151 |
Etude de l’influence du stroma BRCA1 muté sur les étapes précoces de transformation tumorale dans le modèle du cancer du sein / Influence of BRCA1-mutated stroma on the early steps of the tumoral transformation in the breast cancer model.Portier, Lucie 13 December 2017 (has links)
L’objectif de ce travail a consisté à évaluer le rôle d’un microenvironnement avec une haplo-insuffisance hétérozygote du gène BRCA1 dans les événements précoces de la transformation tumorale du cancer du sein. Dans ce but, nous avons modélisé un stroma BRCA1-muté en utilisant des cellules souches / stromales mésenchymateuses (MSCs) obtenues par différenciation de cellules souches pluripotentes induites (iPSCs) issues d’une patiente porteuse de la mutation (MSCs BRCA1+/-). Ces cellules mutées pour BRCA1 ont été comparées à des MSCs sans la mutation (MSCs BRCA1+/+) générées à partir d’iPSCs BRCA1+/+. Ce travail de thèse a porté sur l’influence du stroma BRCA1-muté à travers deux axes : le caractère pro-angiogénique des MSCs BRCA1+/- et l’induction d’une transition épithélio-mésenchymateuse (TEM) sur des cellules mammaires normales (HME1).Nous montrons que les MSCs BRCA1-muté présentent des propriétés pro-angiogéniques significativement augmentées en surexprimant le facteur hypoxique HIF-1α et des facteurs de la famille du VEGF, PDGF et Angpt se traduisant par des capacités augmentées à former des structures vasculaires in vitro et in vivo. Les MSCs BRCA1-muté présentent également des capacités migratoires supérieures en produisant et sécrétant la périostine (POSTN), une protéine de la matrice extracellulaire impliquée dans l’adhésion, la motilité et la migration cellulaires. Ces capacités ont été validées par une approche de siRNA spécifique pour la POSTN. In vivo, nous montrons que la co-injection de MSCs BRCA1-muté et de cellules malignes mammaires murines (4T1-Luc-GFP) a permis d’augmenter significativement la croissance tumorale et la formation de métastases pulmonaires. Ces résultats sont corrélés avec la détection de la POSTN in situ et avec la formation d’un réseau vasculaire tumoral développé, quantifié par marquage du CD34. Par ailleurs nous avons démontré qu’un surnageant de MSCs BRCA1+/- peut induire une TEM des cellules HME1 en favorisant l’acquisition d’un phénotype souche cancéreux (CD24Low/CD44High) et en accélérant leur migration. Enfin nous avons initié la production in vitro d’organoïdes mammaires en utilisant des MSCs et des HME1 afin d’étudier plus précisément les mécanismes moléculaires de cette TEM après contact et des possibles événements précoces de la transformation maligne. Nos résultats indiquent que les MSCs peuvent participer à l’initiation tumorale et à la progression métastatique dans un contexte d’une mutation hétérozygote du gène BRCA1. La POSTN pourrait représenter à la fois un marqueur pronostique mais également une cible thérapeutique pour ces cancers du sein héréditaires. / The aim of this study was to evaluate the role of a BRCA1 heterozygous haplo-deficient microenvironment in the early events of tumour transformation of breast cancer. For this purpose we modeled a BRCA1-mutated stroma using mesenchymal stem / stromal cells (MSCs) obtained by differentiation of induced pluripotent stem cells (iPSCs) from a patient carrying the mutation (MSCs BRCA1+/-). These BRCA1-mutated cells were compared to MSCs without the mutation (MSCs BRCA1+/+) generated from iPSCs BRCA1+/+. This study focuses on two aspects of BRCA1-mutated stroma, namely the pro-angiogenic properties of BRCA1+/- MSCs and the induction of an epithelial-mesenchymal transition (EMT) on normal breast cells (HME1).We have shown that BRCA1-mutated MSCs exhibit enhanced pro-angiogenic properties by overexpressing the hypoxic factor HIF-1α and factors from VEGF, PDGF and Angpt families resulting in increased capacities to form vascular structures in vitro and in vivo. BRCA1-mutated MSCs exhibit also higher migratory capabilities by production and secretion of periostin (POSTN), an extracellular matrix protein, which is involved in cell adhesion, motility and migration. These capacities have been validated by a specific siRNA approach for POSTN. In vivo, the coinjection of BRCA1-mutated MSCs with murine breast cancer cell line (4T1-Luc-GFP) promotes tumour growth and the formation of lung metastases. These results are correlated with in situ POSTN detection and with the formation of a developed tumour vascular network, quantified by CD34 staining. We also demonstrated that supernatant of BRCA1+/- MSCs can induce an EMT on HME1 cells by promoting the acquisition of stemness properties (CD24Low/CD44High) and accelerating their migration. Finally we initiated the in vitro production of mammary organoids using MSCs and HME1 in order to study more precisely the molecular mechanisms of this EMT after contact and possible early events of the malignant transformation. These results indicate that MSCs can participate to tumour initiation and metastatic progression in heterozygous BRCA1-mutated background. POSTN could represent a prognostic marker and a therapeutic target for these hereditary breast cancers.
|
152 |
Immune context of malignant rhabdoid tumors : description and identification of new therapeutic targets / Contexte immunitaire des tumeurs rhabdoïdes : description et identification de nouvelles cibles thérapeutiquesLeruste, Amaury 11 February 2019 (has links)
Les tumeurs rhabdoïdes (TR) constituent un rare cancer indifférencié du jeune enfant et du nourrisson, avec un âge médian au diagnostic de 20 mois. Ces tumeurs sont caractérisées par une inactivation biallélique du gène suppresseur de tumeur SMARCB1, un des membres du complexe SWI/SNF, acteur majeur du remodelage de la chromatine, sans autre altération génomique récurrente. Le pronostic des TR est péjoratif, le taux de survie globale atteignant 30% dans la plupart des séries, malgré des approches thérapeutiques conventionnelles particulièrement agressives. Les approches d’immunothérapies ont obtenu un succès certain dans certains cancers de l’adulte, et récentes analyses de l’infiltrat immun des cancers pédiatriques ne montrent pas un fort taux de tumeurs infiltrées à l’exception de rare types de cancers dont les TR intracrâniennes. Nous avons donc procédé à une analyse multimodale de l’infiltrat immun de cohortes de patients ainsi que d’un modèle de TR murines établi dans notre laboratoire. Nous avons identifié une forte proportion de tumeurs infiltrées dans certains sous-groupes de TR. Cet infiltrat était composé à la fois de cellules myéloïdes incluant des populations au phénotype immunosuppresseur, et lymphocytaires T notamment de phénotype résident mémoire caractérisées par une forte expansion clonale probablement spécifique d’un antigène tumoral. Nous avons identifié des cibles thérapeutiques communes aux tumeurs humaines et au modèle murin syngénique, et trouvé que cibler l’infiltrat lymphocytaire T ou myéloïde était susceptible d’induire une réponse tumorale complète avec induction d’une mémoire immunitaire, confirmant le caractère immunogénique des TR, et apportant de nouvelles stratégies thérapeutiques utiles en clinique. Enfin, nous avons identifié que les TR étaient le site d’une réexpression de rétrovirus endogènes, dépendante de celle de SMARCB1, avec activation des voies de l’interféron, apportant une base à une immunogénicité des TR issue du génome non codant. / Rhabdoid tumors (RT) are highly undifferentiated cancers occurring in infancy and early childhood, with a median age at diagnosis about 20 months. These tumors are characterized by the biallelic inactivation of SMARCB1 tumor suppressor gene, core member of the SWI/SNF complex, one major chromatin remodeling actor, in an otherwise highly stable genome. The prognosis of RT is dismal with overall survival hardly reaching 30% in most series, despite particularly aggressive conventional treatment. Immunotherapy approaches has gained a striking success within some adult cancer types and recent analyses of immune cell content of pediatric cancers don’t reveal a high rate of infiltrated tumors, except in few tumor types such as intracranial rhabdoid tumors. Then, we conducted a comprehensive analysis of the immune context of both human RT cohorts and a mouse RT model, including at single cell level. We identified a high recurrence of infiltrated tumors, in a RT-subgroup related manner, composed of both myeloid cells including cells with immune suppressive phenotypes, and T cells with notably a tissue resident memory phenotype demonstrating a high clonal expansion highly suggestive of immunogenicity. We identified common targetable immune populations between human and mouse RTs, and found that targeting both T and myeloid infiltrating cells was able to induce complete anti-tumor response with induced memory, confirming the immunogenic properties of RTs, and identifying new therapeutic strategies of clinical relevance. We finally identified that RTs were the site of SMARCB1-dependent endogenous retroviruses reexpression, with subsequent activation of interferon signaling, likely triggering the immune response in the context of RT, and providing a basis of non-coding genome-driven immunogenicity for these tumors.
|
153 |
Dendritic Cells in Head and Neck Cancer Microenvironment : From Mechanisms to Biomarkers / Les cellules dendritiques dans le micro-environnement tumoral des cancers ORL : des mécanismes aux biomarqueursHoffmann, Caroline 08 October 2019 (has links)
L’objectif de ce travail était de comprendre l’état moléculaire des cellules dendritiques (CD) dans le microenvironnement tumoral. En intégrant l’analyse de tumeurs humaines par cytométrie en flux, de transcriptome, de secretome tumoral et l’analyse d’une base de données d’interaction CD-lymphocyte T générées in vitro, j’ai obtenus 2 résultats majeurs. Tout d’abord, nous proposons une nouvelle classification de CD activées humaines, qui sont soit « secrétantes », c’est-à-dire spécialisées dans la production de cytokines et chemokines, soit « aidantes » c’est-à-dire spécialisées dans l’induction de la sécrétion de nombreuses cytokines T helper après co-culture. Les CD infiltrant les tumeurs ORL inflammées correspondaient au type « sécrétantes ». Au-delà du nouveau concept biologique, cette classification est base théorique importante pour l’immunothérapie à base d’adjuvants. Deuxièmement, nous avons montré que l’inflammation tumorale n’était pas un facteur pronostic majeur des cancers ORL, mais que MMP2 et l’effraction extra-capsulaire étaient des facteurs pronostiques indépendants de la survie liée à la maladie. Nous avons pu classer les patients en 4 niveaux de risque et montré qu’ils avaient des chances équivalentes de réponse à l’immunothérapie. Nos données sont une base pour un essai clinique dirigé par biomarqueur, proposant de la chimiothérapie ou de l’immunothérapie néoadjuvantes, dans le but de diminuer le pourcentage de patients présentant des récidives sévères et précoces / The objective of the thesis was to decipher the molecular state of tumor infiltrating dendritic cell (DC) and their relation to the tumor microenvironment. By combining the analysis of human tumor samples by flow cytometry and RNA sequencing, of tumor secretome and of a large dataset of in vitro DC-Tcell interactions I obtained 2 main findings. First, we reported a novel classification of human activated DC, that are either “secretory” that is specialized in secreting cytokines and chemokines, or “helper” that is specialized at inducing the secretion of a broad range of T helper cytokines after cell co-culture. DC infiltrating inflamed human head and neck cancer matched the “secretory” phenotypic and transcriptomic signatures. Beyond this novel biological concept, this classification is of importance as a theoretical basis for adjuvant-based immunotherapy. Secondly, we showed that tumor inflammation was not the main prognostic factor for oral cavity cancer (OCC) patients, but that MMP2 and the presence of extra-nodal extension were independent predictors of reduced disease-specific survival. We could stratify OCC into 4 prognostic groups and showed that they had similar expected rates of response to immunotherapy. Our data may serve to design a biomarker-driven clinical trial proposing neoadjuvant chemotherapy or immunotherapy to high-risk patients, with the goal of reducing the percentage of OCC patients that will present with early and severe recurrences.
|
154 |
Role fibroblastů při hojení ran a rakovině / Role of fibroblasts in wound healing and cancerMateu Sanz, Rosana January 2021 (has links)
Fibroblasts are stromal cells ubiquitously present in the human body. They often appear in a quiescent state and can become activated in response to tissue remodeling signals. Activated fibroblasts acquire biosynthetic, pro-inflammatory and contractile properties, key functions for wound healing. In addition, the presence of permanently activated fibroblasts is one of the hallmarks of cancer. The purpose of this work is to investigate the differences between newborn and adult fibroblasts and keratinocytes in their implication in scarless wound healing, the origin of cancer associated fibroblasts (CAF)s and the influence of fibroblasts in melanoma invasion. Evidence suggests that wounds heal almost without scar in newborns. To understand the mechanisms that contribute to scarless wound healing we focused on the differences between newborn and adult fibroblasts and keratinocytes, which are cells present in human skin and participating in wound healing process. A comparison of the expression profile between newborn and adult fibroblasts showed differentially regulated genes related to the acute phase of the inflammatory response and ECM organization, traits involved in wound healing. We also found that newborn fibroblast showed higher differentiation potential, exhibited markers of pluripotency and...
|
155 |
Characterization of the immunomicroenvironment of glioblastoma : Optimization of an antibody panel for detection of glioblastoma TME cells and effect of C3 on glioma cells. / Karakterisering av immunomikromiljön i glioblastom. : Optimering av en antikroppspanel för detektion av celler i tumörmiljön av glioblastom samt effekt av C3 på gliomaceller.Wahldén, Julia January 2023 (has links)
Glioblastoma (GBM) is the most common primary brain tumor in adults and represents one of the most aggressive tumor types. Despite treatment GBM remains incurable and when treated the tumor recurs more aggressive and treatment resistant than before. The understanding of the underlying pathophysiology of GBM has increased in recent years, yet the high mortality with a 5-year survival rate of less than 10% remains. Previous studies from the research group, have shown that complement protein C3 is upregulated in stromal and glioma cells in lower oxygen tensions, and this indicates that C3 might play a role in certain GBM tumor microenvironment (TME) niches. For this reason, it is of great interest to investigate how glioma cells are affected when stimulated with C3. With cell proliferation-, migration assays and gene expression analysis with real time quantitative polymerase chain reaction (qPCR) it was possible to investigate how glioma cells respond to treatment with C3 and C3a-receptor antagonist. By optimizing an antibody panel for detection of macrophages/microglia, astrocytes and tumor cells we made it possible to study the modulation of immune parameters during treatment with C3A-receptor antagonist in mice. / Glioblastom (GBM) är den vanligaste primära hjärntumören hos vuxna och representerar en av de mest aggressiva tumörtyperna. Trots behandling förblir GBM obotlig och efter behandling återkommer tumören mer aggressiv och behandlingsresistent än tidigare. Förståelsen för den underliggande patofysiologin för GBM har ökat de senaste åren, trots detta kvarstår den höga dödligheten med en 5-årig överlevnadsprognos under 10%. Tidigare studier från forskningsgruppen har också visat att komplementprotein C3 är uppreglerat i gliomceller under lågt syretryck, vilket indikerar att C3 kan spela en roll i olika miljöer som finns i GBM. Av denna anledning är det av stort intresse att undersöka hur gliomceller påverkas när de stimuleras med C3. Med cellproliferation-, migrerings analyser och genuttrycksanalys med real time quantitative polymerase chain reaction (qPCR) var det möjligt att undersöka hur gliomceller, men även andra komponenter av tumor microenvironment (TME) svarar på behandling av C3 och C3A-receptor antagonist. Genom att optimera en antikroppspanel, med detektion av makrofager/ mikroglia, astrocyter och tumörceller har vi nu gjort det möjligt att studera moduleringen av immun parametrar vid behandling av C3A-receptor antagonist i möss.
|
156 |
Matrix Remodeling and Hyaluronan Production by Myofibroblasts and Cancer-Associated Fibroblasts in 3D Collagen MatricesSapudom, Jiranuwat, Damaris Müller, Claudia, Nguyen, Khiet-Tam, Martin, Steve, Anderegg, Ulf, Pompe, Tilo 13 April 2023 (has links)
The tumor microenvironment is a key modulator in cancer progression and has become a novel target in cancer therapy. An increase in hyaluronan (HA) accumulation and metabolism can be found in advancing tumor progression and are often associated with aggressive malignancy, drug resistance and poor prognosis. Wound-healing related myofibroblasts or activated cancer-associated fibroblasts (CAF) are assumed to be the major sources of HA. Both cell types are capable to synthesize new matrix components as well as reorganize the extracellular matrix. However, to which extent myofibroblasts and CAF perform these actions are still unclear. In this work, we investigated the matrix remodeling and HA production potential in normal human dermal fibroblasts (NHFB) and CAF in the absence and presence of transforming growth factor beta -1 (TGF-β1), with TGF-β1 being a major factor of regulating fibroblast differentiation. Three-dimensional (3D) collagen matrix was utilized to mimic the extracellular matrix of the tumor microenvironment. We found that CAF appeared to response insensitively towards TGF-β1 in terms of cell proliferation and matrix remodeling when compared to NHFB. In regards of HA production, we found that both cell types were capable to produce matrix bound HA, rather than a soluble counterpart, in response to TGF-β1. However, activated CAF demonstrated higher HA production when compared to myofibroblasts. The average molecular weight of produced HA was found in the range of 480 kDa for both cells. By analyzing gene expression of HA metabolizing enzymes, namely hyaluronan synthase (HAS1-3) and hyaluronidase (HYAL1-3) isoforms, we found expression of specific isoforms in dependence of TGF-β1 present in both cells. In addition, HAS2 and HYAL1 are highly expressed in CAF, which might contribute to a higher production and degradation of HA in CAF matrix. Overall, our results suggested a distinct behavior of NHFB and CAF in 3D collagen matrices in the presence of TGF-β1 in terms of matrix remodeling and HA production pointing to a specific impact on tumor modulation.
|
157 |
Ets2 and Pten regulate ErbB2-driven mammary tumorigenesis from stromal fibroblastsBalakrishnan, Subhasree 12 September 2016 (has links)
No description available.
|
158 |
An investigation of Atf3, an adaptive-response gene, in breast cancer chemotherapy and stress response.Jalgaonkar, Swati 01 September 2016 (has links)
No description available.
|
159 |
Toll-like Receptor 3 Signaling in Breast Cancer Cells and the Recruitment of Leukocytes to the Tumor MicroenvironmentVenkatesh, Amritha K. 26 July 2012 (has links)
No description available.
|
160 |
Inflammatory Tumor Microenvironment as target in the design of nanoconjugates for the treatment of advanced breast cancerSoriano Teruel, Paula 13 March 2023 (has links)
[ES] Esta tesis doctoral titulada "El microambiente tumoral inflamatorio como objetivo en el diseño de nanoconjugados para el tratamiento del cáncer de mama avanzado" se centra en la evaluación de un nuevo inhibidor del inflamasoma (MM01) como herramienta química para estudiar el papel del inflamasoma en modelos de inflamación y cáncer. El capítulo I incluye una descripción general del sistema inmune, los inflamasomas dependientes de ASC y el papel que juegan en el desarrollo de enfermedades. También se profundiza en el papel de los inflamasomas y de la proteína ASC en la progresión del cáncer de mama. Además, se incluyen conceptos básicos de nanotecnología, nanomedicina y polímeros terapéuticos. Finalmente, se abordan las ventajas de utilizar nanomedicinas como terapia, las interacciones de las nanomedicinas con los sistemas biológicos, los nanofármacos descritos en la literatura, así como sus posibilidades de traslación a la práctica clínica. En los capítulos de resultados, delineamos un novedoso mecanismo de acción para MM01, un modulador de la actividad del inflamasoma recientemente identificado: la inhibición de la oligomerización del ASC y el subsiguiente procesamiento reducido de la pro-caspasa-1 y la inhibición de la actividad de la caspasa-1. Demostramos que MM01 interrumpe el proceso de oligomerización de ASC asociado a la actividad de varios inflamasomas e inhibe la liberación de IL-1ß y la piroptosis en varios modelos celulares de inflamación. MM01 también reduce la infiltración de neutrófilos y la acumulación de citoquinas pro-inflamatorias en un modelo in vivo de peritonitis. Dada la implicación de la función de ASC en múltiples complejos del inflamasoma, el tratamiento con MM01 puede representar un enfoque terapéutico eficaz para tratar aquellas enfermedades en las que está implicada la activación de múltiples inflamasomas. Teniendo en cuenta los resultados obtenidos, empleamos nuestro inhibidor del inflamasoma, MM01, para estudiar el papel del inflamasoma en la progresión tumoral en diferentes modelos de cáncer de mama tanto in vitro como in vivo. Demostramos que diferentes líneas celulares de cáncer de mama responden de forma diferente al tratamiento con MM01. Desarrollamos un ensayo funcional que comprende la evaluación de la migración de las células de cáncer de mama en respuesta al secretoma pro-inflamatorio de los macrófagos M1 (estímulo inflamatorio) en presencia de MM01. Ciertas líneas celulares (como la línea celular EO771) mostraron un aumento de la migración en respuesta al estímulo inflamatorio y una disminución de la migración en respuesta al tratamiento con MM01; sin embargo, también identificamos líneas celulares que responden negativamente al tratamiento con MM01 (como la línea celular 4T1). Por último, demostramos la eficacia de este experimento funcional in vivo demostrando que el tratamiento con MM01 redujo el tamaño del tumor en el modelo ortotópico EO771 pero aumentó el tamaño del tumor y la metástasis pulmonar en el modelo ortotópico 4T1. Estos dos modelos, que recapitulan respuestas contradictorias al tratamiento con nuestro inhibidor del inflamasoma, podrán utilizarse en el futuro para determinar biomarcadores que predigan la respuesta. Por último, desarrollamos una estrategia sintética para obtener un nuevo nanomedicamento que mejora la solubilidad y la orientación tumoral del MM01 en un modelo de cáncer de mama. Implementamos un enfoque híbrido de conjugación-complejación que comprende la conjugación de ß-ciclodextrina con un ácido lineal poli-L-glutámico (PGA) (L-PGA-ßCD) para proporcionar la capacidad de atrapar MM01 dentro de los anillos de ciclodextrina. El nanosistema obtenido mostró una mejor solubilidad en soluciones acuosas en comparación con la forma libre de MM01. Nuestro nanosistema demostró una mejor eficacia en un modelo ortotópico de cáncer de mama al producir una mayor reducción del tamaño del tumor en aquellos ratones tratados con la nanomedicina L-PGA-CD-MM01. / [CA] Aquesta tesi doctoral titulada "El microambient tumoral inflamatori com a objectiu en el disseny de nanoconjugats per al tractament del càncer de mama avançat" se centra en l'avaluació d'un nou inhibidor del inflamasoma (MM01) com a eina química per a estudiar el paper del inflamasoma en models d'inflamació i càncer. El capítol I inclou una descripció general del sistema immune, els inflamasomas dependents de ASC i el paper que juguen en el desenvolupament de malalties. També s'aprofundeix en el paper dels inflamasomas i de la proteïna ASC en la progressió del càncer de mama. A més, s'inclouen conceptes bàsics de nanotecnologia, nanomedicina i polímers terapèutics. Finalment, s'aborden els avantatges d'utilitzar *nanomedicinas com a teràpia, les interaccions de les nanomedicines amb els sistemes biològics, els nanofármacs descrits en la literatura, així com les seues possibilitats de translació a la pràctica clínica. En els capítols de resultats, delineem un nou mecanisme d'acció per a MM01, un modulador de l'activitat del s*inflamasoma recentment identificat: la inhibició de la oligomerización del ASC i el subsegüent processament reduït de la pro-caspasa-1 i la inhibició de l'activitat de la caspasa-1. Vam demostrar que MM01 interromp el procés de oligomerización de ASC associat a l'activitat de diversos inflamasomas i inhibeix l'alliberament de IL-*1ß i la piroptosis en diversos models cel·lulars d'inflamació. MM01 també redueix la infiltració de neutròfils i l'acumulació de citocines pro-inflamatòries en un model in vivo de peritonitis. Donada la implicació de la funció de ASC en múltiples complexos del *inflamasoma, el tractament amb MM01 pot representar un enfocament terapèutic eficaç per a tractar aquelles malalties en les quals està implicada l'activació de múltiples *inflamasomas. Tenint en compte els resultats obtinguts, emprem el nostre inhibidor del *inflamasoma, MM01, per a estudiar el paper del *inflamasoma en la progressió tumoral en diferents models de càncer de mama tant in vitro com in vivo. Vam demostrar que diferents línies cel·lulars de càncer de mama responen de forma diferent del tractament amb MM01. Desenvolupem un assaig funcional que comprén l'avaluació de la migració de les cèl·lules de càncer de mama en resposta al secretoma pro-inflamatori dels macròfags M1 (estímul inflamatori) en presència de MM01. Unes certes línies cel·lulars (com la línia cel·lular EO771) van mostrar un augment de la migració en resposta a l'estímul inflamatori i una disminució de la migració en resposta al tractament amb MM01; no obstant això, també identifiquem línies cel·lulars que responen negativament al tractament amb MM01 (com la línia cel·lular 4T1). Finalment, vam demostrar l'eficàcia d'aquest experiment funcional in vivo demostrant que el tractament amb MM01 va reduir la grandària del tumor en el model ortotòpic EO771 però va augmentar la grandària del tumor i la metàstasi pulmonar en el model ortotòpic 4T1. Aquests dos models, que recapitulen respostes contradictòries al tractament amb el nostre inhibidor del *inflamasoma, podran utilitzar-se en el futur per a determinar biomarcadors que prediguen la resposta. Finalment, desenvolupem una estratègia sintètica per a obtindre un nou nanomedicament que millora la solubilitat i l'orientació tumoral del MM01 en un model de càncer de mama. Implementem un enfocament híbrid de conjugació-complexació que comprén la conjugació de ß-ciclodextrina amb un àcid lineal *poli-L-glutàmic (PGA) (L-PGA-ßCD) per a proporcionar la capacitat d'atrapar MM01 dins dels anells de ciclodextrina. El nanosistema obtingut va mostrar una millor solubilitat en solucions aquoses en comparació amb la forma lliure de MM01. El nostre nanosistema va demostrar una millor eficàcia en un model ortotòpic de càncer de mama en produir una major reducció de la grandària del tumor en aquells ratolins tractats amb la nanomedicina L-PGA-CD-MM01. / [EN] This PhD thesis entitled "The inflammatory tumor microenvironment as a target in the design of nanoconjugates for the treatment of advanced breast cancer" focuses on the evaluation of a novel inflammasome inhibitor (MM01) as a chemical tool to study the role of the inflammasome in models of inflammation and cancer. Chapter I includes an overview of the immune system, ASC-dependent inflammasomes and the role they play in disease development. It also delves into the role of inflammasomes and ASC protein in breast cancer progression. In addition, basic concepts of nanotechnology, nanomedicine and therapeutic polymers are included. Finally, the advantages of using nanomedicines as therapeutics, the interactions of nanomedicines with biological systems, the nanodrugs described in the literature, as well as their translation possibilities to clinical practice are discussed. In the results chapters, we delineate a novel mechanism of action for MM01, a recently identified modulator of inflammasome activity: inhibition of ASC oligomerization and subsequent reduced pro-caspase-1 processing and inhibition of caspase-1 activity. We demonstrate that MM01 disrupts the ASC oligomerization process associated with the activity of several inflammasomes and inhibits IL-1ß release and pyroptosis in several cellular models of inflammation. MM01 also reduces neutrophil infiltration and pro-inflammatory cytokine accumulation in an in vivo model of peritonitis. Given the involvement of ASC function in multiple inflammasome complexes, treatment with MM01 may represent an effective therapeutic approach to treat those diseases in which multiple inflammasome activation is involved. Considering the results obtained, we employed our inflammasome inhibitor, MM01, to study the role of the inflammasome in tumor progression in different breast cancer models both in vitro and in vivo. We demonstrate that different breast cancer cell lines respond differently to MM01 treatment. We developed a functional assay involving the assessment of breast cancer cell migration in response to the pro-inflammatory M1 macrophage secretome (inflammasome stimulus) in the presence of MM01. Certain cell lines (such as the EO771 cell line) showed increased migration in response to the inflammatory stimulus and decreased migration in response to MM01 treatment; however, we also identified cell lines that respond negatively to MM01 treatment (such as the 4T1 cell line). Finally, we demonstrated the efficacy of this functional experiment in vivo by showing that MM01 treatment reduced tumor size in the orthotopic EO771 model but increased tumor size and lung metastasis in the orthotopic 4T1 model. These two models, which recapitulate conflicting responses to treatment with our inflammasome inhibitor, may be used in the future to determine biomarkers predictive of response. Finally, we developed a synthetic strategy to obtain a novel nanomedicine that enhances the solubility and tumor targeting of MM01 in a breast cancer model. We implemented a hybrid conjugation-complexation approach comprising the conjugation of ß-cyclodextrin to a linear poly-L-glutamic acid (PGA) (L-PGA-ßCD) to provide the ability to trap MM01 within the cyclodextrin rings. The obtained nanosystem showed improved solubility in aqueous solutions compared to the free form of MM01. Our nanosystem demonstrated improved efficacy in an orthotopic breast cancer model by producing a greater reduction in tumor size in those mice treated with the L-PGA-CD-MM01 nanomedicine. / Soriano Teruel, P. (2023). Inflammatory Tumor Microenvironment as target in the design of nanoconjugates for the treatment of advanced breast cancer [Tesis doctoral]. Universitat Politècnica de València. https://doi.org/10.4995/Thesis/10251/192498
|
Page generated in 0.1105 seconds