• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 6
  • 2
  • 1
  • 1
  • Tagged with
  • 11
  • 11
  • 11
  • 9
  • 5
  • 4
  • 4
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 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.
1

Adoptive T Cell Therapy for Treatment of Metastatic Melanoma

Sadeghi, Arian January 2011 (has links)
Malignant melanoma is a common type of solid tumor that causes high cancer-related mortality in young adults of Northern Europe. The incidence of melanoma increases rapidly which renders us a special responsibility to investigate this disease in depth. One recent promising approach to treat malignant melanoma is adoptive cell therapy with tumor-directed autologous T cells. This thesis aims to improve this therapy in four different studies. We first sought to establish a protocol for the assessment of melanoma-specific T-cell cultures in order to screen for optimal specificity and reactivity in a robust, reliable and simple manner. The conclusion was that reactive cells could be found in a majority of patients and could be screened for specificity by stimulation with melanoma cell lines. In the next study, 28 melanoma patients with advanced disease were treated with autologous tumor-infiltrating T cells. Objective responses (18%) including one sustained complete response were observed. This is the first study in cancer patients with autologous T cell transfer combined with low-dose s.c. IL-2 as supportive cytokine. In the following two studies we wanted to improve management and culture conditions of the T cells. When investigating methods for improved handling and preservation of large numbers of T cells, we observed that freeze-thawing of T cells could impair the metabolic activity of the T cells. Another conclusion was that rapid expansion of T cells could lead to loss of antigenic specificity and apoptosis. These adverse effects could be prevented with short time recovery. In order to improve expansion methods, mass expansion of T cells in an automated bioreactor was evaluated. We concluded that the bioreactor is suitable for this task and allows for higher cell densities and absolute cell numbers compared to traditional culturing conditions without influencing cell phenotype or reactivity. Taken together, my current studies present guiding principles and encouragement for the further development of immunotherapies for treatment of patients with malignant melanoma.
2

Magneto-plasmonic nanoparticle platform for detection of rare cells and cell therapy

Wu, Chun-Hsien, active 21st century 10 September 2015 (has links)
Magnetic and plasmonic properties combined in a single nanostructure provide a synergy that is advantageous in a number of biomedical applications, such as contrast enhancement in multimodal imaging, simultaneous capture and detection of circulating tumor cells, and photothermal therapy of cancer. These applications have stimulated significant interest in development of magneto-plasmonic nanoparticles with optical absorbance in the near-infrared region and a strong magnetic moment. In this dissertation, we addressed this need to create a novel immunotargeted magneto-plasmonic nanoparticle platform. The nanostructures were synthetized by self-assembly of primary 6 nm iron oxide core-gold shell particles, resulting in densely packed spherical nanoclusters. The close proximity of the primary particles in the nanoclusters generates a greatly improved response to an external magnetic field and strong near-infrared plasmon resonances. A procedure for antibody conjugation and PEGylation to the hybrid nanoparticles was developed for biomedical applications which require molecular and biocompatible targeting. Furthermore, we presented two biomedical applications based on the immunotargeted hybrid nanoparticle platform, including circulating tumor cell (CTC) detection and cell-based immunotherapy of cancer. In the CTC detection assays, rare cancer cells were specifically targeted by antibody-conjugated nanoparticles and efficiently separated from normal blood cells by a magnetic force in a microfluidic chamber. The experiments in whole blood showed capture efficiency greater than 90% for a variety of cancers. We also explored photoacoustic imaging to detect nanoparticle-labeled CTCs in whole blood. The results showed excellent sensitivity to delineate the distribution of hybrid nanoparticles on the cancer cells. Thus, these works paves the way for a novel CTC detection approach which utilizes immunotargeted magneto-plasmonic nanoclusters for a simultaneous magnetic capture and photoacoustic detection of CTCs. In another application, we introduced a novel approach to label cytotoxic T cells using the magnetic nanoparticles with an expectation to enhance T cell recruitment in tumor under external magnetic stimulus. A series of in vitro experiments demonstrated highly controllable manipulation of labeled T cells. Thus, these results highlight the promise of using our nanoparticle platform as a multifunctional probe to manipulate and track immune cells in vivo and further improve the efficacy of cell-based cancer immunotherapy. / text
3

Targeting Tumour Antigen Heterogeneity with Dual-Specific Adoptive Cell Transfer

Fisher, Robert January 2021 (has links)
Through the years, cancer therapies have progressed rapidly, pouring out novel treatments such as gene therapy, small molecule therapies and immunotherapy. One such immunotherapy, adoptive cell transfer (ACT), augmented through the addition of a chimeric antigen receptor (CAR), has proven success in treatment of hematological malignancies. Additionally, oncolytic viruses (OV) and OV-based (OVV) therapies, have shown promising results in both clinical and pre-clinical studies. In most instances, when applied as a monotherapy, the aforementioned treatment methods are incapable of inducing complete tumour remission. The Wan lab has developed an approach combining ACT with OVV therapies that dramatically increase therapeutic benefit resulting in complete regression of well-established solid tumours. Despite promising results, certain tumours can still escape this combination therapy through antigen loss resulting in antigen negative relapse (ANR). To further augment the therapy, the addition of a secondary receptor (CAR) provides the ACT multiple avenues of attack to prevent ANR. In this dissertation, we define culture conditions that promote strong expression of the CAR alongside confirmation of function in an in vitro setting. Following, it is demonstrated that OVV boosted dual-targeting T cells carry strong T cell activity by measure of cytokine release in vivo. Despite promising T cell activity data, dual-specific T cells are unable to improve tumour control and survival once relapse occurs. The failure to control relapse remains unclear however evidence points towards lack of T cell persistence, poor CAR function in vivo and a lack of endogenous T cell response leading to compounding effects that prevent dual-targeting T cells from preventing ANR. Although dual specific therapies have shown poor efficacy in preventing ANR, further study must be completed to identify areas of improvement – such as persistence, as the potential for success in using dual-targeting T cells coupled with OVVs still lies untapped. / Thesis / Master of Science (MSc)
4

Adoptive T cell therapy of breast cancer: defining and circumventing barriers to T cell infiltration in the tumour microenvironment.

Martin, Michele 03 November 2011 (has links)
In the era of personalized cancer treatment, adoptive T cell therapy (ACT) shows promise for the treatment of solid cancers. However, partial or mixed responses remain common clinical outcomes due to the heterogeneity of tumours. Indeed, in many patients it is typical to see a response to ACT in one tumour nodule, while others show little or no response. Thus, defining the tumour features that distinguish those that respond to ACT from those that do not would be a significant advance, allowing clinicians to identify patients that might benefit from this treatment approach. The first chapter of this thesis provides the necessary background to understand the principals behind and components of ACT. This chapter also offers selected historical advances contributing to the current state of the field. The second chapter introduces a novel murine model of breast cancer developed to investigate the tumour-specific mechanisms associated with immune evasion in an ACT setting. The third chapter describes the in vivo characterization of mammary tumour cell lines derived from our mouse model that reliably showed complete, partial or no response to ACT. Using these cell lines, we were able to characterize in vivo tumour-specific differences in cytotoxic T cell trafficking, infiltration, activation, and proliferation associated with response to ACT. In the fourth chapter, we used bioinformatics approaches to develop a preliminary predictive gene signature associated with response to ACT in our mammary tumour model. We used this signature to predict outcome and then test a number of murine mammary tumours in vivo, with promising results, wherein 50% of tumours responded to ACT as predicted based upon gene expression. Thus, using an innovative model for breast cancer, these results suggest that there are tumour-specific features that can be used a priori to predict how a tumour will respond to adoptive T cell therapy. Importantly, these findings might facilitate the design of immunotherapy trials for human breast cancer. / Graduate
5

Endogenous Lymphocytes Play a Critical Role in the Elimination of Solid Tumors in the Context of Adoptive Cell Combined with Oncolytic Vaccination / COOPERATION BETWEEN ENDOGENOUS LYMPHOCYTES AND ACT

Simovic, Boris January 2016 (has links)
A major obstacle in the implementation of adoptive cell therapy (ACT) for solid tumors is CD8+ T cell quantity and functional quality. In order to address this issue, the ACT field has directed considerable effort toward the generation of less-differentiated memory T cells (Tm), which demonstrate superior effector function and engraftment over effector T cells. An obstacle in using Tm for ACT is their requirement for in vivo activation before full effector function can be acquired. We sought to determine if a rhabdovirus expressing a defined tumor antigen (i.e. a rhabdoviral oncolytic vaccine) could activate adoptively-transferred Tm in vivo and eliminate established tumors. We used ex vivo cultured DUC18 TCR-transgenic Tm combined with a rhabdoviral oncolytic vaccine to target established CMS5 fibrosarcomas in both balb/c and NRG mice, and we compared the efficacy of the combination treatment versus monotherapies. Our data demonstrate that the rhabdoviral oncolytic vaccine was capable of expanding adoptively-transferred Tm in order to eliminate established tumors. Furthermore, synergy between ACT and oncolytic vaccination was required for optimal therapeutic outcome. Interestingly, we observed a population of endogenous, tumor-primed lymphocytes which appeared to be required for complete tumor elimination and subsequent memory formation. This was in contrast to the current consensus in the ACT field which is that endogenous lymphocytes are detrimental to therapeutic outcome, thus necessitating lymphodepletion prior to the commencement of therapy. Our data suggest that endogenous lymphocytes may be a beneficial cell population which is overlooked by current approaches to ACT. / Thesis / Master of Science (MSc) / Current approaches to the T cell therapy of cancer are hindered by poor cell quality. It is simple to grow higher quality T cells, but it is difficult to grow very large numbers of them. Furthermore, higher quality T cells need a signal in order to “switch on” before they can start killing cancer cells. Here, we use a cancer-targeting virus as a signal for these cells to activate, grow to very large numbers in the patient, and destroy their tumor. Our vaccine also switches on other immune cells in the patient, which help guarantee the destruction of the tumor. The significance of this work is that it will improve T cell therapy for cancer by opening the possibility of using higher-quality T cells which are much better at killing cancer than the currently used type of T cells.
6

Improving Adoptive Cell Therapy to Overcome Tumor Resistance / MS-275 Enhances Antitumor Immunity During Adoptive Cell Therapy to Overcome Tumor Resistance

Nguyen, Andrew 20 December 2021 (has links)
Cancer immunotherapy has gained attention in recent years for its successes in potentiating immune responses that can elicit tumor control. In particular, adoptive cell therapy (ACT), which involves the autologous/allogeneic transplant of ex vivo-cultivated tumor-specific T lymphocytes, can mediate potent tumor recognition and killing; however, durable clinical responses are often difficult to obtain in solid tumors. Solid tumors and their unique microenvironments have the capacity to evade and suppress antitumor immune responses and represent significant hurdles for effective ACT. Recently, we have discovered that chemical inhibition of histone deacetylases via MS-275 (Entinostat) during ACT can subvert tumor resistance to foster potent, broad-spectrum antitumor immunity. Overall, the work described supports the efficacy of ACT in the treatment of immunosuppressive, solid tumors; however, consistency in durable clinical outcomes can only be achieved through the concurrent therapeutic targeting of tumor resistance mechanisms. This thesis uses pre-clinical models to describe how tumor resistance to ACT can manifest, and demonstrates that concurrent MS-275 delivery drives extensive immunomodulation to promote sustained tumor clearance. This includes: 1) The polarization of tumor-infiltrating myeloid cells into cytotoxic effectors with the ability to reject immune escape variants 2) The inflammatory remodeling of the tumor microenvironment to potentiate epitope spreading against secondary tumor antigens 3) The transcriptional reprogramming of adoptively transferred T cells to overcome tumor-burden-dependent exhaustion We expect that the results will help facilitate the development of next-generation ACT platforms that will feature strategies for multi-mechanistic perturbation of tumor resistance. / Thesis / Doctor of Philosophy (PhD) / The host immune system has the ability to recognize and destroy tumor cells. Therapeutic platforms that leverage antitumor immune cells, specifically T cells, have shown potency in the elimination of cancer. In the clinic, cancer immunotherapies have demonstrated early success against hematological malignancies; however, are unreliable in the treatment of solid tumors. Solid tumors utilize intrinsic and adapted mechanisms of resistance to mitigate the effectiveness of cancer immunotherapy. This thesis pursues research questions aimed at understanding how tumors resist immunotherapy, what mechanisms are utilized, and how to overcome these obstacles. We anticipate that these results will contribute to the development and incorporation of strategies to subvert tumor resistance and potentiate T cells against solid tumors.
7

Strategies to Improve the Usability and Efficacy of CAR-T cell Therapy in NHL

Jackson, Zachary Gene 26 May 2023 (has links)
No description available.
8

Des antigènes particulaires synthétiques pour manipuler les fonctions anticorpsindépendantes des lymphocytes B : intérêt dans les stratégies d’induction de tolérance allo-immune / B cells loaded with synthetic particulate antigens : an alternative platform to generate antigen-specific regulatory T cells for adoptive cell therapy

Sicard, Antoine 27 June 2016 (has links)
Dans des modèles expérimentaux, une tolérance d'allogreffe a pu être induite en transférant des lymphocytes T CD4+ régulateurs (Treg) spécifiques d'antigènes (Ag) du donneur expandus ex vivo. Les données ont démontré l'importance des Treg d'allospécificité indirecte (Treg indirects) dans l'induction d'une tolérance à long terme. L'expansion de Treg indirects ex vivo est problématique, principalement à cause de la difficulté d'obtenir en grand nombre des cellules présentatrices d'antigène autologues (CPA)pour stimuler les Treg. Les lymphocytes B (LB) sont des APC accessibles, présentes en grand nombre et ont un fort potentiel régulateur. Cependant, l'utilisation de LB autologues comme APC est rendue problématique par leur incapacité à présenter les Ag dont ils ne sont pas spécifiques.Dans ce travail de thèse, nous avons développé une approche nanobiotechnologique permettant de transformer des LB polyclonaux autologues en puissants stimulateurs de Treg spécifiques de l'Ag.Des Ag particulaires synthétiques (SPAg) ont été générés en fixant sur des nanosphères fluorescentes de 400 nm de diamètre : (i) des anticorps monoclonaux dirigés contre un domaine constant de la chaine légère kappa du récepteur des LB, et (ii) des Ag modèles.Les SPAg se comportent comme des Ag particulaires naturels lorsqu'ils sont incubés in vitro avec des LB murins ou humains. Les SPAg se lient à la surface des LB kappa+, déclenchent un signal d'activation et sont internalisés dans leur endosomes. Les LB chargés en SPAg induisent l'activation et la prolifération des lymphocytes T CD4+ spécifiques de l'Ag in vitro.Des propriétés régulatrices peuvent être conférées aux LB chargés en SPAg en les stimulant avec du CpG. Les LB régulateurs générés n'induisent pas de prolifération des T CD4+ effecteurs mais, au contraire, entrainent une prolifération importante des Treg.Cette approche apparait comme une alternative innovante pour expandre des Treg spécifiques de l'Ag ex vivo / Allograft tolerance has been obtained in experimental models with adoptive transfer of ex vivo-expanded regulatory T cells (Treg) specific for donor antigens. Preclinical data have shown that Treg specific for indirectly presented alloantigens (indirect Treg) are mandatory for long-term tolerance. However, the ex vivo expansion of indirect Treg faces limitations,related essentially to the source of autologous antigen-presenting cells (APCs) used to stimulate T cells in vitro. B cells are (i) potent regulatory cells and (ii) APCs able to establish a privileged crosstalk with CD4+ T cells. However, the use of B cells as APCs is made problematic due to their inability to internalize and present non-cognate antigens. We have developed a novel nanobiotechnology-based approach to turn autologous polyclonal B cells into potent stimulators of antigen-specific T reg.Synthetic particulate antigens (SPAg) were generated by immobilizing (i) monoclonal antibodies directed against a framework region of B cell receptor (BCR) kappa-light chains and (ii) model antigens on fluorescent nanospheres of 400 nm in diameter.SPAg behaved like genuine particulate antigens when incubated in vitro with polyclonal murine B cells. SPAg bound to surface BCR of any kappa-positive B cells, triggered activation signal and were internalized in late endosomal compartment of B cells. SPAgloaded B cells induced activation and proliferation of antigen-specific T cells. This approach was transposable to humans’ cells. Importantly, regulatory properties could be conferred toSPAg-loaded B cells by CpG stimulation. SPAg-loaded regulatory B cells prevented proliferation of effector CD4+ T cells and induced proliferation of antigen-specific Treg in vitro.Autologous polyclonal B cells loaded with SPAg appear as an innovative platform to expand Treg ex vivo. This approach may improve the efficiency and costs of current procedures
9

Negative co-signaling in the expansion and function of human antigen-specific T-cells for adoptive cell therapy

Lak, Shirin 08 1900 (has links)
Immunotherapy, especially the adoptive transfer of T cells and immune checkpoint blockade therapy, have revolutionized cancer therapy. In particular, utilizing antigen-specific T cells for adoptive cell therapy has enabled the development of specific and effective strategies. It has paved the way for developing more accurate and personalized cancer immunotherapies. Adoptive cell therapy (ACT) results depend on the characteristics of ex vivo expanded T cells, such as their differentiation and clonal diversity. However, ex vivo expanded specific T cells often express several inhibitory receptors involved in T-cell exhaustion and markers of terminal effector differentiation. Accordingly, we hypothesized that blocking one or several inhibitory receptors during the ex vivo expansion could improve the expansion and differentiation of antigen-specific T cells. Preconditioning the ACT products and combinatorial immunotherapy approaches are newly developed concepts in cancer therapy to optimize cancer immunotherapy for a larger group of patients. To study the development of antigen-specific T-cells in combination with checkpoint blockade, we have adopted a method that allows the expansion of rare antigen-specific T cell precursors from PBMCs via multiple stimulations, using antigen-pulsed dendritic cells. In the current study, we utilized our protocol to generate and expand antigen-specific CD8+ T cells targeting the oncogenic Epstein-Barr virus (EBV)-LMP2 and a tumor-associated antigen (TAA) from the Wilms Tumor 1 (WT1) protein. We employed two approaches to abolish the negative regulatory receptors, antibody-mediated blockade and deletion via CRISPR/Cas9. We evaluated the impact of checkpoint blockade on antigen-specific T cells development, proliferation, and function. Additionally, TCR clonality and transcriptomic changes were assessed by genomic studies, including single-cell RNA (scRNA) sequencing and T-cell receptor sequencing. Supporting our hypothesis, we observed that blocking both PD-L1 and TIM3 (not any of them alone) significantly enhanced LMP2 and WT1-specific T cell generation and expansion. Additionally, checkpoint blockade resulted in higher specific T cell function, including cytokine production and in vitro targeted cytotoxicity. Using scRNA-seq and TCR sequencing approaches, we first remarked that the specific T cells are highly oligoclonal and identified a few dominant shared clones between donors. Immune checkpoint blockade did not confer consistent transcriptional signatures but may have a clonotype and donor-specific impact on the expression of activation and exhaustion-related genes. Overall, immune checkpoint blockade did not markedly alter the clonal composition of the T-cell product. We also evaluated the impact of CD5 deletion in antigen-specific T cell priming and expansion as an inhibitory receptor and a part of the immune response synapse. However, in a human ACT setting, our data show that the CRISPR/Cas9 mediated CD5 deletion only has modest effects on antigen-specific T-cell generation. However, future combinations with the blockade of other immune checkpoint may be warranted. Conclusion We demonstrated that blocking PD-L1 and TIM3 during the ex vivo expansion improves antigen-specific T-cell yield. We show that blocking multiple checkpoints can synergistically optimize specific T-cell production without compromising the response's specificity. It is a rapidly implementable strategy to enhance the number and quality of ex vivo expanded antigen-specific T cells for immunotherapy. / Le transfert adoptif de cellules T et le traitement par le blocage des points de contrôle immunologiques ont révolutionné le traitement du cancer. En particulier, l'utilisation de cellules T antigène-spécifiques en thérapie cellulaire adoptive a facilité le développement d'immunothérapies anticancéreuses plus précises et personnalisées. Les résultats de la thérapie cellulaire adoptive (TCA) sont liés à la qualité des cellules T spécifiques expansées ex vivo, telles que leur état de différenciation et leur diversité clonale. Cependant, le pré-conditionnement des produits de thérapie cellulaire adoptive et les traitements d'immunothérapie combinatoire sont de nouveaux concepts en développement de la thérapie du cancer pour optimiser l'immunothérapie du cancer dans un plus grand groupe de patients. Nous avons formulé l’hypothèse que le blocage d'un ou plusieurs récepteurs inhibiteurs au cours de l'expansion ex vivo favorise une meilleure expansion et une meilleure fonction des cellules T destinées à la TCA. Pour étudier l’expansion et la différenciation de cellules T antigène-spécifiques lors d’un blocage des points de contrôle, nous avons adopté une méthode qui nous permet de stimuler et expanser de rares cellules T antigène-spécifiques à partir de cellules mononuclées du sang périphérique (PBMCs) via de multiples stimulations utilisant des cellules dendritiques chargées avec l’antigène d’intérêt. Nous avons utilisé deux approches pour supprimer l’activité des récepteurs régulateurs négatifs, les anticorps bloquants des points de contrôle et la délétion génique via CRISPR/Cas9. Nous démontrons que le blocage combiné de PD-L1 et TIM3 améliore considérablement la l'expansion de cellules T CD8+ spécifiques à des antigènes viraux et tumoraux. De plus, le blocage des points de contrôle a entraîné la génération de cellules T spécifiques fonctionelles tel que démontré par la production de cytokines et la cytotoxicité in vitro. En utilisant de séquençage de l'ARN en cellule unique (scRNA-seq) et de séquençage des récepteurs des lymphocytes T (TCP-seq), nous avons remarqué que les cellules T spécifiques sont très fortment oligoclonales. Nous avons également identifié quelques clones dominants partagés entre les donneurs. L’application de l’inhibition des points de controles ne confère pas de signatures transcriptionelles particulières mais pourrait affecter certains clones provenant de certains donneurs davantage que d’autres. De plus, le peu de changements dans la composition clonale des cellules expandues suggèrent que le blocage de ces points de contrôle immunologiques n’altère pas de façon significative le produit cellulaire obtenu. Des données récentes soutiennent également un rôle du CD5 dans la régulation de l'activation des cellules T naïves et leur état fonctionnel. Cependant, dans un contexte compatible avec la TCA, nos données montrent que la suppression de CD5 via CRISPR/Cas9 n'a que des effets modestes sur la génération de cellules T antigène-spécifiques. Par contre, la combinaison éventuelle avec l’inhibition de d’autres points de contrôle immunologiques pourrait être envisagée. En conclusion, nos travaux fournissent une nouvelle méthode pour générer des cellules T spécifiques pour la TCA et la caractérisation à plus haute résolution de cellules T spécifiques expansées ex vivo. Nous avons donc démontré que le blocage combiné de plusieurs points de contrôle peut optimiser de manière synergique la production de cellules T spécifiques sans compromettre la spécificité de la réponse. Il s'agit là d'une stratégie rapidement applicable pour améliorer le nombre et la qualité des cellules T antigène-spécifiques expansées ex vivo pour l'immunothérapie.
10

Caractérisation de récepteurs de cellules T reconnaissant des antigènes spécifiques aux cellules leucémiques pour leur utilisation dans le cadre de thérapies

Aubin, Marie-France 08 1900 (has links)
La leucémie aigüe myéloïde est un cancer hautement létal notamment parce que le taux de rechutes est élevé, ce qui traduit l’importance du développement de nouvelles thérapies. Ces dernières peuvent tirer avantage du fait que les cellules leucémiques peuvent exprimer des antigènes qui ne sont pas exprimés par les tissus sains, soit les antigènes spécifiques aux tumeurs (TSA). À cet effet, nos collaborateurs ont découvert une source importante « d'aberrantly expressed TSA » (aeTSA) dans les régions non codantes de l’ADN. Ces aeTSA sont présentés par les molécules de CMH I et plusieurs ont provoqué la réactivité des lymphocytes T (LTs) in vitro. En plus d'être spécifiques aux cellules cancéreuses, ces aeTSA sont partagés entre plusieurs patients ce qui fait d'eux des cibles intéressantes dans le cadre d’immunothérapies. Sachant que c’est le récepteur de cellules T (TCR) qui confère la spécificité aux LTs, le but est d'isoler et de caractériser des TCR anti-aeTSA en vue de leur utilisation comme outils thérapeutiques. Pour ce faire, l’expansion de LTs CD8+ naïfs provenant de donneurs sains a été réalisée grâce à une co-culture avec des cellules dendritiques autologues chargées avec l'aeTSA d’intérêt. Les LTs CD8+ spécifiques du aeTSA ont été triés à l’aide d'un marquage dextramères et l’ARN a été isolé afin de réaliser le séquençage du TCR. Ce dernier a révélé que le répertoire de TCR anti-aeTSA est nettement oligoclonal, facilitant l'identification des séquences des chaînes α et β des clonotypes les plus abondants. En revanche, les répertoires de TCR anti-LMP2 426-434 (antigène viral) et anti-WT1 37-45 (antigène associé aux tumeurs) étaient plus diversifiés. De plus, des tests d'avidité fonctionnelle réalisés à l'aide d'ELISpot en concentrations décroissantes de peptides ont révélé que l'avidité fonctionnelle des LTs qui reconnaissent les aeTSA est similaire à celle du peptide LMP2 426-434, ce qui suggère que les aeTSA stimulent des réponses T de hautes avidités. Ensuite, la délétion du TCR endogène a été réalisée à l'aide de la technique CRISPR-Cas9, montrant plus de 90% d'efficacité. À des fins d'optimisation de protocoles, le TCR 1G4 spécifique de NY-ESO-1 a été introduit dans le locus TRAC et, simultanément, le knock-out de la chaîne α du TCR endogène a été réalisé afin de limiter les mésappariements et la compétition entre ces deux TCR. Les prochaines étapes seront d’introduire le gène codant pour le TCR spécifique d'aeTSA dans des LTs et de vérifier que les cellules éditées sont réactives envers ces aeTSA. Finalement, ce projet pourrait ouvrir la voie au ciblage d'aeTSA à l’aide de l’ingénierie du TCR pour rediriger un grand nombre de LTs envers les cellules leucémiques. / Acute myeloid leukemia is a highly lethal cancer for which effective immunotherapies are actively sought. These immunotherapies can take advantage of the fact that leukemia cells can express antigens that are not expressed by healthy tissues, namely tumor-specific antigens (TSA). In this regard, our collaborator's team has discovered an important source of aberrantly expressed TSA (aeTSA) in the non-coding regions of DNA. These aeTSAs are presented by MHC 1 molecules and can elicit T cells reactivity in vitro. In addition to being specific to cancer cells, these aeTSAs are shared between several patients, which makes them interesting targets in the context of immunotherapies. Knowing that the T cell receptor (TCR) is responsible for T cells specificity, the goal is to isolate and characterize anti-aeTSA TCRs for their use as therapeutic tools. To this end, we expanded aeTSA-specific T cells from naive CD8+ T cells obtained from healthy donors through co-culture with autologous dendritic cells loaded with the relevant aeTSA. The aeTSA-specific CD8+ T cells identified by dextramer staining were sorted for RNA extraction TCR sequencing. Amplicon sequencing reveals that the expanded anti-aeTSA TCR repertoire is markedly oligoclonal, facilitating the identification of dominant TCR α and β chains. In contrast, the anti-LMP2 426-434 (viral antigen) and anti-WT1 37-45 (tumor-associated antigen) TCR repertoires were more diverse. In addition, functional avidity tests, performed using ELISpot in decreasing concentrations of peptides, revealed that the functional avidity of T cells recognizing aeTSA is similar to LMP2 426-434 peptide, suggesting that aeTSAs stimulate high-avidity responses. Then, endogenous TCR knock-out was performed using the CRISPR-Cas9 technique, showing more than 90% efficiency. For protocol optimization purposes, the 1G4 TCR specific for NY-ESO-1 was introduced into the TRAC locus and, simultaneously, the knock-out of the α chain of the endogenous TCR was achieved in order to limit mismatches and competition between these two TCRs. The next steps will be to introduce the gene coding for the aeTSA-specific TCR into T cells and to validate that the edited cells are reactive toward these aeTSAs. Ultimately, this project could pave the way for targeting aeTSAs using TCR engineering to redirect large numbers of T cells toward leukemic cells.

Page generated in 0.0936 seconds