• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 24
  • 7
  • 4
  • 2
  • 2
  • 2
  • 2
  • 1
  • Tagged with
  • 50
  • 50
  • 39
  • 29
  • 23
  • 22
  • 18
  • 17
  • 12
  • 10
  • 10
  • 8
  • 8
  • 8
  • 8
  • 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.
21

Functional genetic screening and therapeutic targeting of recurrent glioblastoma

Chokshi, Chirayu R January 2022 (has links)
Glioblastoma (GBM) remains the most aggressive and prevalent malignant primary brain tumor in adults. Unchanged since 2005, standard of care (SoC) consists of surgical resection, followed by radiation therapy (RT) with concurrent and adjuvant chemotherapy with temozolomide (TMZ). Despite these therapeutic efforts, patients succumb to recurrent disease with a median overall survival of 14.6 months and a five-year survival rate of 5.5-6.8%. Therapeutic failure is largely explained by ITH and the presence of treatment-resistant GBM stem-like cells (GSCs). Given the lack of understanding of recurrent GBM and absence of second line therapies patients, I hypothesize that genome-scale functional genetic interrogation will unravel recurrent GBM-specific tumor biology and inform development of novel therapeutics. First, I compared primary and recurrent GBM at the genetic, transcriptomic, proteomic and functional genetic levels. These analyses map a multilayered genetic response to drive tumor recurrence, identifying protein tyrosine phosphatase 4A2 (PTP4A2) as a novel modulator of self-renewal, proliferation and tumorigenicity at GBM recurrence. Mechanistically, genetic perturbation and a small molecule inhibitor of PTP4A2 repress axon guidance activity through a dephosphorylation axis with roundabout guidance receptor 1 (ROBO1) and exploit a genetic dependency on ROBO signaling. Importantly, engineered anti-ROBO1 single-domain antibodies also mimic the effects of PTP4A2 inhibition. Given the genetic dependency on ROBO signaling and enrichment of ROBO1 expression in GBM tissues, I undertook a campaign to evaluate ROBO1 as a therapeutic target in recurrent GBM and develop anti-ROBO1 chimeric antigen receptor T (CAR-T) cells using camelid single-domain antibodies targeting human ROBO1. I optimized the design of anti-ROBO1 CAR-T cells and tested the anti-tumor activity of these modalities in in vitro using patient-derived recurrent GBM lines and orthotopic patient-derived xenograft models. I present data to expand the repertoire of GBM-enriched antigens suitable for effective CAR-T cell therapy. Given that resistance to SoC and disease relapse are inevitable for GBM patients, pre-clinical and clinical advancement of immunotherapeutic modalities, combined with recent insights into the tumor immune microenvironment, are poised to improve clinical outcomes for this patient population. / Thesis / Doctor of Philosophy (PhD) / Glioblastoma remains the most lethal and prevalent primary brain tumor in adults. Standard of care for patients remains unchanged since 2005, consisting of surgery to remove visible tumor at diagnosis (primary tumor), followed by radiation therapy and chemotherapy to treat remaining tumor cells. Despite these therapeutic efforts, tumor relapse (recurrent tumor) is inevitable with no standardized second-line therapy. Patients succumb to recurrent disease with a median overall survival of 14.6 months and only 5.5-6.8% of patients survive five years post diagnosis. Therapy failure and tumor relapse are explained by immense diversity among tumor cells at the DNA and protein levels, giving rise to a subset of tumor cells with abilities to resist therapy and seed the recurrent tumor. Previous studies have presented evolution of tumor cells through therapy, with recurrent tumor cells harboring novel changes at the DNA and protein levels. However, the impact of these changes on tumor cell function has not been evaluated. In this thesis, we developed and applied a genetic screening technique to determine the functional role of thousands of genes in primary and recurrent tumor cells from the same patient. This analysis revealed numerous genes that exhibit differential effects on survival of primary and recurrent tumor cells, including genes that drive recurrent tumor cell growth but are dispensable in primary tumor cells. Functional remodeling of these genes and pathways revealed a new functional role of multiple proteins belonging to a process called axonal guidance in recurrent tumor cells. To evaluate the therapeutic potential of these findings, we deeply interrogated the mechanism by which axonal guidance drives recurrent tumor cells and targeted crucial molecular players using chemical and immunological therapies. Using models that predict clinical effectiveness, we engineered and tested a novel therapy that redirects immune cells to target recurrent tumor cells driven by dysfunctional axonal guidance activity. The goal of this thesis was to discover the functional differences between primary and recurrent tumor cells, thereby leveraging this information to engineer candidate therapies for treatment of recurrent glioblastoma.
22

The Multiple Faces of Genetically-Modified T Cells : Potential Applications in Therapy

Hillerdal, Victoria January 2014 (has links)
In this PhD thesis the potential of T-cells as therapy for disease are explored. The applications of genetically modified T-cells for treatment of cancer and autoimmune disease; the functionality and optimal activation of T-cells are discussed. Successful treatment of cancer with T-cell receptor (TCR)-modified T-cells was first reported in 2006, and is based on recognition of a specific peptide by the TCR in the context of the MHC molecule. As antigen presentation in tumors is often defective and to avoid MHC-restriction, chimeric antigen receptors (CAR) molecules containing an antibody part for recognition of cell surface antigens and TCR and co-receptor signaling domains have been developed. Activated T-cells mount an efficient immune response resulting in the killing of the cancer cell and initiating T-cell proliferation. The rationale for using genetically modified T-cells instead of isolating tumor infiltrating lymphocytes from the tumor and expanding them (TIL therapy) is that it is often very difficult to obtain viable lymphocytes that are able to expand enough in order to use them for therapy. This thesis explores the possibility of using prostate-specific antigens to target T-cells towards prostate cancer. The prostate has many unique tissue antigens but most patients with metastatic prostate cancer have undergone prostatectomy and consequently have “prostate antigen” expression only in cancer cells. We targeted the prostate antigens TARP and PSCA with a HLA-A2 restricted TCR and a CAR respectively. In both cases the tumor-specific T-cells were able to generate potent proliferative and cytotoxic responses in vitro. The PSCA CAR-modified T-cells delayed subcutaneous tumor growth in vivo. It is evident from our in vivo experiments that the PSCA CAR T-cells were unable to completely cure the mice. Therefore, we aimed to improve the quality of the transferred T-cells and their resistance to the immunosuppressive tumor microenvironment. Stimulation with allogeneic lymphocyte-licensed DCs improved the resistance to oxidative stress and antitumor activity of the T-cells. We further investigated the potential of genetically modified regulatory T-cells (Tregs) to suppress effector cells in an antigen-specific manner. Using a strong TCR we hypothesize that the phenotype of the TCR-transduced Tregs may be affected by antigen activation of those cells. We found that the engineered Tregs produced cytokines consistent with Th1, Th2 and Treg phenotypes.
23

Mechanisms of lck-dependent proliferation during thymocyte development /

Tasch, Michael A. January 2000 (has links)
Thesis (Ph. D.)--University of Washington, 2000. / Vita. Includes bibliographical references (leaves 139-193).
24

Enhancing adoptive immunotherapy : redirecting immune subsets and metabolic pathways / Optimisation des immunothérapies : manipulation de sous populations immunitaires et exploitation du métabolisme

Yong, Carmen 15 September 2017 (has links)
Le transfert adoptif de cellules T exprimant un récepteur chimérique reconnaissant un antigène (CAR), est un traitement qui génère des réponses impressionnantes dans les cancers hématologiques mais est beaucoup moins efficace pour le traitement de tumeurs solides. Les tumeurs solides modulent leur microenvironnement induisant des formes multiples d’immunosuppression qui inhibent l’efficacité des fonctions effectrices des cellules T ayant infiltrées la tumeur. Au cours de ma thèse, j’ai évalué le potentiel de deux stratégies pour améliorer les réponses anti-tumorales des cellules T CAR. La première se focalise sur l’étude du rôle potentiel des cellules immunes non T, exprimant un CAR sur la stimulation des fonctions et de la persistance de cellules T CAR+ dans le microenvironnement tumoral. Afin d’étudier la fonction des cellules CAR non T, nous avons généré un modèle de souris transgénique (vav-CAR) dans lequel les cellules immunes expriment un CAR reconnaissant l’antigène tumoral Her2 (ErbB2). Comme attendu, les cellules T CAR+ possèdent des fonctions anti-tumorales, mais nous avons aussi mis en évidence que les macrophages et les cellules NK exprimant le CAR montraient une réponse cytokinique, cytotoxique et phagocytiques spécifiques de l’antigène. De plus, en utilisant le modèle vav-CAR, nous avons démontré le potentiel des cellules immunes CAR+ dans le rejet des tumeurs et cela indépendamment des cellules T CD8+. Les cellules T CD4+ sont essentielles puisque leur élimination réduit considérablement les réponses anti-tumorales dans notre modèle vav-CAR. Il a été démontré que certaines sous-populations de cellules T auxiliaires participent aux réponses anti-tumorales avec les cellules Th1 et Th17 démontrant une efficacité plus robuste que les autres sous-populations. Notre deuxième stratégie s’est focalisée sur l’étude de l’impact du métabolisme au cours de la polarisation des cellules T CD4+ et plus particulièrement lors de la différenciation des cellules T CAR+ en cellules Th1. En effet, l’activation et différenciation des cellules T sont fortement associées à une augmentation des besoins métaboliques. Dans le microenvironnement tumoral, en raison de la forte demande en ressources de la tumeur, la déprivation en nutriments ainsi générée peut limiter l’accès aux nutriments d’autres types cellulaires et ainsi altérer le devenir et les fonctions des cellules immunes greffés infiltrant la tumeur. En conséquence, modifier les cellules immunes CAR+ afin qu’elles puissent résister à la compétition métabolique du microenvironnement tumoral pourrait leur permettre de conserver leurs fonctions effectrices. En étudiant l’impact de la déprivation en nutriments sur la différenciation des cellules T, nous avons trouvé que des concentrations limitantes en glutamine, l’acide aminé le plus abondant du plasma, inhibaient le potentiel des cellules T à se différencier vers la voie Th1 associée à la production d’IFNγ. Au contraire, cette condition favorisait la conversion de cellules T CD4 naïves en cellules régulatrices Foxp3+ ayant des fonctions suppressives (Tregs). De plus, nous avons montré que la présence d’un seul métabolite dérivé de la glutamine, l’α-ketoglutarate (αKG), suffisait à augmenter les fonctions effectrices anti-tumorales de plusieurs sous-types de cellules T auxiliaires CAR+, augmentant la production d’IFNγ et diminuant l’expression de FOXP3. Ainsi, durant ma thèse, j’ai développé un modèle murin vav-CAR, générant un outil permettant d’étudier et manipuler les fonctions de multiples populations de cellules immunitaires exprimant un CAR. Ce modèle permettra de promouvoir l’utilisation de cellules immunes optimisées exprimant un CAR dans le cadre d’immunothérapies dirigées contre des tumeurs solides. De plus, en utilisant ce modèle, nous avons identifié un métabolite de la glutamine, qui orchestre les réponses immunitaires au moyen d’une reprogrammation métabolique des cellules T CD4. / The adoptive transfer of T cells expressing a chimeric antigen receptor (CAR) as a treatment for cancer has achieved impressive responses in haematological malignancies, but has been less successful in the treatment of solid tumors. The tumor microenvironment of solid tumors presents multiple forms of immunosuppression, inhibiting the efficient effector function of infiltrating anti-tumor T cells. During my PhD, we assessed the potential of two strategies to enhance the anti-tumor function of CAR T cells. The first focuses on the potential of other CAR-expressing immune subsets to stimulate CAR T cell function and persistence in the tumor microenvironment. To elucidate the function of CAR-expressing non-T lymphocytes, we generated a transgenic mouse model (vav-CAR) in which immune cells express a CAR against the Her2 (ErbB2) tumor antigen. As expected, CAR T cells harboured anti-tumor function but we also found that CAR-modified macrophages and natural killer cells (NKs) exhibited significant antigen specific cytokine secretion, cytotoxicity and phagocytosis. Moreover, using the vav-CAR model, we demonstrated the potential of CAR immune cells to mediate tumor rejection independently of CD8+ T cells. CD4+ T cells were critical for this response as their deletion severely abrogated the anti-tumor responses in our vav-CAR model. Distinct T helper subsets have been shown to participate to anti-tumor responses, with Th1 and Th17 cells demonstrating a more robust efficacy as compared to other T helper subsets. Our second strategy was focused on the impact of metabolism in the polarisation of CD4+ T cells, in particular the differentiation of CAR T cells to Th1 lineage. T cell activation and polarisation is highly associated with increased metabolic needs. Given that nutrient deprivation in the tumor microenvironment, due to a high demand of the tumor for resources, can limit the nutrients available for other cell types, the fate and function of adoptively transferred immune cells may be altered upon entering the tumor. Therefore, modifying CAR immune cells to resist metabolic suppression in the tumor microenvironment may help retain their effector functions. Upon assessing the effects of nutrient deprivation on T cell differentiation, we found that limiting concentrations of glutamine, the most abundant amino acid in the plasma, inhibited the potential of T cells to undergo Th1 differentiation with associated IFNγ secretion. Rather, this condition resulted in the conversion of naïve CD4+ T cells into suppressive Foxp3+ regulatory T cells (Tregs). Furthermore, we determined that a single glutamine-derived metabolite, α-ketoglutarate (αKG), enhanced the anti-tumor effector functions of multiple CAR T helper subsets, increasing the production of IFNγ and reducing FOXP3 expression.Thus, during my PhD, I generated a vav-CAR model, providing a platform in which the function of multiple CAR-bearing immune subsets can be studied and manipulated. This model will promote the utilisation of optimized CAR-bearing immune cells in adoptive immunotherapy for solid tumors. Furthermore, using the CAR model, we have identified a glutamine metabolite that orchestrates immune responses through the metabolic reprogramming of CD4 T cells.
25

Chimeric antigen receptor (CAR)-modified T cells targeting FLT3 in acute myeloid leukemia (AML) / Chimäre Antigen Rezeptor (CAR)-modifizierte T-Zellen gegen FLT3 bei Akuter Myeloischer Leukämie (AML)

Jetani, Hardikkumar January 2021 (has links) (PDF)
Adoptive immunotherapy using chimeric antigen receptor (CAR)-modified T cells targeting CD19 has shown remarkable therapeutic efficacy against B cell leukemia and lymphoma, and provided proof of concept for therapeutic potential in other hematologic malignancies. Acute myeloid leukemia (AML) is an entity with an unmet medical need for effective and curative treatments. Therefore, there is a strong desire for development of potentially curative CAR-T cell immunotherapy for AML treatment. FMS-like tyrosine kinase 3 (FLT3) is a homodimeric transmembrane protein expressed uniformly by AML blasts. FLT3 plays a vital role in the survival of AML blasts and is a key driver of leukemia-genesis in AML cases with internal tandem duplication (FLT3ITD) and tyrosine kinase domain (TKD) mutations. These attributes suggest that FLT3 could be an excellent target for CAR-T cell immunotherapy. Here, we engineered human CD4+ and CD8+ T cells to express FLT3-specific CARs and demonstrate that they confer potent reactivity against AML cell lines and primary AML blasts that express either wild-type FLT3 or FLT3-ITD. Further, we show that FLT3 CAR-T cells exert potent antileukemia activity in xenograft models of AML and induce complete remissions. We also demonstrate that FLT3-expression on FLT3-ITD+ AML cells can be augmented by FLT3 inhibitors, which lead to increased recognition by CARs and improved efficacy of FLT3 CAR-T cells. We confirmed this principle with three different FLT3 inhibitors which are at distinct stages of clinical development i.e. Phase II/III clinical trial (crenolanib, quizartinib) and clinically approved (midostaurin). Further, we observed the strongest anti-leukemia activity of FLT3 CAR-T cells in combination with crenolanib in vivo. FLT3 is known to be expressed by normal hematopoietic stem and progenitor cells. We evaluated FLT3-expression on normal hematopoietic stem cells (HSCs) using flow cytometry and confirmed lower level of FLT3-expression on HSCs and progenitors compared to AML cells. As anticipated, we found that FLT3 CAR-T cells recognize normal HSCs in vitro and in vivo, and compromise normal hematopoiesis, suggesting that adoptive therapy with FLT3 CAR-T cells will require successive CAR-T cell depletion and allogeneic HSC transplantation (HSCT) to reconstitute the hematopoietic system. Moreover, an FLT3 inhibitor treatment does not increase FLT3-expression on HSCs. Accordingly, we demonstrate that the depletion of FLT3 CAR-T cells is possible with inducible Caspase 9 (iCasp9) safety switch. Collectively, our data establish FLT3 as a novel CAR target in AML with particular relevance in high-risk FLT3-ITD+ AML. Our data demonstrate that FLT3 CAR-T cells act synergistically with FLT3 inhibitors in FLT3-ITD+ AML. i.e. FLT3 inhibitors-induced upregulation of FLT3 in FLT3-ITD+ AML cells enhances their recognition and elimination by FLT3 CAR-T cells. Due to recognition of normal HSCs, the clinical use of FLT3 CART cells is likely restricted to a defined therapeutic window and must be followed by CART cell depletion and allogeneic HSCT for hematopoietic reconstitution. The data provide rational to use FLT3 CAR-T cells in combination with FLT3 inhibitors to augment the anti-leukemia efficacy of FLT3 CAR-T cells in high-risk FLT3-ITD+ AML patients, and to mitigate the risk of relapse with FLT3-negative AML variants, which could otherwise develop under therapeutic pressure. The data provide proof of concept for synergistic use of CAR-T cell immunotherapy and small molecule targeted therapy and encourage the clinical evaluation of this combination treatment in high-risk patients with FLT3-ITD+ AML. / Adoptive Immuntherapie, die Chimäre- Antigenrezeptor (CAR) –modifizierte, gegen CD19 gerichtet T-Zellen verwendet, hat eine bemerkenswerte therapeutische Wirksamkeit gegen B-Zell-Leukämien und -Lymphome und großes therapeutisches Potenzial für die Behandlung anderer hämatologischer Erkrankungen gezeigt. Die Akute Myeloische Leukämie (AML) ist hierbei eine Entität, für die es bisher an wirksamen und kurativen Therapien fehlt und für die die Entwicklung einer potentiell kurativen CAR-T-Zellimmuntherapie von großer Bedeutung ist. FMS-like tyrosine kinase 3 (FLT3) ist ein homodimeres Transmembranprotein, das von AML-Blasten uniform exprimiert wird. FLT3 spielt eine wichtige Rolle beim Überleben von AML-Blasten und ist ein Schlüsselfaktor in der Leukämie-Genese bei AML-Fällen mit interner Tandem-Duplikation (FLT3-ITD) und Tyrosinkinase-Domänen (TKD)-Mutationen. Diese Eigenschaften legen die Vermutung nahe, dass FLT3 ein ausgezeichnetes Target für die CAR-T-Zell-Immuntherapie darstellen könnte. Daher setzten wir dort an und modifizierten humane CD4+ und CD8+ T-Zellen, um FLT3-spezifische CARs zu exprimieren, und konnten nachweisen, dass diese eine starke Reaktivität gegen AML-Zelllinien und primäre AML-Blasten besitzen, die entweder den FLT3-Wildtyp oder FLT3-ITD exprimieren. Weiterhin konnten wir zeigen, dass FLT3 CAR-T-Zellen in AML-Xenograft-Modellen eine starke anti-Leukämie-Aktivität besitzen und vollständige Remissionen hervorrufen können. Zudem gelang der Nachweis, dass die FLT3-Expression auf FLT3-ITD+ AML-Zellen durch FLT3-Inhibitoren verstärkt werden kann, was zu einer erhöhten Erkennung durch die CARs und einer verbesserten Wirksamkeit von FLT3-CAR-T-Zellen führt. Wir konnten dieses Prinzip mit drei verschiedenen FLT3-Inhibitoren belegen, die sich in unterschiedlichen Stadien der klinischen Entwicklung befinden, d. h. aus einer Klinischen Phase II / III-Studie (Crenolanib, Quizartinib) und einem klinisch zugelassenen Inhibitor (Midostaurin). Darüber hinaus beobachteten wir die stärkste anti-Leukämie-Aktivität von FLT3 CAR-T-Zellen in einer Kombination mit Crenolanib in vivo. Es ist bekannt, dass FLT3 von normalen hämatopoetischen Stamm- und Vorläuferzellen exprimiert wird. Wir untersuchten die FLT3-Expression in normalen hämatopoetischen Stammzellen (HSCs) mittels Durchflusszytometrie und bestätigten im Vergleich zu AML-Zellen eine niedrigere FLT3-Expression auf HSCs und Vorläuferzellen. Wie erwartet, zeigte sich, dass FLT3 CAR-T-Zellen normale HSCs in vitro und in vivo erkennen und die normale Hämatopoese beeinträchtigen, was darauf hindeutet, dass eine adoptive Therapie mit FLT3 CAR-T-Zellen eine sukzessive CAR-T-Zell-Depletion und allogene HSC-Transplantation erfordert, um das hämatopoetische System wiederaufzubauen. Darüber hinaus erhöht die Behandlung mit einem FLT3-Inhibitor nicht die FLT3-Expression auf den HSCs. Dementsprechend konnten wir aufzeigen, dass die Depletion von FLT3 CAR-T Zellen mit einer induzierbaren Caspase 9 (iCasp9) als „Sicherheitsschalter“ möglich ist. Zusammenfassend etablieren unsere Daten FLT3 als ein neuartiges CAR-Target in der Behandlung von AML mit besonderer Relevanz für die Hochrisiko-FLT3-ITD+ AML. Unsere Daten zeigen, dass FLT3 CAR-T-Zellen synergistisch mit FLT3-Inhibitoren in FLT3-ITD+ AML wirken, d.h. eine FLT3-Inhibitoren-induzierte Hochregulation von FLT3 in FLT3-ITD+ AML-Zellen bewirkt und dies die Erkennung und Eliminierung durch FLT3-CAR-T-Zellen verstärkt. Durch ihre Eigenschaft der Erkennung von normalen HSCs ist die klinische Verwendung von FLT3 CAR-T-Zellen wahrscheinlich auf ein definiertes therapeutisches Fenster beschränkt und muss durch eine anschließende CAR-T-Zell-Depletion und eine allogene HSCT zur Rekonstitution des hämatopoetischen Systems ergänzt werden. In Anbetracht der Daten scheint es sinnvoll, FLT3-CAR-T-Zellen in Kombination mit FLT3-Inhibitoren zu verwenden, um die anti-leukämische Wirksamkeit von FLT3-CAR-T-Zellen bei Hochrisiko-FLT3-ITD+ AML-Patienten zu erhöhen und das Risiko eines Rückfalls mit FLT3-negativen AML-Varianten zu verringern, die sich sonst therapiebedingt entwickeln könnten. Die Daten stellen ein Proof-of-Concept für den synergistischen Einsatz von CAR-T-Zell-Immuntherapie und niedermolekularen Inhibitoren dar, der eine klinische Evaluation dieser Kombinationsbehandlung bei Hochrisikopatienten mit FLT3-ITD+ AML erstrebenswert macht.
26

Cell-to-cell transmission and intrinsic mechanisms that influence human immunodeficiency virus infection

Pedro, Kyle D. 18 February 2021 (has links)
Early in the course of human immunodeficiency virus (HIV) infection a population of latently infected cells is established which persists despite long-term anti-retroviral treatment. This latent reservoir of HIV-infected cells, which reflects mechanisms of transcriptional repression, is the major barrier to cure. Efforts to target the latent reservoir have been inefficient, indicating a need for a more complete understanding of how HIV transcription is regulated. The molecular networks involved in the regulation of HIV transcription remain incompletely defined. I hypothesized that utilization of a high throughput enhanced yeast one-hybrid assay would reveal novel host transcription factor-long terminal repeat (LTR) interactions and transcriptional networks that regulate HIV. The screen identified 42 human transcription factors and 85 total protein-DNA interactions with HIV LTRs. I investigated a subset of these factors for transcriptional activity in cell-based models of infection. Krüppel-like factors 2 and 3 (KLF2 and KLF3) are repressors of HIV-1 and HIV-2 transcription whereas PLAG1-like zinc finger 1 (PLAGL1) is an activator of HIV-2 transcription. These factors regulate HIV expression through direct protein-DNA interactions and correlate with epigenetic modifications of the HIV LTR. Multiple signals converging from the cellular environment and cell-cell interactions converge at the HIV LTR to determine HIV replication and transcription. Previous work in our lab has shown that strong signaling through the T cell receptor (TCR) was required to support HIV expression and the establishment of an inducible latent infection, whereas weak TCR signaling was insufficient for these outcomes. I hypothesized that dendritic cells-CD4+ T cell interactions provide signals that compensate for weak TCR signaling, supporting HIV-1 expression and generation of inducible latent infection. I used CD4+ T cells that express chimeric antigen receptors in a dendritic cell coculture model to deliver differential signals to CD4+ T cells during cell-to-cell transmission of HIV. I found that signals from dendritic cells compensate for weak TCR signaling, facilitating cell activation, HIV expression and establishment of an inducible infection.
27

Cancer Signals-Triggered T Cell Immunotherapy for Solid Tumors

Nguyen, Huong Thi Xuan January 2022 (has links)
No description available.
28

Chimerické antigenní receptory a jejich využití pro léčbu hematologických malignit / Chimeric antigen receptors in the treatment of hematological malignacies

Fellnerová, Adéla January 2016 (has links)
Chimeric antigen receptors (CARs) are artificial molecules composed of an antibody derived antigen recognition domain which is fused with the signal transduction domain derived from the physiological TCR. CAR technology used to transduce patients T-cells and endow them with the specificity to a certain surface antigen, has been a major breakthrough in cancer immunotherapy in the last decade. This strategy has been most successful for treating hematologic malignancies. Various CAR approaches and applications are currently tested mainly in the United States where many clinical trials have been launched. In contrast, in the Czech Republic, there are only a few teams focused on this topic with no clinical trials going on. During my work on this diploma thesis and in close collaboration with MUDr. Pavel Otáhal, PhD., who is working on implementation of CAR technology into the Czech clinics for the treatment of B-cell malignancies, individual functional CARs were prepared and tested. CAR expressing Jurkat T-cell lines were generated using a lentiviral vector transduction system. CAR functionality was determined by two different assays. We have shown that individual CARs are able to recognize the B-cell lineage specific antigens CD19 and CD20 and significantly up-regulate the activation molecule CD69 upon...
29

Estabelecimento de uma plataforma para produção de vetores lentivirais para a modificação de linfócitos T com CAR anti-CD19 / Establishment of a platform for the production of lentiviral vectors for the modification of anti-CD19 CAR-T cells

Moço, Pablo Diego 23 July 2018 (has links)
A imunoterapia utilizando linfócitos T modificados com receptor quimérico de antígenos (CAR) tem se mostrado eficaz no tratamento de leucemia e linfomas resistentes à quimioterapia. A proteína CD19 é considerada um alvo ideal porque é expressa na maioria dos tumores de linfócitos B e linfócitos B normais, mas não em outras células. Estudos clínicos recentes mostraram excelentes respostas de linfócitos T-CAR em uma variedade de tumores de células B. Os vetores lentivirais são o método mais comumente utilizado para modificação genética em ensaios clínicos. Este estudo teve como objetivo desenvolver uma plataforma eficiente para a produção de lentivírus e testar a funcionalidade desses vetores para que possam ser usados para modificar geneticamente linfócitos T. A transfecção transiente de céulas HEK293T com plasmídeos na proporção de 3:1:1:1 (transgene:gag-pol:VSV-G:rev) utilizando lipossomos catiônicos e 5 mM de butirato de sódio resultou nos títulos virais mais elevados. Isso representa um aumento de 17 vezes no título viral da transfecção com polietilenoimina (PEI). Três métodos para concentracao lentiviral foram utilzados nesse trabalho, ultracentrifugação, filtração tangencial e ultrafiltração. A ultrafiltração sobre membrana com corte de peso molecular (MWCO) de 100 kDa resultou na maior taxa de recuperação de partículas virais viáveis, aproximadamente 82%. As partículas virais produzidas por este processo demonstraram ser funcionais para a transdução de linfócitos T. Além disso, o receptor quimérico (CAR) se mostrou específico contra o antígeno CD19 de células B, resultando na ativação dos linfócitos T-CAR e gerando citotoxicidade contra células CD19+ in vitro. Houve uma redução de aproximadamente 87% das células alvo, quando analisado por citometria de fluxo e uma citotoxicidade média de 50% foi observada por ensaios colorimétricos. / Immunotherapy using T cells modified with chimeric antigen receptor (CAR) has been proven effective in the treatment of leukemia and lymphomas resistant to chemotherapy. CD19 protein has been shown to be an ideal target because it is expressed on most B-cell tumors and normal B cells, but not in other cells. Recent clinical studies have shown excellent responses of CAR T-cells in a variety of B-cell tumors. Lentiviral vectors are the most commonly used method for genetic modification in clinical trials. This study aimed to develop an efficient platform for lentiviral production and to test the functionality of those vectors so that they can be used in to genetically modify T cells. Transient transfection of HEK293T cells with plasmids in a 3:1:1:1 ratio (transgene:gag-pol:VSV-G:rev) using cationic liposomes and 5 mM sodium butyrate resulted in the highest viral titers. That represents a 17-fold increase in viral titer from polyethylenimine (PEI) transfection. Three methods for lentiviral concentration were used in this work, ultracentrifugation, tangential filtration and ultrafiltration. Membrane ultrafiltration with 100 kDa molecular weight cutoff (MWCO) resulted in the highest recovery rate of viable viral particles, approximately 82%. The viral particles produced by this process have been shown to be functional for the transduction of T cells. In addition, the chimeric receptor (CAR) was shown to be specific against the B cell antigen CD19, resulting in the activation of CAR-T cells and generating cytotoxicity against CD19+ cells in vitro. There was a reduction of approximately 87% of the target cells when analyzed by flow cytometry and an average cytotoxicity of 50% was observed by colorimetric assays.
30

Anomalies des programmes de réponse lymphocytaire après stimulation du récepteur à l’antigène dans la leucémie lymphoïde chronique / Abnormalities of lymphocyte response programs after antigen receptor stimulation in chronic lymphocytic leukemia

Schleiss, Cédric 21 December 2018 (has links)
Une cellule reçoit en permanence des signaux de son environnement. Cette stimulation induit une cascade de signalisation activant un programme génique et protéomique dynamique aboutissant à une réponse cellulaire adaptée. Dans la leucémie lymphoïde chronique (LLC), la stimulation du récepteur à l’antigène induit un programme et une réponse anormale à l’origine de la prolifération leucémique. Notre objectif est de caractériser ce programme cellulaire pathologique. Pour cela, nous avons mis en place un modèle de stimulation afin de reproduire ex vivo cette stimulation du récepteur à l’antigène de cellules primaires issues de patients porteurs de LLC et d’activer ce programme cellulaire. Nous avons alors analysé la dynamique transcriptionnelle et protéomique activée dans ces cellules afin de caractériser les anomalies de ce programme. Cette étude nous a permis de mettre en évidence la spécificité de ce programme prolifératif et de caractériser les gènes clés de ce programme tumoral. Ces gènes constituent de potentielles cibles thérapeutiques innovantes. / A cell constantly receives signals from its environment. This stimulation induces a signalling cascade activating a dynamic genic and proteomic program, leading to an adapted cellular response. In chronic lymphocytic leukemia (CLL), an antigen receptor stimulation induces a program and an abnormal response behind leukemic proliferation. Our aim was to characterize the pathological cell program. To achieve this, we have implemented a stimulation model to reproduce ex vivo antigen receptor stimulation of primary cells from CLL patients and activate this cellular program. We then analyzed the transcriptional and proteomic dynamics activated in these cells in order to characterize the abnormalities of this program. This study allows us to highlight the specificity of this proliferative program and to identify key genes of tumor program. These genes constitute potential new therapeutic targets.

Page generated in 0.0979 seconds