• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 23
  • 8
  • 2
  • 1
  • 1
  • Tagged with
  • 38
  • 38
  • 14
  • 12
  • 12
  • 12
  • 10
  • 9
  • 8
  • 8
  • 8
  • 7
  • 7
  • 7
  • 6
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
31

Remediação das vias p53/Arf e interferon-beta como uma estratégia de imunoterapia do câncer: uma abordagem de transferência gênica / Remediation of the p53/Arf and Interferon-beta pathways as a cancer immunotherapy strategy: a gene transfer approach

Ruan Felipe Vieira Medrano 08 January 2018 (has links)
As células tumorais prosperam como consequência da capacidade de resistir aos mecanismos de morte celular e de evasão da vigilância imunológica. Nós propomos que, em cânceres que possuem o supressor de tumor p53 selvagem, a remediação de ambas dessas defesas pode ser promovida pela transferência genica combinada de vetores adenovirais portadores dos transgenes de p19Arf (proteína supressora de tumor, parceira funcional de p53) e de interferon-beta (IFNbeta, citocina imunomoduladora). De fato, em resultados anteriores, notamos que a transdução combinada (p19Arf/IFNbeta), mas não os tratamentos individuais, em células de melanoma murino B16F10 resulta em aumento massivo de morte celular. Porém a capacidade destas células em processo de morte de desencadear imunidade antitumoral não foi analisada. Nesta tese e em estudos complementares, buscamos investigar os mecanismos moleculares de morte celular envolvidos na resposta imune estimulada por p19Arf/IFNbeta e explorar sua aplicação como imunoterapia do câncer. Inicialmente, em modelo de vacinação profilática, revelamos que o tratamento combinado em células B16F10 promove a expressão de IL-15, ULBP1, dos receptores de morte FAS/APO1 e KILLER/DR5, assim como uma resposta de células natural killer que rejeitam estas células tratadas quando inoculadas em camundongos imunocompetentes singênicos. Após desafio tumoral no flanco oposto, a progressão desses tumores foi fortemente reduzida devido ao engajamento de linfócitos T CD4+ e CD8+, que apresentaram produção aumentada das citocinas IFN-? e TNF-alfa e medeiam proteção antitumoral de longo prazo. Em seguida, explorando um contexto de imunização diferente, a transferência de gênica in situ foi realizada em carcinoma heterotópico de pulmão e exibiu proteção significativa contra um desafio tumoral secundário, apenas quando o tumor primário foi tratado com p19Arf/IFNbeta. Análise de transcriptoma destes tumores indicou uma assinatura quimiotáxica de neutrófilos e linfócitos T CD8+ através das quimiocinas CCL3, CXCL3 e da IL-1beta. Em apoio destas observações, análises mecanicistas in vitro revelaram que células tratadas com p19Arf/IFNbeta ativam programas apoptóticos de p53 e antivirais de IFNbeta, enquanto sucumbem a um processo de morte por necroptose que também libera moléculas de morte celular imunogênica (MCI), calreticulina, ATP e HMGB1. No entanto, procurando potencializar ainda mais o benefício terapêutico dos nossos vetores, exploramos sua associação com o quimioterápico imunogênico doxorrubicina (Dox), que também é indutor de MCI. E nesta associação, percebemos que a Dox aumenta não apenas os níveis de morte celular, mas também a imunogenicidade das células tratadas, proporcionando em um modelo de vacina terapêutica, um controle tumoral superior em camundongos que já portavam antes da vacinação tumores B16F10 ou MCA205. Além disso, a associação in situ destas terapias restaurou a eficácia de uma dose sub-terapêutica de Dox, que em contraste com sua dose terapêutica, não prejudica a função cardíaca. Finalmente, também exploramos a associação com o bloqueio dos pontos de controle imunológicos PD-1 ou CTLA-4, que no modelo de vacina terapêutica, sua associação induziu maior rejeição completa de tumores B16F10. Em conclusão, aqui apresentamos evidências sobre a capacidade da combinação p19Arf/IFNbeta de induzir morte celular e estimulação imunológica. E ressaltamos seu potencial como uma estratégia de imunoterapia do câncer / Cancer cells thrive as a consequence of resisting cell death mechanisms and escaping from immune surveillance. We propose that, in cancers that harbor the wild-type tumor suppressor p53, remediation of both of these defenses can be achieved by harnessing the adenoviral vector mediated gene transfer of p19Arf (tumor suppressor protein, p53 functional partner) together with interferon-beta (IFNbeta, immunomodulatory cytokine). Indeed, in our initial observations, it was noticed that combined-transduction (p19Arf/IFNbeta), but not the individual treatments, of B16F10 mouse melanoma cells results in massive cell death levels. Yet, the capability of these dying cells to unleash antitumor immunity was not investigated. Here in this thesis and in complementary studies, we sought to investigate the molecular mechanisms of cell death involved in the p19Arf/IFNbeta immune stimulation and explore its potential as a mediator of cancer immunotherapy. First, in a prophylactic B16F10 vaccine model, we revealed that the dual treatment led to the up-regulation of IL-15, ULBP1, FAS/APO1 and KILLER/DR5 death receptors, plus a natural killer cell response that completely rejects treated cells when inoculated in syngeneic immunocompetent mice. Whereas, upon a contralateral tumor challenge, progression was strongly reduced by engaging both CD4+ and CD8+ T cells, which displayed augmented production of IFN-? and TNF-alpha cytokines and provided long term antitumor protection. Next, exploring different immunization context, in situ gene transfer in a heterotopic lung carcinoma exhibited significant protection against a secondary tumor challenge only when the primary tumor was treated with p19Arf/IFNbeta. Transcriptome analysis of these treated tumors indicated a chemotaxic signature of neutrophils and CD8+ T cells with the involvement of CCL3, CXCL3 chemokines and IL-1beta. Moreover, in support of this evidence, mechanistic in vitro studies revealed that p19Arf/IFNbeta treated cells reactivate p53 apoptotic and IFNbeta antiviral programs, while succumbing to a necroptosis cell death processes that also releases immunogenic cell death (ICD) molecules, calreticulin, ATP and HMGB1. Yet, aiming to potentiate therapeutic benefit of our vectors, we explored their association with doxorubicin (Dox) immunogenic chemotherapy, which is also an inducer of ICD. And in this setting, this association with Dox enhances not only cell death levels but also immunogenicity of treated cells, providing superior tumor control in a therapeutic vaccine model, where mice were already bearing B16F10 tumors or MCA205 sarcomas before vaccination. Moreover, associated use of these therapies in situ rescued efficacy of a sub-therapeutic dose of Dox, which in contrast to its therapeutic dose, does not impair cardiac function. Finally, we also evaluated the association with PD-1 or CTLA-4 checkpoint blockade immunotherapy, which in the therapeutic vaccine model induced full tumor rejection in a greater number of mice. In sum, here we provide compelling evidence for the ability of the p19Arf/IFNbeta combined gene transfer to promote cell death and immunogenic stimuli and underscored its potential to be applied as a cancer immunotherapy strategy
32

Etude du rôle des lymphocytes T régulateurs dans la régulation des réponses immunes antitumorales induites par vaccination

Nuttin, Lise 17 February 2011 (has links)
Le système immunitaire est capable de rejeter une tumeur, même si l'interaction entre système immun et tumeurs est et reste complexe. Contrôler à long terme la croissance de la tumeur est un challenge, probablement suite à divers mécanismes de tolérance centrale et périphérique. Différentes approches d'immunothérapie du cancer ont été et sont toujours développées. Nous avons, au laboratoire, investigué et démontré, dans le cadre de deux différents modèles tumoraux murins, les capacités thérapeutiques d’une nouvelle stratégie de vaccinations combinées associant des injections de cellules dendritiques (DC) et de cellules tumorales sécrétrices de GM-CSF.<p><p>Notre travail de thèse a consisté à poursuivre l'étude de cette stratégie de vaccins combinés en étudiant plus particulièrement le rôle des lymphocytes T régulateurs (Treg) dans l'efficacité thérapeutique des vaccinations. <p>Les Treg regroupent différentes populations cellulaires immunosuppressives dérivées du thymus qui jouent un rôle clé dans le maintien de la tolérance périphérique. Les Treg naturels dont le mécanisme de suppression principal nécessite un contact cellule-cellule, expriment de façon constitutive les molécules de surface CD4, CD25 et CTLA-4 mais le marqueur le plus spécifique est le facteur de transcription Foxp3 qui est indispensable à leur développement et à leur fonction suppressive. Dans un premier temps, nous avons donc caractérisé les Treg de rat pour l'expression de Foxp3 et la nature et spécificité antigénique de leur fonction suppressive. Nos résultats démontrent d’une part, une expression de Foxp3 restreinte aux LT CD25+ natifs et liée à une fonction suppressive s'exerçant par contact cellule-cellule et d’autre part, une spécificité antigénique non-restreinte de ces Treg. <p><p>Ensuite, nous avons utilisé ce modèle de vaccination associant des injections de DC et de cellules tumorales sécrétrices de GM-CSF pour analyser comparativement les réponses immunes induites chez les rats vaccinés guéris ou non-guéris et les rats contrôles non vaccinés et identifier les paramètres cruciaux conduisant à l'éradication de la tumeur. Nos résultats ont montré que la principale différence entre rats vaccinés guéris et non guéris ne réside pas dans l'induction de réponses cytotoxiques systémiques spécifiques de la tumeur. Par contre, la guérison est associée à la persistance d'une réponse systémique LT CD4+ TH1 ainsi qu'au recrutement important en intratumoral de LT CD8+ cytotoxiques lié à une faible proportion relative de Treg. <p><p>Comme dans la majorité des études publiées chez l'animal, les DC utilisées dans ces vaccins combinés ont été générées à partir de rats naïfs, par différenciation de précurseurs de la moëlle osseuse en présence de GM-CSF seul. Par analogie avec les vaccins DC administrés aux patients cancéreux, nous avons aussi dérivé des DC à partir de la moëlle osseuse de rats porteurs d’une tumeur (TUM+) et nous avons constaté que ces mêmes vaccins combinés montraient in vivo une efficacité thérapeutique nettement moins bonne que leurs équivalents naïfs. Nous avons établi que cette différence d’efficacité était liée à la présence de Treg fonctionnels dans les vaccins DC dérivés de rats TUM+. Puis, nous avons établi le lien entre la présence de Treg dans les vaccins DC TUM+ et leur moins bonne efficacité thérapeutique en montrant qu'un traitement in vivo des rats TUM+ au témozolomide (TMZ) avant de générer les vaccins DC résultait in vitro, en une moindre expression de Foxp3 et une fonction suppressive diminuée et in vivo, en une bien meilleure survie des rats vaccinés.<p><p>Enfin, dans le but d’une utilisation future en clinique, nous avons développé une approche simplifiée ‘tout in vivo’ de notre modèle de vaccinations combinées, en utilisant la tumeur localement irradiée in vivo comme source d’antigène pour des DC autologues injectées en péri-tumoral et un apport exogène de GM-CSF. Nous avons utilisé des adénovirus associés recombinants porteurs du gène du GM-CSF (AAV1-GM-CSF) pour transduire la tumeur in vivo. Ces injections intratumorales d’AAV1-GM-CSF ont montré de bons résultats en vaccinations combinées puisque 60% des rats ont pu être guéris d’une tumeur 9L pré-implantée. Nous avons ensuite expérimenté l’enrobage des AAV1-GM-CSF dans un polymère biocompatible et thermosensible, le poloxamère, avant de les injecter en intratumoral, sans observer de meilleur effet thérapeutique. Cependant, nous avons constaté que l'utilisation du poloxamère pour enrober du GM-CSF recombinant permettait d'améliorer nettement la survie des rats vaccinés par comparaison à l'utilisation de GM-CSF recombinant seul.<p><p>La stratégie de vaccinations combinées que nous avons largement explorée et validée chez le rat serait une alternative intéressante à développer en clinique, particulièrement en combinaison avec un traitement permettant d'éliminer les Treg à la fois dans les vaccins et chez les patients vaccinés.<p> / Doctorat en Sciences biomédicales et pharmaceutiques / info:eu-repo/semantics/nonPublished
33

Investigação da resposta imunológica antitumoral induzida por células B16F10 tratadas pela combinação p19Arf e interferon-beta em um modelo de vacinação profilático para melanoma murino / Investigation of the antitumor immune response induced by B16F10 cells treated with the p19Arf and Interferon-beta combination in a murine prophylatic model of melanoma vaccine

Medrano, Ruan Felipe Vieira 25 April 2013 (has links)
Dados recentes do nosso laboratório demonstram que somente a co-transdução, não a tradução individual, com vetores adenovirais portadores de Interferon-beta (IFN?) (citocina imuno modulatória) e p19Arf (parceira funcional da proteína supressora de tumor p53) resulta na morte celular massiva do melanoma murino B16F10. A capacidade desse tratamento combinado de induzir uma resposta imune antitumoral ainda não foi avaliada. Dessa maneira, o objetivo do presente trabalho foi investigar se células B16F0 tratadas por essa combinação são capazes de induzir uma resposta imune antitumoral em um modelo de vacinação profilático de melanoma. Para isso, essas células foram co-transduzidas com os vetores AdPGp19 e AdPGIFN? e 48 horas depois, inoculadas como agente vacinal no flanco esquerdo (sítio da vacina) de camundongos C57BL/6 imunocompetentes. Sete dias após a última vacinação, esses animais foram desafiados com células B16F10 naïve no flanco direito (sítio do desafio). A progressão tumoral do desafio foi significativamente reduzida, mesmo quando o desafio tumoral foi feito 73 dias após da vacinação. Porém, como os animais imunizados desenvolveram tumores no sítio da vacina, condições para o uso dessas células tratadas foram avaliadas, revelando que: o número de células e de aplicações usadas durante a vacinação tem influência no aparecimento desse tumores, e que apenas com o tratamento combinado os camundongos permanecem livres de tumor. A influência do sistema imune para este resultado foi revelada após protocolo de imunussupressão. Em seguida, o papel da p19Arf e do IFN? na proteção antitumoral da combinação foi estudado. In vitro, os efeitos antitumorais da combinação parecem ser mais influentes da reposição de p19Arf do que da expressão de IFN?, mas já in vivo, na presença do sistema imune, foram mais dependentes do IFN?. Com a combinação estes efeitos mostraram-se mais pronunciados, induzindo uma proteção antitumoral e maior sobrevida aos animais vacinados. Estes resultados indicam que a combinação p19Arf e IFN? pode ser aplicada como um agente imunoterápico e sugerem que a associação entre morte celular e imuno estimulação pode beneficiar o tratamento contra o câncer / Previously, we have shown in a mouse melanoma model of in situ gene therapy that co-transduction, but not individual application, with adenovirus vectors expressing the Interferon-beta (IFN?) (immune modulatory cytokine) and p19Arf (functional partner of the p53 tumor suppressor) transgenes results in massive cell death and reduced tumor progression. However, the capability of this combined treatment to stimulate an antitumor immune response has not been evaluated. Therefore, the aim of this work was to investigate, trough a prophylactic vaccine model, if B16F10 cells treated by the p19Arf and IFN? combination could induce such immune response. To do so, these cells were co-transduced by the AdPGp19 e AdPGIFN? adenoviral vectors and 48 hours after, inoculated as a vaccine agent in the left flank (vaccine site) of immune competent C57BL/6 mice. Seven days after the last vaccine, a tumor challenge was done with naïve B16F10 cells in the right flank (challenge site). Tumor progression was markedly reduced, even when challenge was done 73 days after the vaccination. However, since these animals developed tumors where the vaccine was applied, more appropriate conditions for the use of these treated cells were pursued, thus revealing that: the number of cells and inoculations can dictate tumor development, and also, that only with the combined treatment was tumor formation abolished. The influence of the immune system for this result was revelead by performing an immune supression protocol. Next, the roles of p19Arf and of IFN? were studied. In vitro, the antitumor effects were stronger upon the introduction of p19Arf than IFN?, but in vivo, in the presence of the immune system, the effects were more IFN? dependent. In fact, these effects were more pronouced with the combined treatment, inducing protection against tumor formation and progression and increasing survival in the vaccinated animals. Taken together, these results demonstrate the application of cells treated by the p19Arf e IFN? combination as an effective vaccine agent and also indicates that the association between cell death and immune stimulation may benefit the treatment of cancer
34

Investigação da resposta imunológica antitumoral induzida por células B16F10 tratadas pela combinação p19Arf e interferon-beta em um modelo de vacinação profilático para melanoma murino / Investigation of the antitumor immune response induced by B16F10 cells treated with the p19Arf and Interferon-beta combination in a murine prophylatic model of melanoma vaccine

Ruan Felipe Vieira Medrano 25 April 2013 (has links)
Dados recentes do nosso laboratório demonstram que somente a co-transdução, não a tradução individual, com vetores adenovirais portadores de Interferon-beta (IFN?) (citocina imuno modulatória) e p19Arf (parceira funcional da proteína supressora de tumor p53) resulta na morte celular massiva do melanoma murino B16F10. A capacidade desse tratamento combinado de induzir uma resposta imune antitumoral ainda não foi avaliada. Dessa maneira, o objetivo do presente trabalho foi investigar se células B16F0 tratadas por essa combinação são capazes de induzir uma resposta imune antitumoral em um modelo de vacinação profilático de melanoma. Para isso, essas células foram co-transduzidas com os vetores AdPGp19 e AdPGIFN? e 48 horas depois, inoculadas como agente vacinal no flanco esquerdo (sítio da vacina) de camundongos C57BL/6 imunocompetentes. Sete dias após a última vacinação, esses animais foram desafiados com células B16F10 naïve no flanco direito (sítio do desafio). A progressão tumoral do desafio foi significativamente reduzida, mesmo quando o desafio tumoral foi feito 73 dias após da vacinação. Porém, como os animais imunizados desenvolveram tumores no sítio da vacina, condições para o uso dessas células tratadas foram avaliadas, revelando que: o número de células e de aplicações usadas durante a vacinação tem influência no aparecimento desse tumores, e que apenas com o tratamento combinado os camundongos permanecem livres de tumor. A influência do sistema imune para este resultado foi revelada após protocolo de imunussupressão. Em seguida, o papel da p19Arf e do IFN? na proteção antitumoral da combinação foi estudado. In vitro, os efeitos antitumorais da combinação parecem ser mais influentes da reposição de p19Arf do que da expressão de IFN?, mas já in vivo, na presença do sistema imune, foram mais dependentes do IFN?. Com a combinação estes efeitos mostraram-se mais pronunciados, induzindo uma proteção antitumoral e maior sobrevida aos animais vacinados. Estes resultados indicam que a combinação p19Arf e IFN? pode ser aplicada como um agente imunoterápico e sugerem que a associação entre morte celular e imuno estimulação pode beneficiar o tratamento contra o câncer / Previously, we have shown in a mouse melanoma model of in situ gene therapy that co-transduction, but not individual application, with adenovirus vectors expressing the Interferon-beta (IFN?) (immune modulatory cytokine) and p19Arf (functional partner of the p53 tumor suppressor) transgenes results in massive cell death and reduced tumor progression. However, the capability of this combined treatment to stimulate an antitumor immune response has not been evaluated. Therefore, the aim of this work was to investigate, trough a prophylactic vaccine model, if B16F10 cells treated by the p19Arf and IFN? combination could induce such immune response. To do so, these cells were co-transduced by the AdPGp19 e AdPGIFN? adenoviral vectors and 48 hours after, inoculated as a vaccine agent in the left flank (vaccine site) of immune competent C57BL/6 mice. Seven days after the last vaccine, a tumor challenge was done with naïve B16F10 cells in the right flank (challenge site). Tumor progression was markedly reduced, even when challenge was done 73 days after the vaccination. However, since these animals developed tumors where the vaccine was applied, more appropriate conditions for the use of these treated cells were pursued, thus revealing that: the number of cells and inoculations can dictate tumor development, and also, that only with the combined treatment was tumor formation abolished. The influence of the immune system for this result was revelead by performing an immune supression protocol. Next, the roles of p19Arf and of IFN? were studied. In vitro, the antitumor effects were stronger upon the introduction of p19Arf than IFN?, but in vivo, in the presence of the immune system, the effects were more IFN? dependent. In fact, these effects were more pronouced with the combined treatment, inducing protection against tumor formation and progression and increasing survival in the vaccinated animals. Taken together, these results demonstrate the application of cells treated by the p19Arf e IFN? combination as an effective vaccine agent and also indicates that the association between cell death and immune stimulation may benefit the treatment of cancer
35

Gendering Gardasil: Framing Gender and Sexuality in Media Representations of the HPV Vaccine

Pisciotta, Maura Kathleen 01 January 2012 (has links)
In an age of biomedicine, technologies, drugs, and treatments are expanding in new and diverse ways. Especially relevant to biomedicalization and this research is how such information is conveyed to the public through the media. Medical information is omnipresent in the media through direct-to-consumer (DTC) advertising and regular coverage of health topics in the news. The accessibility and proliferation of medical information provides an important opportunity to examine the ways in which these topics are framed in the media. This research specifically examines the framing of the HPV vaccine, Gardasil in the mass media. In this study, I explore how Gardasil was framed, how gender and sexuality were utilized within such frames, and what groups influenced these frames. I employ frame analysis to examine the presentation of Gardasil in the mass media. Due to the vaccine's intricate connection to gender and sexuality, I examine how these dimensions are utilized and reproduced in such frames. Gardasil was originally approved only for women, making gender a salient aspect of the vaccine. The current body of research examining Gardasil in the media presents data from the time the vaccine was only available to women. Now that the HPV vaccine is approved for men, this research seeks to contribute to a more comprehensive understanding of how Gardasil was framed in the media now that it is available to men and women. And given that Gardasil prevents a sexually transmitted infection (STI) in men and women, it is uniquely tied to issues of sexuality. To analyze Gardasil in the media, I examine newspaper articles, magazine articles, and media representations from stakeholder groups, including DTC advertising, official statements, and group websites. Analysis of key sources indicates that Merck dominated the framing of the vaccine in DTC advertisements and the news media, illustrating the power of the pharmaceutical industry. Findings indicate that the initial marketing of Gardasil constructed the vaccine as uniquely tied to femininity and later, women's empowerment. However, once the drug was approved for men, messages were reframed to appeal to a wider audience. Overall, the media continued to frame the vaccine specifically for women, further constructing HPV as a "woman's disease." The dominant focus on women concomitantly silenced the sexual health of men and sexual minorities. In conclusion, the marketing, discourse, and structural elements of Gardasil make it less accessible to those most in need, therefore contributing to the ongoing problem of cervical cancer and HPV.
36

Exploring the landscape of actionable HLA I-associated tumor antigens across cancers

Apavaloaei, Anca 08 1900 (has links)
Presque toutes les cellules nucléées expriment des peptides associés au CMH I (HLA I chez l’humain)(MAP) qui sont échantillonnés à partir du protéome cellulaire et transportés vers la surface cellulaire pour inspection par les lymphocytes T CD8. En tant que tel, la collection de MAP à la surface des cellules, ou immunopeptidome, informe les lymphocytes T CD8 de l’état cellulaire interne. L’immunosurveillance du cancer repose sur la capacité des lymphocytes T CD8 à reconnaître les MAP anormaux sur les cellules tumorales et à les éliminer tout en épargnant les cellules saines. Par conséquent, l’existence du cancer indique que bien souvent, les lymphocytes T CD8 spécifiques à la tumeur sont impuissants, dysfonctionnels ou incapables d’exercer leur fonction. Les vaccins anticancéreux peuvent actionner la destruction des tumeurs en stimulant la reconnaissance des MAP anormaux. Toutefois, le développement de vaccins anticancéreux efficaces est entravé par le manque de MAP exploitables, ou antigènes tumoraux (TA), exprimés exclusivement sur les cellules tumorales. La recherche et l’identification de TA ont été largement limitées aux MAP dérivés de mutations non synonymes situées dans des exons canoniques codant pour des protéines. Ces régions génomiques ne représentent que 2% du génome humain. Le fait que les MAP puissent potentiellement dériver de la traduction non canonique de toutes les régions génomiques n’a été pleinement compris que récemment. Ici, nous avons utilisé la protéogénomique pour découvrir des TA exploitables dérivés de produits de traduction canoniques et non canoniques partagés au sein ou entre divers types de cancers humains. Premièrement, nous avons utilisé des cellules souches pluripotentes induites (iPSC) pour identifier les MAP associés à la pluripotence (paMAP) étant partagés par les cellules cancéreuses. Les antigènes pluripotents sont exprimés dans les tissus embryonnaires et absents des tissus adultes sains, mais anormalement réexprimés par les cellules cancéreuses. Ainsi, bien qu'ils ne soient pas mutés, les paMAP constituent des cibles idéales et spécifiques au cancer. Nous avons identifié un ensemble de 48 paMAP dérivés de transcrits codants et non codants (48 %) impliqués dans le maintien de la pluripotence et exprimés de manière aberrante dans plusieurs types de cancer. Ainsi, bien qu’elles proviennent de différents types de cellules et de tissus, des tumeurs 4 distinctes convergent vers un programme transcriptionnel associé à la pluripotence. En effet, l’expression des paMAP dans les cancers est corrélée à l’hypométhylation récurrente de leurs gènes sources, la présence d’aberrations génomiques courantes et l’adoption par les tumeurs de stratégies d’évasion immunitaire communes. Enfin, comme plusieurs paMAP sont immunogènes, leur utilisation comme cibles dans des vaccins anticancéreux pourrait entrer en synergie avec les inhibiteurs disponibles des voies d'évasion immunitaire et améliorer le traitement de plusieurs cancers agressifs. Ensuite, nous avons évalué l’ensemble des TA ayant un potentiel thérapeutique dans deux types de tumeurs présentant une charge mutationnelle particulièrement élevée, le mélanome et le cancer du poumon non à petites cellules (NSCLC). Nous avons constaté que les TA mutés (mTSAs) représentent une minorité (1 %) des TA exploitables dans ces deux types de cancer. Cela peut s'expliquer par une faible expression d'ARN de la plupart des mutations non synonymes ainsi que par leur localisation en dehors des régions génomiques les plus efficaces pour la génération de MAP. En revanche, 99 % des TA dérivent de séquences génomiques non mutées spécifiques au cancer (aeTSA), surexprimées dans le cancer (TAA) ou spécifiques à la lignée cellulaire d'origine (LSA, exprimés par les mélanocytes ou par les cellules épithéliales pulmonaires, pour le mélanome et le NSCLC, respectivement). Tout comme les paMAP, environ 50 % des aeTSA identifiés dans le mélanome et le NSCLC proviennent de séquences non canoniques et sont régulés de manière épigénétique. Alors que les mTSA sont exclusivement spécifiques à chaque patient patient, les aeTSA sont partagés entre les échantillons tumoraux. De plus, leur absence dans les tissus normaux, leur abondance et leur capacité à activer les lymphocytes T CD8 en font des cibles idéales pour traiter les mélanomes et les NSCLC. En conclusion, cette thèse fournit un aperçu de la biogenèse de différents types de TA dans diverses cohortes de patients et ouvre la voie au développement d’immunothérapies ciblées et efficaces contre une grande variété de cancers. / Nearly all nucleated cells express MHC I (HLA I in humans)-associated peptides (MAPs) which are sampled from the cellular proteome and transported to the cell surface for inspection by CD8 T cells. As such, the collection of cell-surface MAPs, or the immunopeptidome, informs CD8 T cells on the inner cell state. Cancer immunosurveillance relies on the capacity of CD8 T cells to recognize abnormal MAPs on tumor cells and eliminate them while sparing healthy cells. Hence, the existence of cancer indicates that tumor-specific CD8 T cells are underpowered, dysfunctional or inhibited from exerting their function. Anti-cancer vaccines can boost tumor killing by stimulating the recognition of abnormal MAPs. The development of effective anti-cancer vaccines is limited by the identification of actionable MAPs, or tumor antigens (TAs), expressed exclusively on tumor cells. The TA search space has been largely limited to MAPs derived from non-synonymous mutations in canonical protein-coding exons which represent a mere 2% of the human genome. That MAPs can derive from the non-canonical translation of potentially all genomic regions has only recently been fully appreciated. Herein, we used proteogenomics to discover actionable TAs derived from canonical and non-canonical translation products shared within or across different types of human cancer. First, we used induced pluripotent stem cells (iPSCs) to identify pluripotency-associated MAPs (paMAPs) shared by cancer cells. Pluripotency antigens are restricted to embryonic tissues and absent from healthy adult tissues but abnormally re-expressed by cancer cells, which makes them ideal tumor-specific targets despite being unmutated. We identified a set of 46 paMAPs derived from coding and allegedly non-coding (48%) transcripts involved in pluripotency maintenance and aberrantly expressed in multiple cancer types. Thus, despite originating from different cell types and tissues, distinct tumor types converged towards a pluripotency-associated transcriptional program. Indeed, the expression of paMAPs across cancers correlated with recurrent source gene hypomethylation, genomic aberrations, and immune evasion properties. Several paMAPs were immunogenic, thus their targeting could synergize with available inhibitors of immune evasion pathways to improve the outcome of multiple aggressive cancers. 7 Next, we evaluated the actionable TA landscape of two tumor types with particularly high mutational load, melanoma and non-small cell lung cancer (NSCLC). We found that mutated TAs (mTSAs) represent a minority (1%) of actionable TAs in both cancer types, which can be explained by a low RNA expression of most non-synonymous mutations and their localization outside genomic regions proficient for MAP generation. By contrast, 99% of TAs derived from unmutated genomic sequences specific to cancer (aeTSAs), overexpressed in cancer (TAAs), or specific to the cell lineage of origin (LSAs, expressed by melanocytes or by lung epithelial cells, for melanoma and NSCLC LSAs, respectively). As for paMAPs, around 50% of aeTSAs in melanoma and NSCLC were non-canonical and were epigenetically regulated. Whereas mTSAs were exclusively patient-specific, aeTSAs were shared among tumor samples and exhibited all characteristics of targetable TAs, including tumor-specificity, high abundance, and immunogenicity. Altogether, this thesis provides insights into the biogenesis of different TA types in various patient cohorts and paves the way for the development of effective TA-based immunotherapies against a large variety of cancers.
37

Aberrations in Cytokine Signaling in Leukemia: Variations in Phosphorylation and O-GlcNAcylation

Tomic, Jelena 31 August 2012 (has links)
Tumor-induced immunosuppression can occur by multiple mechanisms, each posing a significant obstacle to immunotherapy. Evidence presented in this dissertation suggests that aberrant cytokine signaling, as a result of altered metabolism of Chronic Lymphocytic Leukemia (CLL) cells, confers a selective advantage for tumor survival and growth. Cells from CLL patients with aggressive disease (as indicated by high-risk cytogenetics) were found to exhibit prolongation in Interferon (IFN)-induced STAT3 phosphorylation, and increased levels of reactive oxygen species (ROS) in these cells reflected these signaling processes. Changes in the relative balance of phospho-STAT3 and phospho-STAT1 levels, in response to combinations of IL-2 + Toll-like receptor (TLR)-7 agonist + phorbol esters, as well as IFN, were associated with the immunosuppressive and immunogenic states of CLL cells. In addition, immunosuppressive leukemic cells were found to express high levels of proteins with O-linked N-acetylglucosamine (O-GlcNAc) modifications, due to increased metabolic activity through the Hexosamine Biosynthetic Pathway (HBP), which caused impaired intracellular signaling responses and affected disease progression. A conclusion of the studies presented here is that the intrinsic immunosuppressive properties of leukemic cells may be overcome by agents such as Resveratrol that target metabolic pathways of these cells.
38

Aberrations in Cytokine Signaling in Leukemia: Variations in Phosphorylation and O-GlcNAcylation

Tomic, Jelena 31 August 2012 (has links)
Tumor-induced immunosuppression can occur by multiple mechanisms, each posing a significant obstacle to immunotherapy. Evidence presented in this dissertation suggests that aberrant cytokine signaling, as a result of altered metabolism of Chronic Lymphocytic Leukemia (CLL) cells, confers a selective advantage for tumor survival and growth. Cells from CLL patients with aggressive disease (as indicated by high-risk cytogenetics) were found to exhibit prolongation in Interferon (IFN)-induced STAT3 phosphorylation, and increased levels of reactive oxygen species (ROS) in these cells reflected these signaling processes. Changes in the relative balance of phospho-STAT3 and phospho-STAT1 levels, in response to combinations of IL-2 + Toll-like receptor (TLR)-7 agonist + phorbol esters, as well as IFN, were associated with the immunosuppressive and immunogenic states of CLL cells. In addition, immunosuppressive leukemic cells were found to express high levels of proteins with O-linked N-acetylglucosamine (O-GlcNAc) modifications, due to increased metabolic activity through the Hexosamine Biosynthetic Pathway (HBP), which caused impaired intracellular signaling responses and affected disease progression. A conclusion of the studies presented here is that the intrinsic immunosuppressive properties of leukemic cells may be overcome by agents such as Resveratrol that target metabolic pathways of these cells.

Page generated in 0.0843 seconds