Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Rep (Hoboken) ; 5(2): e1459, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34245130

RESUMO

BACKGROUND: Data are steadily accruing that demonstrate that intestinal tumors are frequently derived from multiple founding cells, resulting in tumors comprised of distinct ancestral clones that might cooperate or alternatively compete, thereby potentially impacting different phases of the disease process. AIM: We sought to determine whether tumors with a multi-ancestral architecture involving at least two distinct clones show increased tumor number, growth, progression, or resistance to drug intervention. METHODS: Mice carrying the Min allele of Apc were generated that were mosaic with only a subset of cells in the intestinal epithelium expressing an activated form of PI3K, a key regulatory kinase affecting several important cellular processes. These cells were identifiable as they fluoresced green, whereas all other cells fluoresced red. RESULTS: Cell lineage tracing revealed that many intestinal tumors from our mouse model were derived from at least two founding cells, those expressing the activated PI3K (green) and those which did not (red). Heterotypic tumors with a multi-ancestral architecture as evidenced by a mixture of green and red cells exhibited increased tumor growth and invasiveness. Clonal architecture also had an impact on tumor response to low-dose aspirin. Aspirin treatment resulted in a greater reduction of heterotypic tumors derived from multiple founding cells as compared to tumors derived from a single founding cell. CONCLUSION: These data indicate that genetically distinct tumor-founding cells can contribute to early intratumoral heterogeneity. The coevolution of the founding cells and their progeny enhances colon tumor progression and impacts the response to aspirin. These findings are important to a more complete understanding of tumorigenesis with consequences for several distinct models of tumor evolution. They also have practical implications to the clinic. Mouse models with heterogenous tumors are likely better for predicting drug efficacy as compared to models in which the tumors are highly homogeneous. Moreover, understanding how interactions among different populations in a single heterotypic tumor with a multi-ancestral architecture impact response to a single agent and combination therapies are necessary to fully develop personalized medicine.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasias Intestinais/genética , Animais , Antineoplásicos/farmacologia , Carcinogênese/genética , Carcinogênese/patologia , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Intestinais/tratamento farmacológico , Neoplasias Intestinais/patologia , Camundongos , Camundongos Transgênicos
2.
Oncoimmunology ; 10(1): 1912892, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33996265

RESUMO

Concurrent blockade of different checkpoint receptors, notably PD-1 and CTLA-4, elicits greater anti-tumor activity for some tumor types, and the combination of different checkpoint receptor inhibitors is an active area of clinical research. We have previously demonstrated that anti-tumor vaccination, by activating CD8 + T cells, increases the expression of PD-1, CTLA-4, LAG-3 and other inhibitory receptors, and the anti-tumor efficacy of vaccination can be increased with checkpoint blockade. In the current study, we sought to determine whether anti-tumor vaccination might be further improved with combined checkpoint blockade. Using an OVA-expressing mouse tumor model, we found that CD8 + T cells activated in the presence of professional antigen presenting cells (APC) expressed multiple checkpoint receptors; however, T cells activated without APCs expressed LAG-3 alone, suggesting that LAG-3 might be a preferred target in combination with vaccination. Using three different murine tumor models, and peptide or DNA vaccines targeting three tumor antigens, we assessed the effects of vaccines with blockade of PD-1 and/or LAG-3 on tumor growth. We report that, in each model, the anti-tumor efficacy of vaccination was increased with PD-1 and/or LAG-3 blockade. However, combined PD-1 and LAG-3 blockade elicited the greatest anti-tumor effect when combined with vaccination in a MycCaP prostate cancer model in which PD-1 blockade alone with vaccination targeting a "self" tumor antigen had less efficacy. These results suggest anti-tumor vaccination might best be combined with concurrent blockade of both PD-1 and LAG-3, and potentially other checkpoint receptors whose expression is increased on CD8 + T cells following vaccine-mediated activation.


Assuntos
Antígenos CD , Vacinas Anticâncer , Neoplasias , Receptor de Morte Celular Programada 1 , Vacinas de DNA , Animais , Linfócitos T CD8-Positivos , Masculino , Camundongos , Neoplasias/tratamento farmacológico , Proteína do Gene 3 de Ativação de Linfócitos
3.
Cancers (Basel) ; 12(10)2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-33008010

RESUMO

Metastatic castration-resistant prostate cancer (mCRPC) is a challenging disease to treat, with poor outcomes for patients. One antitumor vaccine, sipuleucel-T, has been approved as a treatment for mCRPC. DNA vaccines are another form of immunotherapy under investigation. DNA immunizations elicit antigen-specific T cells that cause tumor cell lysis, which should translate to meaningful clinical responses. They are easily amenable to design alterations, scalable for large-scale manufacturing, and thermo-stable for easy transport and distribution. Hence, they offer advantages over other vaccine formulations. However, clinical trials with DNA vaccines as a monotherapy have shown only modest clinical effects against tumors. Standard therapies for CRPC including androgen-targeted therapies, radiation therapy and chemotherapy all have immunomodulatory effects, which combined with immunotherapies such as DNA vaccines, could potentially improve treatment. In addition, many investigational drugs are being developed which can augment antitumor immunity, and together with DNA vaccines can further enhance antitumor responses in preclinical models. We reviewed the literature available prior to July 2020 exploring the use of DNA vaccines in the treatment of prostate cancer. We also examined various approved and experimental therapies that could be combined with DNA vaccines to potentially improve their antitumor efficacy as treatments for mCRPC.

4.
Commun Biol ; 2: 79, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30820474

RESUMO

Finding improved therapeutic strategies against T-cell Non-Hodgkin's Lymphoma (NHL) remains an unmet clinical need. We implemented a theranostic approach employing a tumor-targeting alkylphosphocholine (NM600) radiolabeled with 86Y for positron emission tomography (PET) imaging and 90Y for targeted radionuclide therapy (TRT) of T-cell NHL. PET imaging and biodistribution performed in mouse models of T-cell NHL showed in vivo selective tumor uptake and retention of 86Y-NM600. An initial toxicity assessment examining complete blood counts, blood chemistry, and histopathology of major organs established 90Y-NM600 safety. Mice bearing T-cell NHL tumors treated with 90Y-NM600 experienced tumor growth inhibition, extended survival, and a high degree of cure with immune memory toward tumor reestablishment. 90Y-NM600 treatment was also effective against disseminated tumors, improving survival and cure rates. Finally, we observed a key role for the adaptive immune system in potentiating a durable anti-tumor response to TRT, especially in the presence of microscopic disease.


Assuntos
Memória Imunológica/efeitos da radiação , Linfoma de Células T/radioterapia , Tomografia por Emissão de Pósitrons/métodos , Radioisótopos de Ítrio/uso terapêutico , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Memória Imunológica/imunologia , Linfoma de Células T/diagnóstico por imagem , Linfoma de Células T/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Distribuição Tecidual/imunologia , Distribuição Tecidual/efeitos da radiação , Carga Tumoral/imunologia , Carga Tumoral/efeitos da radiação , Proteína Tumoral 1 Controlada por Tradução , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Radioisótopos de Ítrio/sangue , Radioisótopos de Ítrio/farmacocinética
5.
Am J Cancer Res ; 9(1): 53-63, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30755811

RESUMO

Imaging of immunotherapy targets using positron emission tomography (PET) can allow for noninvasive monitoring of their dynamic expression and may allow for patient stratification in the future. Therefore, two tracers targeting CTLA-4, one a full antibody and the other a F(ab')2 fragment, were radiolabeled with 64Cu and validated in humanized mouse models. Ipilimumab was digested to develop ipilimumab-F(ab')2, and both the intact antibody and the fragment were conjugated with NOTA to chelate 64Cu for PET. The tracers were administered to both control NBSGW mice and humanized mice (PBL mice, engrafted with human peripheral blood lymphocytes), and PET was conducted out to 48 h post-injection. PET region-of-interest analysis, ex vivo biodistribution studies, and tissue staining were used to confirm that the tracers specifically accumulated in CTLA-4+ tissues. Following injection of tracers (n = 3-5 per group), specific uptake was noted in the salivary gland tissues of the humanized mice. This uptake, a result of graft-versus-host disease onset, was proven to be due to human T-cells through staining of the tissues for human CD3 and CTLA-4. 64Cu-NOTA-ipilimumab demonstrated the highest absolute uptake in the salivary glands of PBL mice, peaking at 7.00 ± 2.19 %ID/g. In contrast, 64Cu-NOTA-ipilimumab-F(ab')2 uptake was 2.40 ± 0.86 %ID/g at the same time point. However, the F(ab')2 agent cleared from circulation more quickly than the intact antibody, providing higher salivary gland-to-blood ratios, which reached 1.78 ± 0.72 at 48 h post-injection, compared to 64Cu-NOTA-ipilimumab at 1.19 ± 0.49. Uptake of the tracers in the salivary glands of control mice, and the nonspecific tracer in the PBL mice, was lower at all time points as well. PET imaging with both 64Cu-NOTA-ipilimumab and 64Cu-NOTA-ipilimumab-F(ab')2 was able to localize CTLA-4+ tissues. These tracers may thus help elucidate the mechanisms of response to CTLA-4-targeted checkpoint immunotherapy treatments.

6.
Cancer Immunol Res ; 6(11): 1364-1374, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30201735

RESUMO

Expression of T-cell checkpoint receptors can compromise antitumor immunity. Blockade of these receptors, notably PD-1 and LAG-3, which become expressed during T-cell activation with vaccination, can improve antitumor immunity. We evaluated whether T-cell checkpoint expression could be separated from T-cell activation in the context of innate immune stimulation with TLR agonists. We found that ligands for TLR1/2, TLR7, and TLR9 led to a decrease in expression of PD-1 on antigen-activated CD8+ T cells. These effects were mediated by IL12 released by professional antigen-presenting cells. In two separate tumor models, treatment with antitumor vaccines combined with TLR1/2 or TLR7 ligands induced antigen-specific CD8+ T cells with lower PD-1 expression and improved antitumor immunity. These findings highlight the role of innate immune activation during effector T-cell development and suggest that at least one mechanism by which specific TLR agonists can be strategically used as vaccine adjuvants is by modulating the expression of PD-1 during CD8+ T-cell activation. Cancer Immunol Res; 6(11); 1364-74. ©2018 AACR.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Ativação Linfocitária/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Receptores Toll-Like/agonistas , Transferência Adotiva , Aminoquinolinas/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Subunidade p35 da Interleucina-12/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Ovalbumina/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor de Morte Celular Programada 1/imunologia , Receptores Toll-Like/imunologia
7.
Cancer Immunol Res ; 5(8): 630-641, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28634215

RESUMO

Antitumor vaccines encoding self-antigens generally have low immunogenicity in clinical trials. Several approaches are aimed at improving vaccine immunogenicity, including efforts to alter encoded epitopes. Immunization with epitopes altered for increased affinity for the major histocompatibility complex (MHC) or T-cell receptor (TCR) elicits greater numbers of CD8 T cells but inferior antitumor responses. Our previous results suggested that programmed death 1 (PD-1) and its ligand (PD-L1) increased on antigen-specific CD8 T cells and tumor cells, respectively, after high-affinity vaccination. In this report, we use two murine models to investigate whether the dose, MHC affinity, or TCR affinity of an epitope affected the antitumor response via the PD-1/PD-L1 axis. T cells activated with high-affinity epitopes resulted in prolonged APC:T-cell contact time that led to elevated, persistent PD-1 expression, and expression of other checkpoint molecules, in vitro and in vivo Immunization with high-affinity epitopes also decreased antitumor efficacy in the absence of PD-1 blockade. Thus, APC:T-cell contact time can be altered by epitope affinity and lead to therapeutically relevant changes in vaccine efficacy mediated by changes in PD-1 expression. These findings have implications for the use of agents targeting PD-1 expression or function whenever high-affinity CD8 T cells are elicited or supplied by means of vaccination or adoptive transfer. Cancer Immunol Res; 5(8); 630-41. ©2017 AACR.


Assuntos
Antígeno B7-H1/imunologia , Vacinas Anticâncer/imunologia , Epitopos/imunologia , Neoplasias/terapia , Receptor de Morte Celular Programada 1/imunologia , Transferência Adotiva , Animais , Antígeno B7-H1/genética , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/genética , Receptores de Antígenos de Linfócitos T/imunologia , Vacinação/métodos
8.
Pharmacol Ther ; 174: 27-42, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28185916

RESUMO

DNA vaccines offer many advantages over other anti-tumor vaccine approaches due to their simplicity, ease of manufacturing, and safety. Results from several clinical trials in patients with cancer have demonstrated that DNA vaccines are safe and can elicit immune responses. However, to date few DNA vaccines have progressed beyond phase I clinical trial evaluation. Studies into the mechanism of action of DNA vaccines in terms of antigen-presenting cell types able to directly present or cross-present DNA-encoded antigens, and the activation of innate immune responses due to DNA itself, have suggested opportunities to increase the immunogenicity of these vaccines. In addition, studies into the mechanisms of tumor resistance to anti-tumor vaccination have suggested combination approaches that can increase the anti-tumor effect of DNA vaccines. This review focuses on these mechanisms of action and mechanisms of resistance using DNA vaccines, and how this information is being used to improve the anti-tumor effect of DNA vaccines. These approaches are then specifically discussed in the context of human prostate cancer, a disease for which DNA vaccines have been and continue to be explored as treatments.


Assuntos
Vacinas Anticâncer/imunologia , Neoplasias da Próstata/terapia , Vacinas de DNA/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Humanos , Imunidade Inata , Masculino , Neoplasias da Próstata/imunologia , Falha de Tratamento , Resultado do Tratamento
9.
Oncoimmunology ; 5(10): e1223002, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27853647

RESUMO

Increasing transgene expression has been a major focus of attempts to improve DNA vaccine-induced immunity in both preclinical studies and clinical trials. Novel mini-intronic plasmids (MIPs) have been shown to cause elevated and sustained transgene expression in vivo. We sought to test the antitumor activity of a MIP, compared to standard DNA plasmid immunization, using the tumor-specific antigen SSX2 in an HLA-A2-restricted tumor model. We found that MIP vaccination elicited a greater frequency of antigen-specific CD8+ T cells when compared to conventional plasmid, and protected animals from subsequent tumor challenge. However, therapeutic vaccination with the MIP resulted in an inferior antitumor effect, and CD8+ tumor-infiltrating lymphocytes from these mice expressed higher levels of surface LAG3. Antitumor efficacy of MIP vaccination could be recovered upon antibody blockade of LAG3. In non-tumor bearing mice, MIP immunization led to a loss of epitope dominance, attenuated CD8+ cytokine responses to the dominant p103 epitope, and increased LAG3 expression on p103-specific CD8+ T cells. Further, LAG3 expression on CD8+ T cells was associated with antigen dose and persistence in spite of DNA-induced innate immunity. These data suggest that for antitumor immunization, approaches leading to increased antigen expression following vaccination might optimally be combined with LAG3 inhibition in human trials. On the other hand, mini-intronic vector approaches may be a superior means to elicit LAG3-dependent tolerance in the treatment of autoimmune diseases.

10.
PLoS One ; 11(2): e0150170, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26919712

RESUMO

A widely accepted paradigm in the field of cancer biology is that solid tumors are uni-ancestral being derived from a single founder and its descendants. However, data have been steadily accruing that indicate early tumors in mice and humans can have a multi-ancestral origin in which an initiated primogenitor facilitates the transformation of neighboring co-genitors. We developed a new mouse model that permits the determination of clonal architecture of intestinal tumors in vivo and ex vivo, have validated this model, and then used it to assess the clonal architecture of adenomas, intramucosal carcinomas, and invasive adenocarcinomas of the intestine. The percentage of multi-ancestral tumors did not significantly change as tumors progressed from adenomas with low-grade dysplasia [40/65 (62%)], to adenomas with high-grade dysplasia [21/37 (57%)], to intramucosal carcinomas [10/23 (43%]), to invasive adenocarcinomas [13/19 (68%)], indicating that the clone arising from the primogenitor continues to coexist with clones arising from co-genitors. Moreover, neoplastic cells from distinct clones within a multi-ancestral adenocarcinoma have even been observed to simultaneously invade into the underlying musculature [2/15 (13%)]. Thus, intratumoral heterogeneity arising early in tumor formation persists throughout tumorigenesis.


Assuntos
Adenocarcinoma/patologia , Adenoma/patologia , Carcinoma in Situ/patologia , Transformação Celular Neoplásica , Neoplasias Intestinais/patologia , Células-Tronco Neoplásicas/patologia , Adenocarcinoma/genética , Adenoma/genética , Animais , Carcinoma in Situ/genética , Linhagem da Célula , Transformação Celular Neoplásica/genética , Células Clonais/patologia , Modelos Animais de Doenças , Progressão da Doença , Evolução Molecular , Proteínas de Ligação a Ácido Graxo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Genes APC , Genes Reporter , Integrases/genética , Mucosa Intestinal/patologia , Neoplasias Intestinais/genética , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mosaicismo , Invasividade Neoplásica , RNA não Traduzido/genética , Ratos , Transgenes , Microambiente Tumoral
11.
Cancer Prev Res (Phila) ; 7(1): 105-13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24196829

RESUMO

Colorectal cancer often arises from adenomatous colonic polyps. Polyps can grow and progress to cancer, but may also remain static in size, regress, or resolve. Predicting which polyps progress and which remain benign is difficult. We developed a novel long-lived murine model of colorectal cancer with tumors that can be followed by colonoscopy. Our aim was to assess whether these tumors have similar growth patterns and histologic fates to human colorectal polyps to identify features to aid in risk stratification of colonic tumors. Long-lived Apc(Min/+) mice were treated with dextran sodium sulfate to promote colonic tumorigenesis. Tumor growth patterns were characterized by serial colonoscopy with biopsies obtained for immunohistochemistry and gene expression profiling. Tumors grew, remained static, regressed, or resolved over time with different relative frequencies. Newly developed tumors demonstrated higher rates of growth and resolution than more established tumors that tended to remain static in size. Colonic tumors were hyperplastic lesions (3%), adenomas (73%), intramucosal carcinomas (20%), or adenocarcinomas (3%). Interestingly, the level of ß-catenin was higher in adenomas that became intratumoral carcinomas than those that failed to progress. In addition, differentially expressed genes between adenomas and intramucosal carcinomas were identified. This novel murine model of intestinal tumorigenesis develops colonic tumors that can be monitored by serial colonoscopy, mirror growth patterns seen in human colorectal polyps, and progress to colorectal cancer. Further characterization of cellular and molecular features is needed to determine which features can be used to risk-stratify polyps for progression to colorectal cancer and potentially guide prevention strategies.


Assuntos
Pólipos do Colo/patologia , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Proteína da Polipose Adenomatosa do Colo/genética , Pólipos Adenomatosos/patologia , Animais , Colonoscopia , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 110(28): 11523-8, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23798428

RESUMO

Intestinal tumors from mice and humans can have a polyclonal origin. Statistical analyses indicate that the best explanation for this source of intratumoral heterogeneity is the presence of interactions among multiple progenitors. We sought to better understand the nature of these interactions. An initial progenitor could recruit others by facilitating the transformation of one or more neighboring cells. Alternatively, two progenitors that are independently initiated could simply cooperate to form a single tumor. These possibilities were tested by analyzing tumors from aggregation chimeras that were generated by fusing together embryos with unequal predispositions to tumor development. Strikingly, numerous polyclonal tumors were observed even when one genetic component was highly, if not completely, resistant to spontaneous tumorigenesis in the intestine. Moreover, the observed number of polyclonal tumors could be explained by the facilitated transformation of a single neighbor within 144 µm of an initial progenitor. These findings strongly support recruitment instead of cooperation. Thus, it is conceivable that these interactions are necessary for tumors to thrive, so blocking them might be a highly effective method for preventing the formation of tumors in the intestine and other tissues.


Assuntos
Transformação Celular Neoplásica , Neoplasias Intestinais/patologia , Animais , Células Epiteliais/patologia , Genes APC , Humanos , Neoplasias Intestinais/genética , Camundongos , Camundongos Endogâmicos C57BL
13.
Cancer Res ; 72(12): 2931-6, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22525701

RESUMO

Aberrations in the phosphoinositide 3-kinase (PI3K) signaling pathway play a key role in the pathogenesis of numerous cancers by altering cellular growth, metabolism, proliferation, and apoptosis. Mutations in the catalytic domain of PI3K that generate a dominantly active kinase are commonly found in human colorectal cancers and have been thought to drive tumor progression but not initiation. However, the effects of constitutively activated PI3K upon the intestinal mucosa have not been previously studied in animal models. Here, we show that the expression of a dominantly active form of the PI3K protein in the mouse intestine results in hyperplasia and advanced neoplasia. Mice expressing constitutively active PI3K in the epithelial cells of the distal small bowel and colon rapidly developed invasive adenocarcinomas in the colon that spread into the mesentery and adjacent organs. The histologic characteristics of these tumors were strikingly similar to invasive mucinous colon cancers in humans. Interestingly, these tumors formed without a benign polypoid intermediary, consistent with the lack of aberrant WNT signaling observed. Together, our findings indicate a noncanonical mechanism of colon tumor initiation that is mediated through activation of PI3K. This unique model has the potential to further our understanding of human disease and facilitate the development of therapeutics through pharmacologic screening and biomarker identification.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Apoptose , Biomarcadores Tumorais , Proliferação de Células , Transformação Celular Neoplásica , Neoplasias Colorretais/metabolismo , Ativação Enzimática , Genótipo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Via de Sinalização Wnt
14.
Cancer Prev Res (Phila) ; 4(6): 916-23, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21636550

RESUMO

Previous studies have shown that intestinal tumors from Apc(Min)(/+) (Min) mice and familial adenomatous polyposis (FAP) patients are often polyclonal. We sought to determine whether polyclonality is unique to tumors arising from hereditary predispositions or, instead, is a common feature of intestinal tumorigenesis in other pathways to tumorigenesis. Ethylnitrosourea-induced intestinal tumors from mice wild type at the Apc locus and chimeric for the Rosa26 lineage marker were analyzed. Many were overtly polyclonal, being composed of a mixture of Rosa26(+) and Rosa26(-) neoplastic cells. Statistical analyses revealed that polyclonality could be explained by interactions between two initiated clones separated by a very short distance. The frequency of overtly polyclonal tumors and the range of interactions estimated in this model are similar to those observed when analyzing familial tumors from Min mice. Thus, polyclonality does not depend on the familial pathway to tumorigenesis. Interactions between two initiated clones might provide a selective advantage during the early stages of intestinal tumorigenesis.


Assuntos
Neoplasias Intestinais/classificação , Neoplasias Intestinais/patologia , Proteínas/fisiologia , Polipose Adenomatosa do Colo , Animais , Carcinógenos/toxicidade , Transformação Celular Neoplásica , Células Cultivadas , Quimerismo , Células Clonais , Etilnitrosoureia/toxicidade , Feminino , Genes APC/fisiologia , Neoplasias Intestinais/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , RNA não Traduzido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...