• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 9
  • 1
  • Tagged with
  • 9
  • 9
  • 4
  • 4
  • 3
  • 3
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 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

Type I Interferon-Mediated Killing of Cancer Cells with IAP-Targeted Combination Immunotherapy

Beauregard, Caroline January 2016 (has links)
SMAC mimetic compounds (SMCs) are small molecule antagonists of the Inhibitor of Apoptosis (IAP) family of proteins. Binding of SMCs to the IAPs results in the sensitization of cancer cells to apoptosis in the presence of death ligands, such as tumour necrosis factor alpha (TNFα). I hypothesize that type I interferon (IFN) stimulation in cancer cells and in immune cells leads to the production of TNFα, which can then synergize with SMCs to kill cancer cells. The combined treatment of SMC and IFNα induces tumour regression in mice, and this effect is completely abrogated upon treatment with TNFα-neutralizing antibody. The synergistic effects are mediated by tumour cells and by contribution of immune cells, particularly macrophages and dendritic cells, as the systemic depletion of phagocytic innate immune cells results in an increase in tumour volume following combination treatment. The characterization of immune cell contribution will aid in the translation of the SMC combination therapy into clinical applications for the treatment of cancer.
2

Immunomodulatory Effects of Inhibitor of Apoptosis (IAP) Antagonists on Dendritic Cells

Labelle, Madeline Jones 06 December 2023 (has links)
The Inhibitor of Apoptosis (IAP) proteins are a highly conserved group of anti-apoptotic proteins that regulate various pathways, particularly those that affect proliferation and cell death. Smac mimetics compounds (SMCs) are IAP antagonists that induce the degradation of two IAPs, cellular IAP 1 (cIAP1) and cellular IAP 2 (cIAP2). cIAP1 and cIAP2 are negative regulators of the alternative NF-κB pathway, which is critical to the regulation, activation, proliferation, and survival of immune cells. Consequently, SMCs can affect immunological responses by providing co-stimulatory signals for antigen-presenting cells or promoting proliferation and activation of T cells. Due to their potent immunomodulatory properties, SMCs are an ideal candidate for new vaccine adjuvants. I sought to demonstrate the potential of SMCs as a vaccine adjuvant by evaluating SMCs effects on dendritic cells (DCs). I demonstrated that SMC treatment of bone marrow derived dendritic cells (BMDCs) induces degradation of both cIAP1 and cIAP2 and leads to activation of the alternative NF-κB signalling pathway. Furthermore, SMC treatment led to upregulation of proteins associated with DC maturation, as well as secretion of pro-inflammatory cytokines. Despite the activating effects elicited by SMCs in vitro, the use of SMCs as an adjuvant for peptide vaccination failed to prevent tumour growth. Further work to determine the best use of SMCs as adjuvants in vivo needs to be done to explore the potential of this class of drugs. Thus, these findings will guide the use of SMCs in adjuvant vaccine therapies for robust protective immunity.
3

Engineering Novel TNFα-armed Oncolytic Viruses for Combination Immunotherapy with SMAC Mimetics

Pichette, Stephanie January 2016 (has links)
Small molecular Inhibitor of Apoptosis (IAP) antagonists, known as Smac mimetic compounds (SMCs), are a novel class of anti-cancer drugs currently undergoing clinical trials. SMCs were designed to mimic the function of the pro-apoptotic protein, Smac, which directly depletes cells of cIAP1 and cIAP2, and consequently renders tumour cells sensitive to death in the presence of proinflammatory ligands such as TNFα. The Korneluk lab recently reported that SMCs synergize with the attenuated oncolytic virus Vesicular stomatitis virus (VSVΔ51) by eliciting an enhanced immune response in mice, such that the combined therapy is vastly superior to stand-alone therapies. To improve on this SMC-mediated synergistic response, I generated variants of TNFα-armed VSVΔ51. Due to high ectopic expression of TNFα in infected cells, a five times lower viral dose of TNFα-armed VSVΔ51 combined with SMC treatment was sufficient to improve the survival rate as compared to SMC and VSVΔ51 co-therapy. This improved synergistic response is attributed to a bystander effect whereby the spread of TNFα from infected cells leads to the death of neighbouring, uninfected cells in the presence of a SMC. In addition, the double treatment induced vasculature collapse in solid tumours, revealing another mechanism by which cytokine-armed VSVΔ51 in combination with a SMC can induce cancer cell death. This approach demonstrates great potential for engineered oncolytic virus and SMCs as a new combination immunotherapy for cancer treatment.
4

Regulation of the Cellular Inhibitor of Apoptosis 1 (cIAP1) Translation by IRES Trans-Acting Factors and Impact on Cancer

Faye, Mame Daro January 2015 (has links)
Apoptosis is the mechanism by which complex multicellular organisms induce the programmed death of damaged cells, thus maintaining tissue homeostasis. One of the main hallmarks of cancer, apoptosis is tightly regulated by pro- and anti-apoptotic factors whose equilibrium will decide of the fate of the cell. Among these factors, the cellular inhibitor of apoptosis cIAP1 is a key regulator of nuclear factor-κB dependent signaling and of caspase-8 mediated apoptosis. cIAP1 expression is controlled primarily at the translational level through an internal ribosome entry site (IRES) that facilitates the recruitment of the ribosome to the translation initiation start independently of the 5’ cap. We have previously identified four putative IRES trans-acting factors (ITAFs) that bind specifically to the cIAP1 IRES, namely NF45, NF90, IGF2BP1 and RH1. My research project characterised NF45 as an ITAF that positively regulates the IRES-mediated translation of cIAP1 and of the Xlinked inhibitor of apoptosis, XIAP. This regulation is important for maintaining Survivin and Cyclin E protein levels and insuring proper cell division. Furthermore, I showed that IGF2BP1 is another ITAF that is overexpressed in rhabdomyosarcoma cancer (RMS) and positively regulates cIAP1 translation, thus leading to apoptotic resistance in these cells. Importantly, the use of Smac mimetics, chemical compounds that cause cIAP1 proteasomal degradation, induces TNFα-mediated apoptosis of RMS cells and leads to growth inhibition of RMS xenograft tumors as well as significantly improved survival. Finally, I show that certain modulators of innate immunity synergize with Smac mimetics to improve the killing of RMS cancer cells. Hence, cIAP1 translation regulation by NF45 and IGF2BP1 is highly important for maintaining proper functioning of the cell and dysregulation of these ITAFs can lead to carcinogenesis.
5

Phenotypical and Functional Characterization of Polarized Human Macrophages

Iqbal, Salma January 2015 (has links)
Macrophages can be polarized into M1 and M2 macrophages based on the composition of the milieu. Human macrophages have been poorly characterized. In this study, various macrophage subsets were generated by treating monocyte-derived macrophages (MDMs) with IFNγ (M1), IL-4 (M2a), LPS and IL-1β (M2b) or IL-10 (M2c) which were characterized with respect to their cell surface marker profile and functional profile in the context of cytokine production, susceptibility to HIV infection and apoptosis. Each polarization state demonstrated a unique cell surface marker profile and cytokine profile. In addition M1 macrophages were shown to produce IFNγ post TLR stimulation. Moreover, M1 macrophages were highly sensitive to apoptosis following Smac mimetic treatment. Furthermore, M2a and M2c macrophages were resistant to apoptosis, induced by PI3K blockage and IAPs degradation respectively, and at the same time supported productive HIV infection unlike the other macrophage subsets. These findings might lead to better understanding of HIV reservoir formation and be used to develop therapies to eradicate it.
6

Selective Induction of Cell Death by Smac Mimetics in Primary Human Proinflammatory and Anti-inflammatory Macrophage Subsets

Ali, Hamza 23 February 2021 (has links)
The inflammatory and anti-inflammatory macrophages have been implicated in many diseases including rheumatoid arthritis, inflammatory bowel disease and chronic rhinosinusitis. Recent studies suggest targeting macrophage function and activation may represent a potential target to treat these diseases. Herein, I investigated the effect of second mitochondria-derived activator of caspases (SMAC) mimetics (SMs), the inhibitors of apoptosis (IAPs) proteins, on the killing of normal human pro- and anti-inflammatory macrophage subsets. It has been shown that human monocytes are highly susceptible to the cytotoxic effects of SMs, however, differentiated macrophages (M0) develop resistance to the cytocidal abilities of SMs. Whether human macrophage subsets are also resistant to the cytotoxic effects of SM remains unknown. My results show that differentiation of M0 macrophages towards M1 state rendered them highly susceptible to SM-induced cell death, whereas M2a, M2b and M2c differentiated subsets were resistant, with M2c being the most resistant. SM-induced cell death in M1 macrophages was mediated by apoptosis as well as necroptosis and activated both extrinsic and intrinsic pathways of apoptosis. The susceptibility of M1 macrophages to SM-induced cell death was attributed to the IFN-𝛾-mediated polarization as JAK inhibitor reversed their susceptibility. In contrast, M2c and M0 macrophages experienced cell death through necroptosis pathway following simultaneous blockage of the IAPs pathways by SM-LCL161 and the caspase pathways by the pan-caspase inhibitors (zVAD.fmk). I investigated the molecular mechanism governing SM-induced cell death in M1 macrophages. My results show that in contrast to the cancer cell lines, SM-induced cell death in M1 macrophages is independent of endogenously produced TNF-⍺, the canonical and non- canonical NF-𝜅B pathways. The susceptibility of M1 macrophages to SM-induced cell death was found to be dependent on IFN-𝛾-mediated differentiation through the JAK-STAT pathway and subsequent activation of IRF-1. In addition, the selective cell death in SM-treated M1 macrophages is mediated by simultaneous degradation of cellular IAP-2 (cIAP-2) and RIPK-1/3 through the activation of mTORC signaling pathway. Overall, the results suggest that survival of human macrophages is critically linked to the activation of the IAPs pathways. Moreover, agents blocking cIAP-1/2, mTORC and IRF-1 can be exploited therapeutically to address inflammation-related diseases. These observations hold a promising therapeutic strategy to limit the activation of proinflammatory M1 macrophages and eventually controlling the M1-associated diseases.
7

Augmentation of anti-myeloma engineered T cells by pharmacological or genetic interventions / Augmentation of anti-myeloma T cells

Afsahi, Arya January 2023 (has links)
Multiple myeloma is an aggressive plasma cell cancer that consistently acquires multi-drug resistance and relapses despite initial treatment successes. Patients may go through greater than 10-lines of therapy, highlighting the need for more effective treatment options. Immunotherapies are the latest evolution in targeted cancer treatments, and thus far have displayed impressive results in several hematological cancers, including multiple myeloma. T cells possess robust anti-tumor functions which can be harnessed and refined for the treatment of cancers. Genetic engineering of T cells to express a chimeric antigen receptor (CAR) confers antigen-specific tumor-targeting, and adoptive transfer of patient-derived CAR-engineered T (CAR T) cells has been efficacious in relapsed/refractory multiple myeloma. Despite the high efficacy, CAR T cell therapy for myeloma is associated with serious adverse events, which limits dose levels and patient eligibility. We have developed a novel synthetic antigen receptor platform, called the T cell antigen coupler (TAC) receptor, which has shown comparatively higher efficacy with a reduced pro-inflammatory profile compared with CAR T cells in pre-clinical models. The TAC receptor was purpose-built to co-opt the natural T cell activation machinery and lacks the costimulatory signaling typically incorporated in CAR designs. This thesis investigates strategies to augment TAC T cell function against for multiple myeloma through the evaluation of ancillary pharmacological and protein stimuli that would complement the anti-tumor functions of TAC T cells without modifying the TAC receptor design. In chapter 2, I investigated a strategy combining TAC T cells with the SMAC mimetic LCL161 to provide transient costimulatory effects. While LCL161 boosted TAC T cells survival and proliferation, the drug also enhanced susceptibility of TAC T cells to apoptosis and offered no advantage to the TAC T cells when challenged with myeloma. In chapter 3, I engineered TAC T cells to secrete IL-27 in an attempt to modulate the myeloma microenvironment and support T cell cytolytic function. IL-27 did not enhance the anti-tumor activity of TAC T cells but forced expression of IL-27 led to a reduction in the production of pro-inflammatory cytokines without altering cytotoxicity. In appendix I, I describe the process of optimizing CRISPR/Cas9 editing of primary TAC T cells. This methodology was required for much of the work in chapter 2. Ph.D. Thesis – Arya Afsahi McMaster University – Biochemistry and Biomedical Sciences v In appendix II, I describe an assessment of mRNA-engineering as a method to produce TAC T cells. This approach proved to be therapeutically futile and was not pursued beyond the work described herein. The work presented here highlights methods of combining TAC T cells with a clinically relevant SMAC mimetic, or the cytokine IL-27, and provides insights into the biological mechanisms that are affected by these approaches. / Thesis / Doctor of Philosophy (PhD)
8

Les inhibiteurs de l'apoptose, une nouvelle cible thérapeutique dans les glioblastomes / Inhibitor of apoptosis proteins, a new therapeutic target in glioblastomas

Souberan, Aurélie 15 December 2017 (has links)
Les glioblastomes (GBs) sont les tumeurs primitives du SNC les plus agressives de l’adulte. Les causes d’échec thérapeutiques sont multiples, comme une résistance des cellules tumorales à l’apoptose, l’existence de cellules souches cancéreuses ou un microenvironnement pro-tumoral. La découverte de molécules thérapeutiques qui pourraient avoir une action pléiotrope est particulièrement attractive. Dans ce contexte nous nous sommes intéressés aux mimétiques de Smac (MS), antagonistes des inhibiteurs de l’apoptose (IAP), qui inhibent le plus souvent cIAP1, cIAP2, XIAP et ML-IAP. Nous avons recherché si les IAP pouvaient être des cibles thérapeutiques dans les GBs humains en étudiant leur expression et leurs valeurs pronostiques éventuelles : les IAP sont exprimés dans les GBs et ML-IAP est associé à un plus mauvais pronostic. Nous avons choisi d’utiliser pour la suite de nos expériences, un MS qui avait une action sur les IAP et en particulier ML-IAP : le GDC-0152. Le GDC-0152 induit l’apoptose in vitro, augmente la survie des souris porteuses de GB et ralentit la croissance tumorale in vivo. Ensuite nous avons recherché si l’effet du GDC-0152 pouvait être différent en fonction du taux d'oxygène. En effet, les GBs sont des tumeurs hypoxiques. Nous avons cultivé en normoxie et en hypoxie, quatre lignées de cellules souches de GBs. En normoxie, le GDC-0152 induit la différenciation des cellules souches (voie NF-κB) et en hypoxie il induit l’apoptose et diminue la prolifération cellulaire (voie ATR).Ces travaux soulignent l’importance du modèle préclinique utilisé dans la caractérisation de l'effet de nouvelles molécules et le potentiel thérapeutique des MS dans les GBs. / Glioblastomas (GBs) are the most aggressive primary brain tumors in adults. The causes of therapeutic failure are unknown and are multiples, such as tumor cell resistance to apoptosis, the presence of cancer stem cells or a pro-tumor microenvironment. Thus, the discovery of therapeutic molecules with pleiotropic action is particularly interesting. In this context, we are interested in smac mimetics (SM), antagonists of inhibitor of apoptosis proteins (IAPs) and most often antagonize cIAP1, cIAP2, XIAP and ML-IAP.We investigated whether IAPs could be attractive therapeutic targets in human GBs by studying their expression and their possible prognostic values. All IAPs were expressed in various degrees in GBs and ML-IAP was associated with a worse prognosis. Therefore, we chose GDC-0152 for the rest of our experiments because it antagonizes the different IAPs and in particular ML-IAP. We showed that GDC-0152 induces apoptosis in vitro, increases the survival of GB-bearing mice and slows tumor growth in vivo.We investigated whether the effect of GDC-0152 could be different depending on the oxygen level. Indeed, GBs are part of the most hypoxic tumors. For this purpose, four GB stem cell lines were grown in normoxia and hypoxia. We found that GDC-0152 has an anti-tumor effect regardless of oxygen level, but the signaling pathways involved were different. In normoxia, GDC-0152 induces differentiation of GB stem cells (NF-κB pathway) and in hypoxia it induces apoptosis and decreases cell proliferation (ATR pathway).This work highlights the importance of the preclinical model used in the characterization of a new molecule effects and the therapeutic potential of SM in GBs.
9

Conditional Activation and Synergistic Enhancement of Smac Mimetic Peptides with Nucleic Acids

Altrichter, Yannic 14 March 2022 (has links)
Smac-Mimetika (SMCs) sind eine Klasse von Tetrapeptid-abgeleiteten Medikamenten, die dem homodimeren, pro-apoptotischen Protein Smac nachempfunden sind. Sie binden und hemmen die Apoptose-Inhibitoren (IAPs), eine Klasse von anti-apoptotischen Proteinen, die von vielen medikamentenresistenten Krebszellen überexprimiert werden. In dieser Arbeit wurden Strategien zur Steigerung und Kontrolle der Aktivität von SMCs durch Konjugation an Oligonukleotide (ON) untersucht. ONs wie Desoxyribonukleinsäure (DNA) oder das peptidbasierte Analogon Peptidnukleinsäure (PNA) bieten einzigartige Erkennungseigenschaften, die zur Detektion und/oder zur Modulation der Expression krebsspezifischer Genprodukte genutzt werden können. Letzteres kann z. B. mit Antisense-Oligonukleotiden (ASOs) erreicht werden, kurzen einzelsträngigen DNA- oder RNA-Molekülen, die die Translation einer komplementären Zielsequenz blockieren. Im ersten Teil der Arbeit wurden Konjugate aus SMCs und ASOs auf Synergie-Effekte getestet. Viele menschliche Krebszelllinien sind gegen SMCs resistent, weil andere anti-apoptotische Proteine wie das zelluläre FLICE-ähnliche Protein (c-FLIP) als Ausfallsicherung wirken. Es konnte gezeigt werden, dass diese Resistenz durch Kupplung eines SMC an ein anti-c-FLIP ASO überwunden werden kann. Im zweiten Teil wurden kurze ON-Sonden verwendet, um ein templiertes Reaktionssystem zur gezielten Aktivierung eines SMCs in Gegenwart von X-linked Apoptose-Inhibitor (XIAP) mRNA zu erzeugen. Es wurden zwei verschiedene Ansätze untersucht: Ein templierter Acyl-Transfer, bei dem hochaffine, bivalente SMCs aus niedrig affinen, monovalenten Vorläufern erzeugt werden und eine Demaskierung der für die Bindungsaffinität von SMCs entscheidenden, N-terminalen Aminogruppe durch templierte Reduktion eines Azids. Für die zweite Strategie wurden zwei verschiedene Reaktionen verglichen, die Staudinger-Reduktion mit Phosphinen und eine katalytische Photoreduktion mit einem Ruthenium-Komplex. / Smac mimetic compounds (SMCs) are a class of tetrapeptide-derived drugs, modelled after the homodimeric, pro-apoptotic protein Smac. They bind and antagonize Inhibitor of Apoptosis Proteins (IAPs), a class of anti-apoptotic proteins overexpressed by many drug-resistant cancer cells. In this work, strategies to enhance and control the activity of SMCs by conjugating them to oligo-nucleotides (ON) were investigated. ONs like deoxyribonucleic acid (DNA) or the peptide-based analog peptide nucleic acid (PNA) offer unique recognition properties that can be used to detect and/or modulate the expression of cancer-specific gene products. The latter can be achieved with antisense oligonucleotides (ASOs), short single-stranded DNA or RNA molecules that block the translation of a complementary sequence of interest. In the first part of this work, conjugates between SMCs and ASOs were tested for synergy. Many human cancer cell lines are resistant to SMCs because other anti-apoptotic proteins like the cellular FLICE-like protein (c-FLIP) act as a failsafe. It could be demonstrated that by joining an SMC with an anti-c-FLIP ASO this resistance can be overcome. In the second part, short ON probes were used to create a templated reaction system to conditionally activate an SMC in the presence of x-linked inhibitor of apoptosis (XIAP) mRNA. Two different approaches were explored: A templated acyl transfer that yields high affinity bivalent SMCs from low affinity, monovalent precursors and unmasking of the N-terminal amino group, which is crucial for the binding affinity of SMCs, by templated reduction of an azide. For the second strategy, two different chemistries were compared, Staudinger reduction with phosphines and a catalytic photoreduction using a ruthenium complex.

Page generated in 0.0427 seconds