Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres











Gamme d'année
1.
Proc Natl Acad Sci U S A ; 119(19): e2123483119, 2022 05 10.
Article de Anglais | MEDLINE | ID: mdl-35507878

RÉSUMÉ

Immunotherapy approaches focusing on T cells have provided breakthroughs in treating solid tumors. However, there remains an opportunity to drive anticancer immune responses via other cell types, particularly myeloid cells. ATRC-101 was identified via a target-agnostic process evaluating antibodies produced by the plasmablast population of B cells in a patient with non-small cell lung cancer experiencing an antitumor immune response during treatment with checkpoint inhibitor therapy. Here, we describe the target, antitumor activity in preclinical models, and data supporting a mechanism of action of ATRC-101. Immunohistochemistry studies demonstrated tumor-selective binding of ATRC-101 to multiple nonautologous tumor tissues. In biochemical analyses, ATRC-101 appears to target an extracellular, tumor-specific ribonucleoprotein (RNP) complex. In syngeneic murine models, ATRC-101 demonstrated robust antitumor activity and evidence of immune memory following rechallenge of cured mice with fresh tumor cells. ATRC-101 increased the relative abundance of conventional dendritic cell (cDC) type 1 cells in the blood within 24 h of dosing, increased CD8+ T cells and natural killer cells in blood and tumor over time, decreased cDC type 2 cells in the blood, and decreased monocytic myeloid-derived suppressor cells in the tumor. Cellular stress, including that induced by chemotherapy, increased the amount of ATRC-101 target in tumor cells, and ATRC-101 combined with doxorubicin enhanced efficacy compared with either agent alone. Taken together, these data demonstrate that ATRC-101 drives tumor destruction in preclinical models by targeting a tumor-specific RNP complex leading to activation of innate and adaptive immune responses.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Tumeurs , Immunité acquise , Animaux , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Humains , Immunité innée , Souris , Tumeurs/anatomopathologie
2.
Clin Immunol ; 187: 37-45, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-29031828

RÉSUMÉ

There is significant debate regarding whether B cells and their antibodies contribute to effective anti-cancer immune responses. Here we show that patients with metastatic but non-progressing melanoma, lung adenocarcinoma, or renal cell carcinoma exhibited increased levels of blood plasmablasts. We used a cell-barcoding technology to sequence their plasmablast antibody repertoires, revealing clonal families of affinity matured B cells that exhibit progressive class switching and persistence over time. Anti-CTLA4 and other treatments were associated with further increases in somatic hypermutation and clonal family size. Recombinant antibodies from clonal families bound non-autologous tumor tissue and cell lines, and families possessing immunoglobulin paratope sequence motifs shared across patients exhibited increased rates of binding. We identified antibodies that caused regression of, and durable immunity toward, heterologous syngeneic tumors in mice. Our findings demonstrate convergent functional anti-tumor antibody responses targeting public tumor antigens, and provide an approach to identify antibodies with diagnostic or therapeutic utility.


Sujet(s)
Antigènes néoplasiques/immunologie , Lymphocytes B/immunologie , Tumeurs/immunologie , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/secondaire , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps , Sites de fixation des anticorps/immunologie , Néphrocarcinome/immunologie , Néphrocarcinome/secondaire , Évolution de la maladie , Femelle , Humains , Tumeurs du rein/immunologie , Tumeurs du rein/anatomopathologie , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Mâle , Mélanome/immunologie , Mélanome/secondaire , Adulte d'âge moyen , Métastase tumorale , Plasmocytes/immunologie , Précurseurs lymphoïdes B , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie
3.
Br J Cancer ; 116(4): 515-523, 2017 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-28118322

RÉSUMÉ

BACKGROUND: Screening of patients for cancer-driving mutations is now used for cancer prognosis, remission scoring and treatment selection. Although recently emerged targeted next-generation sequencing-based approaches offer promising diagnostic capabilities, there are still limitations. There is a pressing clinical need for a well-validated, rapid, cost-effective mutation profiling system in patient specimens. Given their speed and cost-effectiveness, quantitative PCR mutation detection techniques are well suited for the clinical environment. The qBiomarker mutation PCR array has high sensitivity and shorter turnaround times compared with other methods. However, a direct comparison with existing viable alternatives are required to assess its true potential and limitations. METHODS: In this study, we evaluated a panel of 117 patient-derived tumour xenografts by the qBiomarker array and compared with other methods for mutation detection, including Ion AmpliSeq sequencing, whole-exome sequencing and droplet digital PCR. RESULTS: Our broad analysis demonstrates that the qBiomarker's performance is on par with that of other labour-intensive and expensive methods of cancer mutation detection of frequently altered cancer-associated genes, and provides a foundation for supporting its consideration as an option for molecular diagnostics. CONCLUSIONS: This large-scale direct comparison and validation of currently available mutation detection approaches is extremely relevant for the current scenario of precision medicine and will lead to informed choice of screening methodologies, especially in lower budget conditions or time frame limitations.


Sujet(s)
Analyse de mutations d'ADN/méthodes , Hétérogreffes , Tumeurs/génétique , Animaux , Hétérogreffes/métabolisme , Séquençage nucléotidique à haut débit/méthodes , Humains , Souris , Transplantation tumorale , Tumeurs/anatomopathologie , Réaction de polymérisation en chaîne/méthodes , Reproductibilité des résultats , Cellules cancéreuses en culture
4.
Cancer Biol Ther ; 16(8): 1184-93, 2015.
Article de Anglais | MEDLINE | ID: mdl-26046946

RÉSUMÉ

Triple negative breast cancer has an extremely poor prognosis when chemotherapy is no longer effective. To overcome drug resistance, novel drug delivery systems based on nanoparticles have had remarkable success. We produced a novel nanoparticle component 'MDC' from milk-derived colloid. In order to evaluate the anti-cancer effect of MDC, we conducted in vitro and in vivo experiments on cancer cell lines and a primary tumor derived breast xenograft. Doxorubicin (Dox) conjugated to MDC (MDC-Dox) showed higher cancer cell growth inhibition than MDC alone especially in cell lines with high EGFR expression. In a mouse melanoma model, MDC-Dox significantly suppressed tumor growth when compared with free Dox. Moreover, in a primary tumor derived breast xenograft, one of the mice treated with MDC-Dox showed partial regression, while mice treated with free Dox failed to show any suppression of tumor growth. We have shown that a novel nanoparticle compound made of simple milk-derived colloid has the capability for drug conjugation, and serves as a tumor-specific carrier of anti-cancer drugs. Further research on its safety and ability to carry various anti-cancer drugs into multiple drug-resistant primary breast models is warranted.


Sujet(s)
Colloïdes/composition chimique , Doxorubicine/administration et posologie , Vecteurs de médicaments/composition chimique , Lait/composition chimique , Tumeurs du sein triple-négatives/traitement médicamenteux , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Colloïdes/administration et posologie , Doxorubicine/composition chimique , Vecteurs de médicaments/administration et posologie , Systèmes de délivrance de médicaments/méthodes , Femelle , Humains , Sujet immunodéprimé , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Souris de lignée C57BL , Nanoparticules/composition chimique , Tumeurs du sein triple-négatives/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Oncotarget ; 4(10): 1737-47, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-24077805

RÉSUMÉ

Somatic mutations in Isocitrate Dehydrogenase 1 (IDH1) are frequent in low grade and progressive gliomas and are characterized by the production of 2-hydroxyglutarate (2-HG) from α-ketoglutarate by the mutant enzyme. 2-HG is an "oncometabolite" that competitively inhibits α-KG dependent dioxygenases resulting in various widespread cellular changes including abnormal hypermethylation of genomic DNA and suppression of cellular differentiation. Despite the growing understanding of IDH mutant gliomas, the development of effective therapies has proved challenging in part due to the scarcity of endogenous mutant in vivo models. Here we report the generation of an endogenous IDH1 anaplastic astrocytoma model which rapidly grows in vivo, produces 2-HG and exhibits DNA hypermethylation. Using this model, we have demonstrated the preclinical efficacy and mechanism of action of the FDA approved demethylating drug 5-azacytidine in vivo. Long term administration of 5-azacytidine resulted in reduction of DNA methylation of promoter loci, induction of glial differentiation, reduction of cell proliferation and a significant reduction in tumor growth. Tumor regression was observed at 14 weeks and subsequently showed no signs of re-growth at 7 weeks despite discontinuation of therapy. These results have implications for clinical trials of demethylating agents for patients with IDH mutated gliomas.


Sujet(s)
Antimétabolites antinéoplasiques/pharmacologie , Azacitidine/pharmacologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/enzymologie , Gliome/traitement médicamenteux , Gliome/enzymologie , Isocitrate dehydrogenases/génétique , Animaux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Processus de croissance cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Femelle , Gliome/génétique , Gliome/anatomopathologie , Humains , Immunohistochimie , Isocitrate dehydrogenases/métabolisme , Mâle , Souris , Souris nude , Mutation , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Transl Oncol ; 6(2): 197-205, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23544172

RÉSUMÉ

Mutually exclusive genetic alterations in the RET, RAS, or BRAF genes, which result in constitutively active mitogen-activated protein kinase (MAPK) signaling, are present in about 70% of papillary thyroid carcinomas (PTCs). However, the effect of MAPK activation on other signaling pathways involved in oncogenic transformation, such as Notch, remains unclear. In this study, we tested the hypothesis that the MAPK pathway regulates Notch signaling and that Notch signaling plays a role in PTC cell proliferation. Conditional induction of MAPK signaling oncogenes RET/PTC3 or BRAF(T1799A) in normal rat thyroid cell line mediated activation of Notch signaling, upregulating Notch1 receptor and Hes1, the downstream effector of Notch pathway. Conversely, pharmacological inhibition of MAPK reduced Notch signaling in PTC cell. Thyroid tumor samples from transgenic mice expressing BRAF(T1799A) and primary human PTC samples showed high levels of Notch1 expression. Down-regulation of Notch signaling by γ-secretase inhibitor (GSI) or NOTCH1 RNA interference reduces PTC cell proliferation. Moreover, the combination of GSI with a MAPK inhibitor enhanced the growth suppression in PTC cells. This study revealed that RET/PTC and BRAF(T1799A) activate Notch signaling and promote tumor growth in thyroid follicular cell. Taken together, these data suggest that Notch signaling may be explored as an adjuvant therapy for thyroid papillary cancer.

7.
Cancer Immunol Res ; 1(5): 296-302, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-24777967

RÉSUMÉ

Meningiomas are the most common primary intracranial tumors. Surgical resection remains the treatment of choice for these tumors. However, a significant number of tumors are not surgically accessible, recur, or become malignant, necessitating the repetition of surgery and sometimes radiation. Chemotherapy is rarely used and is generally not recognized as an effective treatment. Cancer/testis (CT) genes represent a unique class of genes, which are expressed by germ cells, normally silenced in somatic cells, but activated in various cancers. CT proteins can elicit spontaneous immune responses in patients with cancer and this feature makes them attractive targets for immunotherapy-based approaches. We analyzed mRNA expression of 37 testis-restricted CT genes in a discovery set of 18 meningiomas by reverse transcription PCR. The overall frequency of expression of CT genes ranged from 5.6% to 27.8%. The most frequently expressed was NY-ESO-1, in 5 patients (27.8%). We subsequently analyzed NY-ESO-1 protein expression in a larger set of meningiomas by immunohistochemistry and found expression in 108 of 110 cases. In some cases, NY-ESO-1 expression was diffused and homogenous, but in most instances it was heterogeneous. Importantly, NY-ESO-1 expression was positively correlated with higher grade and patients presenting with higher levels of NY-ESO-1 staining had significantly worse disease-free and overall survival. We have also shown that NY-ESO-1 expression may lead to humoral immune response in patients with meningioma. Considering the limited treatment options for patients with meningioma, the potential of NY-ESO-1-based immunotherapy should be explored.


Sujet(s)
Antigènes néoplasiques/biosynthèse , Protéines membranaires/biosynthèse , Tumeurs des méninges/immunologie , Tumeurs des méninges/thérapie , Méningiome/immunologie , Méningiome/thérapie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps antitumoraux/biosynthèse , Anticorps antitumoraux/génétique , Antigènes néoplasiques/génétique , Antigènes néoplasiques/immunologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Immunohistochimie , Immunothérapie/méthodes , Mâle , Protéines membranaires/génétique , Protéines membranaires/immunologie , Tumeurs des méninges/génétique , Méningiome/génétique , Adulte d'âge moyen , Récidive tumorale locale/immunologie , Récidive tumorale locale/thérapie , Jeune adulte
8.
Mol Cancer Res ; 10(7): 904-13, 2012 Jul.
Article de Anglais | MEDLINE | ID: mdl-22618028

RÉSUMÉ

The Hippo signaling pathway is functionally conserved in Drosophila melanogaster and mammals, and its proposed function is to control tissue homeostasis by regulating cell proliferation and apoptosis. The core components are composed of a kinase cascade that culminates with the phosphorylation and inhibition of Yes-associated protein 1 (YAP1). Phospho-YAP1 is retained in the cytoplasm. In the absence of Hippo signaling, YAP1 translocates to the nucleus, associates with co-activators TEAD1-4, and functions as a transcriptional factor promoting the expression of key target genes. Components of the Hippo pathway are mutated in human cancers, and deregulation of this pathway plays a role in tumorigenesis. Loss of the NF2 tumor suppressor gene is the most common genetic alteration in meningiomas, and the NF2 gene product, Merlin, acts upstream of the Hippo pathway. Here, we show that primary meningioma tumors have high nuclear expression of YAP1. In meningioma cells, Merlin expression is associated with phosphorylation of YAP1. Using an siRNA transient knockdown of YAP1 in NF2-mutant meningioma cells, we show that suppression of YAP1 impaired cell proliferation and migration. Conversely, YAP1 overexpression led to a strong augment of cell proliferation and anchorage-independent growth and restriction of cisplatin-induced apoptosis. In addition, expression of YAP1 in nontransformed arachnoidal cells led to the development of tumors in nude mice. Together, these findings suggest that in meningiomas, deregulation of the Hippo pathway is largely observed in primary tumors and that YAP1 functions as an oncogene promoting meningioma tumorigenesis.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Tumeurs du cerveau , Méningiome , Phosphoprotéines , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Cisplatine/pharmacologie , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Humains , Méningiome/génétique , Méningiome/métabolisme , Souris , Tumeurs expérimentales/métabolisme , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , Petit ARN interférent , Analyse sur puce à tissus , Facteurs de transcription , Cicatrisation de plaie , Protéines de signalisation YAP
10.
Biochemistry ; 48(28): 6633-43, 2009 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-19530647

RÉSUMÉ

The eukaryotic single-stranded DNA-binding protein, replication protein A (RPA), is essential in DNA metabolism and is phosphorylated in response to DNA-damaging agents. Rad52 and RPA participate in the repair of double-stranded DNA breaks (DSBs). It is known that human RPA and Rad52 form a complex, but the molecular mass, stoichiometry, and exact role of this complex in DSB repair are unclear. In this study, absolute molecular masses of individual proteins and complexes were measured in solution using analytical size-exclusion chromatography coupled with multiangle light scattering, the protein species present in each purified fraction were verified via sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)/Western analyses, and the presence of biotinylated ssDNA in the complexes was verified by chemiluminescence detection. Then, employing UV cross-linking, the protein partner holding the ssDNA was identified. These data show that phosphorylated RPA promoted formation of a complex with monomeric Rad52 and caused the transfer of ssDNA from RPA to Rad52. This suggests that RPA phosphorylation may regulate the first steps of DSB repair and is necessary for the mediator function of Rad52.


Sujet(s)
Réparation de l'ADN , ADN simple brin/métabolisme , Protéine Rad52 de réparation-recombinaison de l'ADN/métabolisme , Protéine A de réplication/métabolisme , Chromatographie sur gel , ADN simple brin/ultrastructure , Humains , Lumière , Modèles biologiques , Phosphorylation , Protéine Rad52 de réparation-recombinaison de l'ADN/ultrastructure , Protéine A de réplication/ultrastructure , Diffusion de rayonnements
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE