• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 68
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 81
  • 81
  • 49
  • 25
  • 22
  • 14
  • 12
  • 8
  • 8
  • 7
  • 7
  • 7
  • 7
  • 6
  • 6
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
61

Oncolytic Virus Therapy in Combination with Chemotherapy for Ovarian Cancer.

Bolyard, Chelsea M. January 2013 (has links)
No description available.
62

Improving Oncolytic Viral Therapy for Primary and Metastatic Tumors in the Brain

Meisen, Walter Hans 22 May 2015 (has links)
No description available.
63

IMPROVING ONCOLYTIC VESICULAR STOMATITIS VIRUS THROUGH MODULATION OF THE ANTI-TUMOUR IMMUNE RESPONSE

Stephenson, Kyle B. 04 1900 (has links)
<p>Despite improvements in detection and treatment, cancer is the leading cause of death worldwide. Current treatment modalities have not been able to improve the mortality rates and significant toxicities limit efficacy. Therefore there is a need for development of novel therapeutics.</p> <p>Oncolytic viruses have the ability to efficiently replicate in and destroy tumours while leaving normal tissues unharmed. These treatment platforms have been gaining momentum in recent years due to pre-clinical and clinical successes. Oncolytic viruses are extremely safe with limited toxicity observed in phase I/II clinical trials, and objective responses have been observed in some patients treated with oncolytic viruses. However, there is still room to improve on these therapeutic platforms.</p> <p>Recently, the importance of the induction of anti-tumour immunity during oncolytic virotherapy has been realized and harnessing this immune response can be used to improve current oncolytic virus platforms. To this end we have conducted numerous studies assessing our ability to improve oncolytic VSV through the addition of transgenes to enhance the immunostimulatory properties of oncolytic VSV treatment. These studies showed that only the addition of a highly secreted form of human IL-15 was able to improve VSV therapy through enhanced anti-tumour immunity. However, expressing cell-autonomous transgenes from oncolytic VSV was unable to modify the therapeutic efficacy of VSV due to limited replication, both temporally and geographically within the tumour, and the indirect vascular shutdown induced by VSV infection of tumours. We believe that the drastic vascular shutdown observed following VSV therapy is an important component to the success of VSV and we have investigated which steps in this process are critical for induction of anti-tumour immunity.</p> <p>The research presented in this thesis further enforces the requirement for induction of anti-tumour immune responses in the success of OV therapy. Our findings also indicate that manipulating the tumour as a whole, rather than the virus, will lead to improved oncolytic therapeutics.</p> / Doctor of Philosophy (PhD)
64

Analysis of Human Appendiceal Peritoneal Carcinomatosis Samples Infected with Oncolytic Viruses

Zerhouni, Siham 11 December 2013 (has links)
Peritoneal carcinomatosis (PC), the intra-abdominal dissemination of malignancy, is equated with a 5-year survival of 15%, depending on the source. Appendiceal PC is a challenge to treat as cancer cells are embedded in copious amounts of mucin and are difficult to target. Oncolytic viruses (OVs) preferentially replicate and lyse cancer cells and present a targeted, novel strategy for PC. The hypothesis of this study is that appendiceal PC will show variable susceptibility to OVs and that protein expression in these tumours will predict OV replication efficiency. Human appendiceal PC infected ex-vivo with 4 different OVs displayed variable infectivity and replication by fluorescence microscopy and plaque assay. Immunohistochemistry analysis revealed differential expression of IRF3, pERK and TK in tumour compared to normal appendix. No correlation of protein expression with viral replication was observed. Personalizing OV therapy will be critical in the optimization of future care of patients treated with this modality.
65

Analysis of Human Appendiceal Peritoneal Carcinomatosis Samples Infected with Oncolytic Viruses

Zerhouni, Siham 11 December 2013 (has links)
Peritoneal carcinomatosis (PC), the intra-abdominal dissemination of malignancy, is equated with a 5-year survival of 15%, depending on the source. Appendiceal PC is a challenge to treat as cancer cells are embedded in copious amounts of mucin and are difficult to target. Oncolytic viruses (OVs) preferentially replicate and lyse cancer cells and present a targeted, novel strategy for PC. The hypothesis of this study is that appendiceal PC will show variable susceptibility to OVs and that protein expression in these tumours will predict OV replication efficiency. Human appendiceal PC infected ex-vivo with 4 different OVs displayed variable infectivity and replication by fluorescence microscopy and plaque assay. Immunohistochemistry analysis revealed differential expression of IRF3, pERK and TK in tumour compared to normal appendix. No correlation of protein expression with viral replication was observed. Personalizing OV therapy will be critical in the optimization of future care of patients treated with this modality.
66

Impairment of the Type I Interferon Response in HIV-Infected Macrophages Facilitates their Infection and Killing by the Oncolytic Virus, MG1

Sandstrom, Teslin Stella 28 May 2019 (has links)
HIV remains an incurable viral infection and a significant global health concern. Despite the advent of antiretroviral therapy, there are 36.9 million recorded cases of HIV worldwide, with an additional 1.8 million new infections recorded in 2017 alone. An HIV cure is therefore one of several priorities within the field, and will require HIV “reservoir” cells—comprised of latently-HIV infected CD4+ T cells and productively-infected, tissue resident macrophages—to be selectively killed in vivo. HIV reservoir cells are rarely found within the peripheral circulation, residing instead within inaccessible tissue sanctuaries. Consequently, their characterization has been limited to in vitro laboratory models. To complicate matters further, a definitive cellular surface marker of HIV infected cells has yet to be identified. Impairment of the type I interferon (IFN1) response has been observed during HIV infection, however, making it a unique intracellular maker of HIV-infected cells. The recent development of oncolytic viruses (OV) designed to selectively kill IFN-defective cancer cells also suggests that these IFN1 defects possess therapeutic value. It was therefore hypothesized that the impairment of the IFN1 response in HIV-infected CD4+ cells and macrophages could serve as a target for oncolytic virus-mediated killing. The induction of several antiviral IFN-stimulated proteins, including PKR and ISG15, was inhibited in HIV-infected monocyte-derived macrophages (MDM) following stimulation with IFNα or a synthetic RNA. Consequently, HIV-infected MDM were more susceptible to infection and killing by the oncolytic Maraba virus, MG1. Importantly, MG1-mediated killing required the presence of replication-competent OV, and could not be potentiated by UV-inactivated MG1 or supernatants from MG1-infected cells. The ability of MG1 to target the HIV reservoir was further confirmed using alveolar macrophages collected from the lungs of cART-suppressed individuals living with HIV. These findings indicate that IFN1 defects are a feature of HIV infected cells, which can be exploited for selective killing by OV. This project is therefore unique in that it demonstrates that HIV reservoir cells can be eradicated in a targeted manner by exploiting an intracellular marker of HIV infection. As MG1-based cancer therapies are currently being explored in Phase I/II clinical trials, there is potential for this approach to be adapted for use within the HIV cure field.
67

Modelos fracionários de terapia gênica para o tratamento do câncer

Tavoni, Robinson. January 2019 (has links)
Orientador: Rubens de Figueiredo Camargo / Resumo: Câncer é umas das maiores causas de mortalidade mundial. Os tratamentos mais convencionais, tais como quimioterapia e radioterapia, além de debilitarem muito o paciente também matam as células normais. Em contrapartida, e em fase de tratamento experimental e ensaios clínicos, a terapia gênica tem poucos efeitos colaterais e provoca a morte seletiva das células tumorais. Este trabalho apresenta alguns conceitos do Cálculo Fracionário e o Método de Grünwald-Letnikov para simulação numérica de Equações Diferenciais Fracionárias, também estuda alguns modelos matemáticos de ordem inteira e não inteira, com derivada temporal de Caputo, que visam descrever o tratamento por terapia gênica. A análise de estabilidade e as simulações numéricas exibiram que mudanças no cenário da dinâmica tumoral estão relacionadas com a ordem da derivada fracionária, no qual observou-se uma atuação mais eficiente do tratamento quando o modelo está associado com ordens menores da derivada fracionária. / Abstract: Cancer is one of the biggest causes of worldwide mortality. Conventional treatments, such as chemotherapy and radiotherapy, in addition to weakening the patient also kill normal cells. On the other hand, in experimental treatments and clinical trials, gene therapy has few side effects and causes selective killing of tumor cells. This work presents some concepts of the Fractional Calculus and the Grünwald-Letnikov Method for numerical simulation of Fractional Differential Equations, also studies some mathematical models of integer order and non-integer order, with temporal derivative of Caputo, that aim describe the treatment by gene therapy. Stability analysis and numerical simulations have shown that changes in the scenery of the tumor dynamics are related to the order of the fractional derivative, in which a more efficient treatment performance was observed when the model is associated with smaller orders of the fractional derivative. / Doutor
68

An Interleukin-12-Expressing Oncolytic-Virus Infected Autologous Tumor Cell Vaccine Generates Potent Anti-Tumor Immune Responses

Khan, Sarwat Tahsin 30 July 2018 (has links)
No description available.
69

Immunostimulatory and Oncolytic Properties of Rotavirus Can Overcome Resistance to Immune Checkpoint Blockade Therapy / Voie de signalisation de TLR4 et Rig I dans le neuroblastome : rôle biologique, valeur pronostique et utilisation dans les thérapeutique ciblées

Shekarian, Tala 27 March 2017 (has links)
L'apport des anticorps immunomodulateurs ciblant PD-1, PD-L1 et CTLA-4 ont récemment révolutionné la prise en charge thérapeutique du cancer. Cependant, seule une minorité de patients développent des réponses objectives à ces traitements. Par conséquent, de nouvelles innovation thérapeutiques sont nécessaires afin d'augmenter l'immunogénicité des tumeurs et de surmonter la résistance à la thérapie contre les anticorps immunomodulateurs. Les propriétés oncolytiques de certains virus peuvent être exploitées afin de permettre un amorçage de l'immunité anti-tumorale. Différents virus oncolytiques (OVS) sont actuellement en développement clinique intense en combinaison avec des thérapies par anticorps immunomodulateurs. Nous avons trouvé qu'un vaccin viral pédiatrique disponible dans le commerce a des propriétés oncolytiques. Ce virus pédiatrique peut tuer directement les cellules cancéreuses, avec des caractéristiques de mort cellulaire immunogène. De plus, ce virus a des propriétés pro-inflammatoires et peut activer la voie NF-kB indépendamment des voies de danger cellulaire (TLR et IRF). Ces propriétés biologiques in vitro se traduisent in vivo en une activité anti-tumorale. L'Injection intra-tumorale du vaccin a des effets anti-tumoraux directs mais également à médiation immunitaire. De façon intéressante, dans des modèles de souris immunocompétentes porteuses de tumeurs murines, l'injection intra-tumorale de vaccin a un effet synergique avec des anti CTLA-4 permettant la guérison de 100% des souris. Les vaccins sont des produits pédiatriques et adultes de grade clinique. Par conséquent, des stratégies de vaccination in situ par injection intra-tumorale pourraient être rapidement mises en clinique / Immune checkpoint targeted therapies against PD-1, PD-L1 and CTLA-4 are currently revolutionizing cancer care. However, only a minority of patients develop objective responses with these treatments. Therefore, new therapeutic interventions are needed to increase the immunogenicity of tumors in order to overcome resistance to immune checkpoint blockade therapy. Oncolytic properties of common viruses can be exploited for the priming of anti-tumor immunity and such oncolytic viruses (OVs) are currently in intense clinical development in combination with immune checkpoint targeted therapies. We have found that commercially available virus vaccines do have oncolytic properties. These pediatric vaccine virus can directly kill cancer cells with features of immunogenic cell death. Moreover, it has pro-inflammatory properties and can activate the NF-Kb pathway in a toll-like receptor and IRF3 independent manner. These in vitro biological properties translate in vivo into anti-tumor activity. Intra-tumoral vaccine therapy has anti-tumor effects which are partly immune mediated. Interestingly, in immunocompetent murine pediatric tumor models, intra-tumoral injection overcome resistance and synergize with immune checkpoint targeted therapy. Vaccines are pediatric and adult clinical grade products. Therefore, in situ intra-tumoral immunization strategies could be implemented quickly in the clinic
70

Antagonism Between Trastuzumab and Oncolytic VSV is Overcome by Conjugation to a Microtubule Destabilizer

Garcia, Vanessa January 2015 (has links)
HER2overexpression is associated with poor breast cancer prognosis and increased risk of metastasis. Current HER2targeted therapies include monoclonal antibody based strategies which work by reducing HER2 levels at the cell surface (trastuzumab), by preventing HER2 dimerization (pertuzumab), or via targeted delivery of a cytotoxic payload (trastuzumab emtansine). Although these therapies are successful in some cases, acquired and inherent resistance to these therapeutics remain a treatment hurdle. Oncolytic viruses (OVs) specifically target and lyse cancer cells while leaving normal cells unharmed. One such OV, VSVΔ51, replicates in interferon (IFN) defective cells, a characteristic of approximately 70% of tumours. We hypothesized that the combination of HER2 targeting therapies with VSVΔ51 could improve therapeutic efficacy. We found that HER2 overexpression was associated with increased virus sensitivity and that modulation of HER2 signaling through a subset of activating ligands and inhibitory drugs could influence infection. We further established that the HER2 monoclonal antibodies trastuzumab and pertuzumab mediate an anti-viral effect on VSVΔ51 spread. Finally, we demonstrate that conjugation of a microtubule targeting agent to trastuzumab can overcome the induced anti-viral state and enhance VSVΔ51 spread specifically in cancer cells. Overall, this work highlights the importance of HER2 signaling and activation on VSVΔ51 spread and shows that conjugation of microtubule destabilizing agents to monoclonal antibodies can enhance VSVΔ51 efficacy.

Page generated in 0.0618 seconds