Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 994-1005, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38532525

RÉSUMÉ

Trastuzumab deruxtecan (T-DXd; DS-8201; ENHERTU®) is a human epithelial growth factor receptor 2 (HER2)-directed antibody drug conjugate (ADC) with demonstrated antitumor activity against a range of tumor types. Aiming to understand the relationship between antigen expression and downstream efficacy outcomes, T-DXd was administered in tumor-bearing mice carrying NCI-N87, Capan-1, JIMT-1, and MDA-MB-468 xenografts, characterized by varying HER2 levels. Plasma pharmacokinetics (PK) of total antibody, T-DXd, and released DXd and tumor concentrations of released DXd were evaluated, in addition to monitoring γΗ2AX and pRAD50 pharmacodynamic (PD) response. A positive relationship was observed between released DXd concentrations in tumor and HER2 expression, with NCI-N87 xenografts characterized by the highest exposures compared to the remaining cell lines. γΗ2AX and pRAD50 demonstrated a sustained increase over several days occurring with a time delay relative to tumoral-released DXd concentrations. In vitro investigations of cell-based DXd disposition facilitated the characterization of DXd kinetics across tumor cells. These outputs were incorporated into a mechanistic mathematical model, utilized to describe PK/PD trends. The model captured plasma PK across dosing arms as well as tumor PK in NCI-N87, Capan-1, and MDA-MB-468 models; tumor concentrations in JIMT-1 xenografts required additional parameter adjustments reflective of complex receptor dynamics. γΗ2AX longitudinal trends were well characterized via a unified PD model implemented across xenografts demonstrating the robustness of measured PD trends. This work supports the application of a mechanistic model as a quantitative tool, reliably projecting tumor payload concentrations upon T-DXd administration, as the first step towards preclinical-to-clinical translation.


Sujet(s)
Immunoconjugués , Récepteur ErbB-2 , Trastuzumab , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Trastuzumab/pharmacocinétique , Trastuzumab/pharmacologie , Récepteur ErbB-2/métabolisme , Souris , Humains , Immunoconjugués/pharmacocinétique , Immunoconjugués/pharmacologie , Lignée cellulaire tumorale , Femelle , Camptothécine/analogues et dérivés , Camptothécine/pharmacocinétique , Camptothécine/pharmacologie , Antinéoplasiques immunologiques/pharmacocinétique , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/administration et posologie , Souris nude
2.
Cancer Discov ; 11(11): 2828-2845, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34230008

RÉSUMÉ

Mutations in the STK11 (LKB1) gene regulate resistance to PD-1/PD-L1 blockade. This study evaluated this association in patients with nonsquamous non-small cell lung cancer (NSCLC) enrolled in three phase I/II trials. STK11 mutations were associated with resistance to the anti-PD-L1 antibody durvalumab (alone/with the anti-CTLA4 antibody tremelimumab) independently of KRAS mutational status, highlighting STK11 as a potential driver of resistance to checkpoint blockade. Retrospective assessments of tumor tissue, whole blood, and serum revealed a unique immune phenotype in patients with STK11 mutations, with increased expression of markers associated with neutrophils (i.e., CXCL2, IL6), Th17 contexture (i.e., IL17A), and immune checkpoints. Associated changes were observed in the periphery. Reduction of STAT3 in the tumor microenvironment using an antisense oligonucleotide reversed immunotherapy resistance in preclinical STK11 knockout models. These results suggest that STK11 mutations may hinder response to checkpoint blockade through mechanisms including suppressive myeloid cell biology, which could be reversed by STAT3-targeted therapy. SIGNIFICANCE: Patients with nonsquamous STK11-mutant (STK11mut) NSCLC are less likely than STK11 wild-type (STK11wt) patients to respond to anti-PD-L1 ± anti-CTLA4 immunotherapies, and their tumors show increased expression of genes and cytokines that activate STAT3 signaling. Preclinically, STAT3 modulation reverses this resistance, suggesting STAT3-targeted agents as potential combination partners for immunotherapies in STK11mut NSCLC.This article is highlighted in the In This Issue feature, p. 2659.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , AMP-activated protein kinase kinases , Anticorps monoclonaux , Anticorps monoclonaux humanisés , Antigène CD274/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mutation , Protein-Serine-Threonine Kinases/génétique , Études rétrospectives , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme , Microenvironnement tumoral
3.
Mol Cancer Ther ; 20(6): 1080-1091, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33785652

RÉSUMÉ

Suppressive myeloid cells mediate resistance to immune checkpoint blockade. PI3Kγ inhibition can target suppressive macrophages, and enhance efficacy of immune checkpoint inhibitors. However, how PI3Kγ inhibitors function in different tumor microenvironments (TME) to activate specific immune cells is underexplored. The effect of the novel PI3Kγ inhibitor AZD3458 was assessed in preclinical models. AZD3458 enhanced antitumor activity of immune checkpoint inhibitors in 4T1, CT26, and MC38 syngeneic models, increasing CD8+ T-cell activation status. Immune and TME biomarker analysis of MC38 tumors revealed that AZD3458 monotherapy or combination treatment did not repolarize the phenotype of tumor-associated macrophage cells but induced gene signatures associated with LPS and type II INF activation. The activation biomarkers were present across tumor macrophages that appear phenotypically heterogenous. AZD3458 alone or in combination with PD-1-blocking antibodies promoted an increase in antigen-presenting (MHCII+) and cytotoxic (iNOS+)-activated macrophages, as well as dendritic cell activation. AZD3458 reduced IL-10 secretion and signaling in primary human macrophages and murine tumor-associated macrophages, but did not strongly regulate IL-12 as observed in other studies. Therefore, rather than polarizing tumor macrophages, PI3Kγ inhibition with AZD3458 promotes a cytotoxic switch of macrophages into antigen-presenting activated macrophages, resulting in CD8 T-cell-mediated antitumor activity with immune checkpoint inhibitors associated with tumor and peripheral immune activation.


Sujet(s)
Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Animaux , Modèles animaux de maladie humaine , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Souris
4.
Clin Cancer Res ; 26(23): 6335-6349, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-32943458

RÉSUMÉ

PURPOSE: Danvatirsen is a therapeutic antisense oligonucleotide (ASO) that selectively targets STAT3 and has shown clinical activity in two phase I clinical studies. We interrogated the clinical mechanism of action using danvatirsen-treated patient samples and conducted back-translational studies to further elucidate its immunomodulatory mechanism of action. EXPERIMENTAL DESIGN: Paired biopsies and blood samples from danvatirsen-treated patients were evaluated using immunohistochemistry and gene-expression analysis. To gain mechanistic insight, we used mass cytometry, flow cytometry, and immunofluorescence analysis of CT26 tumors treated with a mouse surrogate STAT3 ASO, and human immune cells were treated in vitro with danvatirsen. RESULTS: Within the tumors of treated patients, danvatirsen uptake was observed mainly in cells of the tumor microenvironment (TME). Gene expression analysis comparing baseline and on-treatment tumor samples showed increased expression of proinflammatory genes. In mouse models, STAT3 ASO demonstrated partial tumor growth inhibition and enhanced the antitumor activity when combined with anti-PD-L1. Immune profiling revealed reduced STAT3 protein in immune and stromal cells, and decreased suppressive cytokines correlating with increased proinflammatory macrophages and cytokine production. These changes led to enhanced T-cell abundance and function in combination with anti-PD-L1. CONCLUSIONS: STAT3 ASO treatment reverses a suppressive TME and promotes proinflammatory gene expression changes in patients' tumors and mouse models. Preclinical data provide evidence that ASO-mediated inhibition of STAT3 in the immune compartment is sufficient to remodel the TME and enhance the activity of checkpoint blockade without direct STAT3 inhibition in tumor cells. Collectively, these data provide a rationale for testing this combination in the clinic.


Sujet(s)
Antinéoplasiques immunologiques/pharmacologie , Antigène CD274/antagonistes et inhibiteurs , Tumeurs du côlon/thérapie , Tumeurs/thérapie , Oligonucléotides/pharmacologie , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Microenvironnement tumoral/immunologie , Essais cliniques de phase I comme sujet , Tumeurs du côlon/immunologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Association de médicaments , Humains , Immunomodulation , Macrophages/immunologie , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Pronostic , Facteur de transcription STAT-3/génétique , Lymphocytes T/immunologie , Cellules cancéreuses en culture
5.
Cancer Discov ; 4(6): 702-15, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24705811

RÉSUMÉ

UNLABELLED: Epithelial-to-mesenchymal transition (EMT) promotes both tumor progression and drug resistance, yet few vulnerabilities of this state have been identified. Using selective small molecules as cellular probes, we show that induction of EMT greatly sensitizes cells to agents that perturb endoplasmic reticulum (ER) function. This sensitivity to ER perturbations is caused by the synthesis and secretion of large quantities of extracellular matrix (ECM) proteins by EMT cells. Consistent with their increased secretory output, EMT cells display a branched ER morphology and constitutively activate the PERK-eIF2α axis of the unfolded protein response (UPR). Protein kinase RNA-like ER kinase (PERK) activation is also required for EMT cells to invade and metastasize. In human tumor tissues, EMT gene expression correlates strongly with both ECM and PERK-eIF2α genes, but not with other branches of the UPR. Taken together, our findings identify a novel vulnerability of EMT cells, and demonstrate that the PERK branch of the UPR is required for their malignancy. SIGNIFICANCE: EMT drives tumor metastasis and drug resistance, highlighting the need for therapies that target this malignant subpopulation. Our findings identify a previously unrecognized vulnerability of cancer cells that have undergone an EMT: sensitivity to ER stress. We also find that PERK-eIF2α signaling, which is required to maintain ER homeostasis, is also indispensable for EMT cells to invade and metastasize.


Sujet(s)
Stress du réticulum endoplasmique/génétique , Transition épithélio-mésenchymateuse/génétique , Facteur-2 d'initiation eucaryote/génétique , eIF-2 Kinase/génétique , Facteur de transcription ATF-4/génétique , Animaux , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Réticulum endoplasmique/métabolisme , Facteur-2 d'initiation eucaryote/métabolisme , Matrice extracellulaire/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Souris SCID , Réponse aux protéines mal repliées , eIF-2 Kinase/métabolisme
6.
Cell Stem Cell ; 8(2): 149-63, 2011 Feb 04.
Article de Anglais | MEDLINE | ID: mdl-21295272

RÉSUMÉ

Women with inherited mutations in the BRCA1 gene have increased risk of developing breast cancer but also exhibit a predisposition for the development of aggressive basal-like breast tumors. We report here that breast epithelial cells derived from patients harboring deleterious mutations in BRCA1 (BRCA1(mut /+) give rise to tumors with increased basal differentiation relative to cells from BRCA1+/+ patients. Molecular analysis of disease-free breast tissues from BRCA1(mut /+) patients revealed defects in progenitor cell lineage commitment even before cancer incidence. Moreover, we discovered that the transcriptional repressor Slug is an important functional suppressor of human breast progenitor cell lineage commitment and differentiation and that it is aberrantly expressed in BRCA1(mut /+) tissues. Slug expression is necessary for increased basal-like phenotypes prior to and after neoplastic transformation. These findings demonstrate that the genetic background of patient populations, in addition to affecting incidence rates, significantly impacts progenitor cell fate commitment and, therefore, tumor phenotype.


Sujet(s)
Tumeurs du sein/métabolisme , Prédisposition génétique à une maladie/génétique , Cellules souches/cytologie , Adulte , Animaux , Tumeurs du sein/génétique , Femelle , Cytométrie en flux , Gène BRCA1 , Humains , Immunohistochimie , Souris , Souris SCID , Adulte d'âge moyen , Données de séquences moléculaires , Phénotype , RT-PCR , Facteurs de transcription de la famille Snail , Cellules souches/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE