (HNSCCa). Viral neoantigens, such as the E6/E7 proteins of HPV16/18, are attractive targets for therapeutic immunization and offer an immune activation strategy that may be complementary to PD-1 inhibition. PATIENTS AND METHODS: We report phase Ib/II safety, tolerability, and immunogenicity results of immunotherapy with MEDI0457 (DNA immunotherapy targeting HPV16/18 E6/E7 with IL12 encoding plasmids) delivered by electroporation with CELLECTRA constant current device. Twenty-two patients with locally advanced, p16+ HNSCCa received MEDI0457. RESULTS: MEDI0457 was associated with mild injection site reactions, but no treatment-related grade 3-5 adverse events (AE) were noted. Eighteen of 21 evaluable patients showed elevated antigen-specific T-cell activity by IFNγ ELISpot, and persistent cellular responses surpassing 100 spot-forming units (SFUs)/106 peripheral blood mononuclear cells (PBMCs) were noted out to 1 year. Induction of HPV-specific CD8+ T cells was observed. MEDI0457 shifted the CD8+/FoxP3+ ratio in 4 of 5 post immunotherapy tumor samples and increased the number of perforin+ immune infiltrates in all 5 patients. One patient developed metastatic disease and was treated with anti-PD-1 therapy with a rapid and durable complete response. Flow-cytometric analyses revealed induction of HPV16-specific PD-1+ CD8+ T cells that were not found prior to MEDI0547 (0% vs. 1.8%). CONCLUSIONS: These data demonstrate that MEDI0457 can generate durable HPV16/18 antigen-specific peripheral and tumor immune responses. This approach may be used as a complementary strategy to PD-1/PD-L1 inhibition in HPV-associated HNSCCa to improve therapeutic outcomes.


Asunto(s)
Neoplasias de Cabeza y Cuello/terapia , Inmunoterapia , Infecciones por Papillomavirus/terapia , Vacunas contra Papillomavirus/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Adulto , Anciano , Antígenos Virales de Tumores/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/clasificación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/virología , Papillomavirus Humano 16/patogenicidad , Papillomavirus Humano 18/patogenicidad , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón gamma/genética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Receptor de Muerte Celular Programada 1/inmunología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
17.
J Cell Physiol ; 234(4): 3170-3179, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30362540

RESUMEN

Recent data indicate that the Simian virus 40 (SV40) infection appears to be transmitted in humans independently from early SV40-contaminated antipolio vaccines. Serum antibodies against SV40 large T antigen (Tag) were analyzed in children/adolescents and young adults. To investigate antibodies reacting to SV40 Tag antigens, serum samples ( n = 812) from children and young adults were analyzed by indirect ELISAs using specific SV40 Tag mimotopes. Mimotopes were synthetic peptides corresponding to SV40 Tag epitopes. In sera ( n = 412) from healthy children up to 17 years old, IgG antibodies against SV40 Tag mimotopes reached an overall prevalence of 15%. IgM antibodies against SV40 Tag were detected in sera of children 6-8 months old confirming and extending the knowledge that SV40 seroconversion occurs early in life. In children/adolescents affected by different diseases ( n = 180) SV40 Tag had a prevalence of 18%, being the difference no significant compared to healthy subjects ( n = 220; 16%) of the same age. Our immunological data indicate that SV40 circulates in children and young adults, both in healthy conditions and affected by distinct diseases. The IgM detection in sera from healthy children suggests that the SV40 infection/seroconversion occurs early in life (>6 months). Our immunological data support the hypothesis that SV40, or a closely related still unknown polyomavirus, infects humans. The SV40 seroprevalence is lower than common polyomaviruses, such as BKPyV and JCPyV, and other new human polyomaviruses. In addition, our immunological surveillance indicates a lack of association between different diseases, considered herein, and SV40.


Asunto(s)
Anticuerpos/sangre , Antígenos Virales de Tumores/inmunología , Epítopos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Infecciones por Polyomavirus/diagnóstico , Seroconversión , Virus 40 de los Simios/inmunología , Adolescente , Factores de Edad , Animales , Línea Celular , Niño , Preescolar , Chlorocebus aethiops , Ensayo de Inmunoadsorción Enzimática , Humanos , Lactante , Infecciones por Polyomavirus/sangre , Infecciones por Polyomavirus/inmunología
18.
Hum Vaccin Immunother ; 15(3): 658-668, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30513252

RESUMEN

OBJECTIVE: Antibody persistence evaluation for all antigens of a fully liquid DTaP-IPV-HB-PRP~T vaccine at 3.5 and 4.5 y of age following different primary series and booster schedules in South Africa and Latin America. METHODS: Participants had completed one of two previous studies (Study 1-South Africa; Study 2-Latin America). In Study 1, participants who had not received HB vaccine at birth received a 6-10-14 week primary series of DTaP-IPV-HB-PRP~T or DTwP/PRP~T-Hib+HB+OPV and a third group who had received HB vaccine at birth received a 6-10-14 week primary series of DTaP-IPV-HB-PRP~T; all received a booster (15-18 months) of the primary series vaccine(s) except for HB in the DTwP/PRP~T-Hib group. In Study 2, participants received HB vaccine at birth, a 2-4-6 month primary series of DTaP-IPV-HB-PRP~T or DTaP-HB-IPV//PRP~T, and a DTaP-IPV-HB-PRP~T or DTaP-HB-IPV//PRP~T booster (12-24 months). Participants were followed up at 3.5 and 4.5 y of age for antibody persistence. RESULTS: Approximately 80% of eligible participants were assessed. In Study 1, a birth dose of HB increased anti-HBs persistence (≥10 mIU/mL) following DTaP-IPV-HB-PRP~T primary and booster vaccination from 76.3% to 96.1% at 3.5 y of age and from 73.3% to 96.1% at 4.5 y of age; in Study 2, anti-HBs persistence was high and similar in each group. For the other antigens, there were no differences between groups or studies at 3.5 or 4.5 y. CONCLUSION: Good persistence of antibodies to each antigen in the DTaP-IPV-HB-PRP~T vaccine up to pre-school age, irrespective of the vaccination schedule during the first 2 y of life.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Anticuerpos Antivirales/sangre , Vacuna contra Difteria, Tétanos y Tos Ferina/inmunología , Vacunas contra Haemophilus/inmunología , Vacunas contra Hepatitis B/inmunología , Esquemas de Inmunización , Vacuna Antipolio de Virus Inactivados/inmunología , Antígenos Bacterianos/inmunología , Antígenos Virales de Tumores/inmunología , Niño , Preescolar , Vacuna contra Difteria, Tétanos y Tos Ferina/administración & dosificación , Femenino , Vacunas contra Haemophilus/administración & dosificación , Anticuerpos contra la Hepatitis B/sangre , Vacunas contra Hepatitis B/administración & dosificación , Humanos , Inmunización Secundaria , Lactante , Masculino , Vacuna Antipolio de Virus Inactivados/administración & dosificación , Estudios Prospectivos , Factores de Tiempo , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología
19.
Clin Cancer Res ; 24(15): 3644-3655, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669806

RESUMEN

Purpose: The causative agent of most cases of Merkel cell carcinoma (MCC) has been identified as the Merkel cell polyomavirus (MCV). MCV-encoded T antigens (Tag) are essential not only for virus-mediated tumorigenesis but also for maintaining MCC cell lines in vitro MCV Tags are thus an appealing target for viral oncoprotein-directed T-cell therapy for MCC. With this study, we aimed to isolate and characterize Tag-specific T-cell receptors (TCR) for potential use in gene therapy clinical trials.Experimental Design: T-cell responses against MCV Tag epitopes were investigated by immunizing transgenic mice that express a diverse human TCR repertoire restricted to HLA-A2. Human lymphocytes genetically engineered to express Tag-specific TCRs were tested for specific reactivity against MCC cell lines. The therapeutic potential of Tag-specific TCR gene therapy was tested in a syngeneic cancer model.Results: We identified naturally processed epitopes of MCV Tags and isolated Tag-specific TCRs. T cells expressing these TCRs were activated by HLA-A2-positive cells loaded with cognate peptide or cells that stably expressed MCV Tags. We showed cytotoxic potential of T cells engineered to express these TCRs in vitro and demonstrated regression of established tumors in a mouse model upon TCR gene therapy.Conclusions: Our findings demonstrate that MCC cells can be targeted by MCV Tag-specific TCRs. Although recent findings suggest that approximately half of MCC patients benefit from PD-1 pathway blockade, additional patients may benefit if their endogenous T-cell response can be augmented by infusion of transgenic MCV-specific T cells such as those described here. Clin Cancer Res; 24(15); 3644-55. ©2018 AACR.


Asunto(s)
Antígenos Virales de Tumores/uso terapéutico , Carcinoma de Células de Merkel/terapia , Terapia Genética , Linfocitos T/inmunología , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/inmunología , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinoma de Células de Merkel/genética , Carcinoma de Células de Merkel/inmunología , Carcinoma de Células de Merkel/virología , Citotoxicidad Inmunológica/genética , Epítopos/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/uso terapéutico , Humanos , Inmunoterapia , Linfocitos/inmunología , Poliomavirus de Células de Merkel/inmunología , Poliomavirus de Células de Merkel/patogenicidad , Ratones , Ratones Transgénicos , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/uso terapéutico
20.
Clin Cancer Res ; 24(9): 2035-2043, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29217527

RESUMEN

Merkel cell carcinoma (MCC) is a rare (∼2,000 U.S. cases/year) but aggressive neuroendocrine tumor of the skin. For advanced MCC, cytotoxic chemotherapy only infrequently (<10% of cases) offers durable clinical responses (>1 year), suggesting a great need for improved therapeutic options. In 2008, the Merkel cell polyomavirus (MCPyV) was discovered and is clonally integrated in approximately 80% of MCC tumors. The remaining 20% of MCC tumors have large numbers of UV-associated mutations. Importantly, both the UV-induced neoantigens in virus-negative tumors and the MCPyV T antigen oncogenes that are required for virus-positive tumor growth are immunogenic. Indeed, antigen-specific T cells detected in patients are frequently dysfunctional/"exhausted," and the inhibitory ligand, PD-L1, is often present in MCC tumors. These findings led to recent clinical trials involving PD-1 pathway blockade in advanced MCC. The combined data from these trials involving three PD-1 pathway blocking agents-avelumab, pembrolizumab, and nivolumab-indicated a high frequency of durable responses in treated patients. Of note, prior treatment with chemotherapy was associated with decreased response rates to PD-1 checkpoint blockade. Over the past year, these striking data led to major changes in advanced MCC therapy, including the first-ever FDA drug approval for this disease. Despite these successes, approximately 50% of patients with MCC do not persistently benefit from PD-1 pathway blockade, underscoring the need for novel strategies to broaden antitumor immune responses in these patients. Here, we highlight recent progress in MCC including the underlying mechanisms of immune evasion and emerging approaches to augment the efficacy of PD-1 pathway blockade. Clin Cancer Res; 24(9); 2035-43. ©2017 AACR.


Asunto(s)
Carcinoma de Células de Merkel/inmunología , Carcinoma de Células de Merkel/terapia , Inmunoterapia , Animales , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Antígenos Virales de Tumores/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor , Carcinoma de Células de Merkel/diagnóstico , Carcinoma de Células de Merkel/etiología , Terapia Combinada , Humanos , Inmunomodulación , Inmunoterapia/métodos , Poliomavirus de Células de Merkel/inmunología , Infecciones por Polyomavirus/complicaciones , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/virología , Receptor de Muerte Celular Programada 1/metabolismo , Nivel de Atención , Carga Tumoral , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA