• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 15
  • 2
  • 2
  • Tagged with
  • 22
  • 22
  • 14
  • 8
  • 8
  • 7
  • 7
  • 5
  • 5
  • 5
  • 4
  • 4
  • 3
  • 3
  • 3
  • 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

The role of the aryl hydrocarbon receptor in autoimmunity and tumor immunity

Kenison-White, Jessica E. 06 March 2021 (has links)
At the intersection between autoimmune disease and cancer lies a disruption in the balance of our body’s critically important immune system, and, specifically, in its regulation. While autoimmune diseases are the result of overactivation and a failure to regulate improper responses to the body’s own tissues, cancer is the result of improper suppression and a failure to recognize and eradicate transformed malignant cells. Although they are fundamentally different conditions, overlap can be found in the pathways which are critical to disease progression and which may represent important therapeutic targets. One such pathway implicated in both autoimmunity and cancer is the aryl hydrocarbon receptor (AhR). AhR activation suppresses immune cell activation through the modulation of T cell differentiation and antigen presenting cell (APC) function. AhR activation shows a beneficial therapeutic effect in models of autoimmune disease, but has also been implicated in driving cancer progression and tumor-mediated immunosuppression. While it is clear that the AhR plays an important role in the immune response, the mechanisms behind AhR regulation of the immune system and the effects of its modulation in autoimmunity and cancer are still not fully understood. Thus, in this work, we investigated the effect of targeting the AhR in models of autoimmunity and cancer, using the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS) and the murine oral cancer (MOC) model of oral squamous cell carcinoma (OSCC). We demonstrated that AhR activation using the endogenous ligand 2-(1’H-indole-3’-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) induced a tolerogenic transcriptional response in mouse and human dendritic cells (DCs) associated with the induction of immunoregulatory/immunosuppressive mechanisms. We further showed that targeting the AhR using a nanoliposome (NLP) delivery platform, which co-encapsulated a MS autoantigen, suppressed the development of EAE in multiple models, both in preventative and therapeutic contexts. This disease suppression was associated with the expansion of antigen-specific FoxP3+ regulatory T cells (Treg cells) and IL10+ type 1 regulatory T cells (Tr1 cells), and a reduction in CNS-infiltrating effector T cells (Teff cells). Using the MOC1 model of OSCC we demonstrated that deletion of the AhR in MOC1 malignant cells completely blocks in vivo tumor growth in an immune system-dependent manner and renders mice completely immune to either local or systemic re-challenge with wildtype MOC1 cells. Suppression of tumor growth was associated with a decrease in the expression of suppressive immune checkpoint markers including PD-L1 and CD39 on macrophages, dendritic cells, and Ly6G+ myeloid cells, and PD-1, CTLA4, Lag3, and CD39 on CD4+ T cells. Further, the AhR was found to control expression of chemokines and immunosuppressive IDO and PD-L1 in malignant cells themselves, suggesting that AhR activity in tumor cells may simultaneously regulate multiple immune checkpoints. Taken together, these results provide new insight into the critical role for the AhR in both autoimmunity and cancer, and confirm it as a valid therapeutic target for both diseases. / 2022-03-05T00:00:00Z
2

B7-H3 suppresses anti-tumor immunity via the CCL2-CCR2-M2 macrophage axis and contributes to ovarian cancer progression / B7-H3はCCL2-CCR2-M2マクロファージ経路を介して抗腫瘍免疫を抑制し、卵巣癌の進展に寄与する

Miyamoto, Taito 23 March 2022 (has links)
京都大学 / 新制・課程博士 / 博士(医学) / 甲第23801号 / 医博第4847号 / 新制||医||1058(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 小林 恭, 教授 竹内 理, 教授 金子 新 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
3

Immunosuppressive tumor microenvironment in Uterine Serous Carcinoma via CCL7 signal with myeloid-derived suppressor cells / 子宮体部漿液性癌における骨髄由来抑制細胞とCCL7シグナルを介した免疫抑制性腫瘍微小環境の解明

Mise, Yuka 24 November 2022 (has links)
京都大学 / 新制・課程博士 / 博士(医学) / 甲第24284号 / 医博第4900号 / 新制||医||1061(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 羽賀 博典, 教授 上野 英樹, 教授 髙折 晃史 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
4

T cell costimulation in anti-tumor immunity and autoimmunity

May, Kenneth F., Jr. 01 December 2004 (has links)
No description available.
5

Traitement anti tumoral par ciblage de TLR3 et découplage des effets opposés des chimiokines pour améliorer l’efficacité des agonistes de TLR3 / Anticancer treatment by TLR3 targeting and uncoupling of the chemokines opposing effects to optimize the efficacy of TLR3 agonists

Conforti, Rosa 24 October 2011 (has links)
Le rationnel pour l’utilisation des agonistes des Toll-Like Récepteurs (TLR) dans le traitement du cancer repose sur leurs effets bénéfiques au niveau des cellules du système immunitaire permettant une stimulation de la réponse immunitaire innée et adaptative. Cependant, certains types de cellules tumorales expriment les TLRs qui, une fois activés, peuvent déclencher des effets “délétères” comme la tumorigénèse.Pour mieux disséquer les effets biologiques directs et indirects d’un agoniste de TLR3, l’acide polyadenylique-polyurydilique (poly(A:U)), nous avons utilisé deux modèles tumoraux murins exprimant TLR3 et ne répondant pas à la chimiothérapie, mais capables de produire des grandes quantités de CCL5 et CXCL10 en réponse au poly(A:U) et à l’IFN de type I. In vivo, la combinaison chimiothérapie – poly(A:U) n’a permis d’obtenir qu’une faible activité anti tumorale sauf lorsqu’une vaccination contre les antigènes tumoraux est inclue dans le protocole de traitement et le récepteur CCR5 est bloqué (souris Ccr5-/- ou souris sauvages traitées avec MetRANTES). L’efficacité anti tumorale du traitement combiné est associée à l’induction de cellules T CD8+CXCR3+IFNγ+ et est perdue chez les souris nu/nu, Trif-/- et Cxcr3-/-. La source du ligand de CCR5 est constituée par les cellules tumorales dont la voie TLR3 est activée. L’efficacité du traitement combiné sur ces cellules a été améliorée en inhibant la production de CCL5 à l’aide d’un shARN spécifique pour CCL5. Ces résultats soutiennent la notion que le poly(A:U) peut directement agir sur l’épithélium tumoral pour promouvoir la libération de CXCL10 qui a un effet bénéfique (recrutement des LTCs), mais aussi la sécrétion de CCL5 qui a un rôle délétère (action sur des immunosuppresseurs au niveau de l’hôte). Le découplage des effets opposés des chimiokines et la vaccination anti tumorale préalable peuvent permettre aux LTCs dépendant des ligands de CXCR3 d’infirmer l’action d’immunosuppression CCR5 dépendante et devraient être intégrés dans les essais cliniques à venir utilisant les agonistes de TLR3. / The rationale for the use of Toll –Like Receptor (TLR) agonists in cancer therapy relies upon their “beneficial” effects on immune cells leading to enhanced innate and adaptive immune responses. However, a variety of cancer epithelia express TLRs which, upon triggering, may mediate “deleterious” effects such as tumorigenesis. To further dissect the direct versus indirect biological effects of the TLR3 agonist polyadenylic-polyuridylic acid (poly(A:U)), we took advantage of two murine tumor models expressing TLR3 that failed to respond to chemotherapy but did produce large amounts of CCL5 and CXCL10 in response to the poly(A:U) and type I IFN. In vivo, the combination of chemotherapy and poly(A:U) mediated low tumoricidal activity unless a vaccination against tumor antigens was included in the regimen and the CCR5 receptor was blocked (CCR5 loss-of-function mice or WT animals treated with MetRANTES). The antitumor efficacy of the combination therapy was associated with the elicitation of CD8+CXCR3+IFN+ T cells and abrogated in nu/nu, Trif-/- and Cxcr3-/- mice. The source of CCR5L is the TLR3-activated tumor cells in that stable inhibition of the chemokine production by specific shRNA CCL5 ameliorated the efficacy of the combination therapy. These results support the notion that poly(A:U) can directly act on tumor epithelia to promote the release of beneficial CXCL10 for the recruitment of intratumoral CTLs but also the release of deleterious CCL5 acting on host immunosuppressors. Uncoupling chemokine release and prior vaccination may enable the CXCR3L-dependent CTLs to overrule the CCR5-dependent suppression and may be integrated in future trials using TLR3 agonists.
6

Lenalidomide targets the T-cell co-stimulatory pathway to mediate immune modulation

Mcdaniel, Jessica Marie 01 January 2012 (has links)
T-cells are lymphocytes that make up part of the adaptive arm of the immune system, and are essential for efficient protection from and eradication of viruses and pathogens. T-cells not only play an important role in protection from external agents, but also regulate and prevent activation towards self-peptides and detect and remove erratically growing cells. Alterations in T-cell activation and suppression contribute to auto-immunity, immunocompromised disorders, and cancer progression. The immune system, and T-cells in particular, provides daily surveillance, recognition and destruction of aberrant cells. Although the immune system is proficient at suppressing malignant progression, tumor cells acquire various methods of immune evasion. Myelodysplastic Syndrome (MDS) is a pre-malignant dysplastic disorder of the bone marrow characterized by ineffective hematopoiesis and clonality in the myeloid lineage, where lack of immune response has been implicated in the propensity for progression to acute myeloid leukemia (AML). Leukemia progression is associated with the acquisition of complex genetic abnormalities. Alterations in immune system regulation have been implicated in various stages of the disease process, although the role of the immune system in response to several therapies in MDS has not been fully discovered. Lenalidomide is a small molecule therapeutic preferentially effective in MDS patients with an interstitial chromosome 5q deletion (del(5q)). Improved erythropoiesis has also been reported to occur in 20-30% of low-risk, non-del(5q) patients. Although lenalidomide is a potent immunomodulatory drug that potentiates T-cell and NK-cell responses, the T-cell compartment in MDS is highly deregulated by aberrant repertoire skewing, decreased function and abnormal naïve and memory cell homeostasis. The presence of lymphoid infiltrates in the bone marrow of lenalidomide-responsive patients suggests that T-cells may participate in the hematopoietic response, but it is unclear if lenalidomide is capable of reversing these functional T-cell defects. We therefore assessed immunological changes in low-risk MDS patients before and after 16-weeks of lenalidomide therapy, and assessed the relationship to erythroid response. Although MDS T-cells were anergic prior to treatment, we have shown that T-cells in responders have a significant increase in antigen-induced proliferative response and T helper type-1 (Th1) cytokine production (IL-2, IFN-γ, TNF-α) compared to non-responders. The change in function positively correlated with an increase in naïve T-cells and a decrease in memory cells, indicating that lenalidomide has immunomodulatory activity to reverse anergy in MDS. Although it is known that lenalidomide may increase T-cell activation and proliferation in the absence of co-stimulatory signals, a direct mechanism of action has yet to be elucidated. Since CD28 is one of the most important co-stimulatory molecules deregulated in cancer, we therefore set out to determine if the expression of CD28 was essential for lenalidomide's mechanism in T-cells. We knocked out CD28 expression in healthy donor T-cells, and sorted on inherently deficient, CD28null, T-cells that accumulate in older healthy donors and found that lenalidomide-induced proliferation and function were completely ablated within the CD28null subset. These data indicate the immunotyrosine-based activation motifs (ITAMs) on the intracellular domain of the CD28 receptor are necessary for lenalidomide action. Interestingly, during the natural aging process, repeated exposure to antigens results in the accumulation of CD28null T-cells that are phenotypically distinct and functionally deficient due to excessive proliferative history in vivo. We therefore examined whether CD28 expression on MDS patient T-cells affected responses to lenalidomide, and if this could be used as a predictive biomarker of responsiveness. We found that patients who fail lenalidomide therapy had higher CD8+ Terminal Effector Memory (TEM), which are inherently CD28null, and that non-responders had an overall increase in CD4+ and CD8+CD28null T-cells, as well as an increase in CD28null cells within the TEM compartment compared to hematologic responders. We then sought to determine the particular protein target of lenalidomide responsible for increased CD28 receptor signaling in T-cells. Several targets in a variety of cell types have been postulated, although the direct mechanism in T-cells is unclear. Our group has previously shown that lenalidomide inhibits the activity of two haplodeficient phosphatases located within the commonly deleted region (CDR) on chromosome 5q in the MDS myeloid clone, Protein Phosphatase 2A (PP2A) and Cdc25c. PP2Ac is known to bind CD28 and is hypothesized to inhibit T-cell co-stimulation. Therefore, it is plausible that lenalidomide and other IMiDs inhibit the phosphatase activity of PP2A which leads to increased activation of T-cell proximal signals dependent on CD28 expression. We examined this hypothesis using molecular modeling and virtual screening and found that all of the IMiDs (lenalidomide, pomalidomide, and thalidomide) can theoretically interact with the catalytic pocket of the PP2A heterotrimer, potentially inhibiting PP2Ac activity. In vitro phosphatase activity assays supported these findings as lenalidomide-inhibition of PP2Ac activity was seen in both ad293 and Jurkat cell lines, and in primary T-cells. Mutations of theorized lenalidomide hydrogen-bond sites within the catalytic pocket of PP2A rendered the enzyme catalytically dead, indicating that these are important residues for enzymatic activity, but unfortunately could not be used to determine if lenalidomide activity was disrupted by mutation of those sites. These data together suggest that the ability of lenalidomide to augment immune activation in vivo in MDS patients, and potentially other diseases, is extremely important to patient response. Also, that CD28 expression on T-cells is essential for lenalidomide immune-mediated tumor eradication through CD28 downstream signaling, and potentially through inhibition of PP2A. These results are useful in designing future lenalidomide-combination therapy trials in other hematologic and solid malignancies, and could be used to help stratify patients for future therapeutic decisions in MDS and other malignancies.
7

Accumulation of exhausted CD8+ T cells in extramammary Paget’s disease / 乳房外Paget病において腫瘍浸潤CD8陽性T細胞は疲弊している

Iga, Natsuko 23 May 2019 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第21957号 / 医博第4499号 / 新制||医||1037(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 河本 宏, 教授 濵﨑 洋子, 教授 武藤 学 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
8

Serum immunoglobulin G Fc region N-glycosylation profiling by matrix-assisted laser desorption/ionization mass spectrometry can distinguish breast cancer patients from cancer-free controls / マトリックス支援レーザー脱離イオン化質量分析装置による血清IgG Fc領域のN型糖鎖修飾プロファイリングにより非がんコントロールと乳がん患者を識別することができる

Kawaguchi, Nobuko 25 July 2016 (has links)
Final publication is available at http://dx.doi.org/10.1016/j.bbrc.2015.12.114 CreativeCommonsAttribution Non-Commercial No Derivatives License の記載が必要 / 京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第19924号 / 医博第4144号 / 新制||医||1017(附属図書館) / 33010 / 京都大学大学院医学研究科医学専攻 / (主査)教授 武藤 学, 教授 野田 亮, 教授 小川 修 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
9

Novel Approaches in Pancreatic Cancer Treatment: Bridging Mechanics, Cells, and Immunity

Imran, Khan Mohammad 04 January 2024 (has links)
The heterogeneity of pancreatic cancer renders many available general therapies ineffective holding the five-year survival rate close to 10% for decades. Surgical resection eligibility, resistance to chemotherapy and limited efficacy of immunotherapy emphasize the dire need for diverse and innovative treatments to combat this challenging disease. This study evaluates co-therapy strategies that combine non-thermal, minimally invasive ablation technology and targeted drug delivery to enhance treatment efficacy. Our research begins by uncovering the multifaceted potential of Irreversible Electroporation (IRE), a cutting-edge non-thermal tumor ablation technique. This study demonstrates IRE-mediated ability to trigger programmed necrotic cell death, induce cell cycle arrest, and modulate immune cell populations within the tumor microenvironment. This transformation from a pro-tumor state to a proinflammatory milieu, enriched with cytotoxic T lymphocytes and neutrophils. IRE-induced proinflammation in the tumor site renders immunologically "cold" tumor into immunologically "hot" tumor and holds significant promise of improving treatment efficacy. Notably, IRE-treated mice exhibited an extended period of progression-free survival, implying clinical potential. The transient nature of these effects suggests potential mechanisms of tumor recurrence highlighting the need for further studies to maximize the efficacy of IRE. Our mechanistic studies evaluated the IFN-STAT1-PD-L1 feedback loop as a possible reason for pancreatic tumor recurrence. Our data also suggest a stronger IFN-PD-L1 feedback loop compared to mammary, osteosarcoma and glioblastoma tumors rendering pancreatic cancer immunologically "cold". This study also investigates the use of histotripsy (a non-thermal, noninvasive, nonionizing ultrasound-guided ablation modality) to treat pancreatic cancer utilizing a novel immunocompromised swine model. We successfully generated human orthotopic pancreatic tumors in the immune deficient pigs, which allowed for consequent investigation of clinical challenges presented by histotripsy. While rigorous clinical studies are indispensable for validation, the promise of histotripsy offers new hope for patients. In parallel, we used our immunocompromised swine model of orthotopic pancreatic cancer to investigate the SonoTran® system, which employs ultrasound-activated oscillating particles to enhance drug delivery within hard-to-reach tumors. Our study demonstrates that SonoTran® significantly enhances the intratumoral penetrance of therapeutic agents, including commonly used chemotherapy drugs like paclitaxel and gemcitabine. Additionally, SonoTran® improved delivery of the anti-epidermal growth factor (EGFR) monoclonal antibody, cetuximab- which is frequently used in cancer immunotherapy. Together, our findings address challenges in the delivery of a range of therapeutics while simultaneously exposing challenges like off-target damage. In conclusion, this study presents a multifaceted approach to confront the complex characteristics of pancreatic cancer. Given the variations in patient response and the complexity of the disease, it is clear that a singular solution is unlikely. Our research, which combines IRE, histotripsy, and SonoTran®, to interrogate a promising array of tools to tackle different challenges to provide tailored treatments. In the ever-evolving landscape of pancreatic cancer therapy, this research opens new avenues to investigate deeper into molecular mechanisms, co-therapy treatment options, future preclinical and clinical studies which eventually encourage the potential for improved patient outcomes. / Doctor of Philosophy / Pancreatic cancer is a formidable disease, known for its late-stage diagnosis and limited treatment options with a poor 5-year survival rate of ~10%. However, a promising frontier in the battle against this lethal disease has emerged through combining mechanical, cell based and immunotherapies to attack the cancer from multiple angles at once. In my PhD research, I explored novel approaches to transform the landscape of pancreatic cancer treatment. We began by investigating Irreversible Electroporation (IRE), a non-thermal method to ablate tumors. Beyond its known function of reducing tumor size, IRE initiated programmed necrotic cell death, halted tumor cell division, and triggered changes in the immune landscape within the tumor. In response to IRE treatment, the immune environment shifted from pro-tumor to proinflammatory state, showing potential for clinical use. Mice treated with IRE experienced extended cancer progression-free survival temporarily, followed by eventual relapse. During relapse, we found that immune cells reverted back to their original, pre- IRE treated state. This observation logically implies combining IRE and immune checkpoint inhibitors aimed towards maintaining the IRE-altered immunological environment. Next, we developed and used novel pig models that closely resemble human pancreatic cancer patients to test histotripsy, a first phase toward making histotripsy as a non-invasive treatment approach for pancreatic cancer. Use of orthotopic tumor in a large animal model and clinical device allowed us to expose some challenges of ultrasound guidance of histotripsy. Notably, the treatment results in partial ablation and a reduction in stroma materials, which play a role in the tumor's resistance to commonly used treatments. While rigorous clinical studies are needed for validation, this approach offers hope in the quest for innovative pancreatic cancer treatment. Another promising approach we investigated involves SonoTran® particles, ultrasound-activated oscillating particles that can increase drug absorption in a targeted fashion. Our study demonstrated increased concentrations of commonly used therapeutic agents within tumors through SonoTran®-facilitated delivery, providing an effective means to overcome drug delivery issues within pancreatic tumors. There is no one size fits all treatment to address the complexity of pancreatic cancer. The future of treatment lies in the integration of IRE, histotripsy and SonoTran® into clinical practice. In summary, this PhD research identified promising novel technologies and combinations of treatments for pancreatic cancer, reaffirming the importance of exploring innovative solutions to combat pancreatic cancer. The dynamic nature of the pancreatic tumor microenvironment underscores the importance of further research to extend the positive impacts of these treatments and improve tumor debulking.
10

INVESTIGATING MECHANISMS OF CANCER VACCINE-INDUCED TUMOR IMMUNITY AND AUTOIMMUNITY

Bernard, Dannie 10 1900 (has links)
<p><strong>INTRODUCTION: </strong>Pre-clinical and clinical data strongly support the feasibility of employing immunotherapy as a strategy to treat cancer.</p> <p><strong>METHODS: </strong>Using the B16F10 murine melanoma model, we have been investigating mechanisms of T cell-mediated antitumor immunity following immunization with dopachrome tautomerase (DCT), a melanoma-associated antigen.</p> <p><strong>RESULTS: </strong>In <strong>Chapter 2</strong>, we uncovered an interesting dichotomy whereby DCT-specific CD4<sup>+</sup> T cell-mediated tumor protection and autoimmunity are dependent on IL-4/STAT-6 and IFN-g/STAT-4, respectively. Our data also revealed that this phenomenon is extrinsic of CD4<sup>+</sup> T cell polarization.</p> <p>To gain further insight into the targets recognized by CD4<sup>+</sup> T cells, we conducted in <strong>Chapter 3</strong> extensive CD4<sup>+</sup> T cell epitope mapping experiments using overlapping peptide libraries. Interestingly, while we were able to identify “helper” epitopes within DCT that were required for maximal CD8<sup>+</sup> T cell expansion, we were unable to identify “effector” epitopes responsible for tumor rejection. Further examination of the requirements for the generation of CD4<sup>+</sup> T cell effector epitopes showed that post-translational modifications of the protein were involved.</p> <p>In <strong>Chapter 4</strong>, we investigated the modest efficacy afforded by DCT immunization in the context of established B16F10 melanomas. Using intratumoral transcriptional analysis, we demonstrated that the vaccine rapidly promoted an IFN-g-dependent immunosuppressive state inside the tumor. Concurrent treatment with the immunomodulatory antibodies anti-4-1BB and anti-PD-1 effectively counteracted this tumor immunosuppression, resulting in complete regression of tumors and long-term survival in 70% of the mice.</p> <p><strong>CONCLUSIONS: </strong>The research described in this thesis sheds new light into the mechanisms by which vaccine-mediated CD4<sup>+</sup> T cell responses participate to tumor rejection and autoimmunity. Moreover, our findings indicate that cancer vaccine-induced tumor immunosuppression significantly limits tumor regression, emphasizing the requirement of combinatorial approaches for successful cancer immunotherapy. Overall, our research offers new insight for future vaccine development.</p> / Doctor of Philosophy (Medical Science)

Page generated in 0.0641 seconds