Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
1.
J Proteome Res ; 21(10): 2331-2340, 2022 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-36049057

RÉSUMÉ

The fibronectin (FN) isoform including the extradomain B (EDB) segment (EDB + FN) is a promising tumor target and is highly expressed in some tumor types, such as breast, head, and neck cancer. To date, mostly immunohistochemistry (IHC) and Western blot have been used for the analysis of EDB + FN. However, complete quantitative measurements of EDB + FN expression in a tumor and circulation are important for the development of anti-EDB therapeutics. To this end, a method using protein enrichment followed by online antipeptide antibody enrichment coupled with a nanoflow LC-MS/MS was developed to quantify EDB + FN in human and cynomolgus plasma, patient-derived xenograft (PDX) tumors, and PDX formalin-fixed paraffin-embedded (FFPE) samples. Mouse plasma EDB + FN was analyzed using a protein immunoaffinity method followed by nanoflow LC-MS/MS. EDB + FN concentrations were 63.1 pmol/g in PDX breast cancer tumor and 49.6 pmol/g in PDX head and neck tumor. Mean plasma concentration was 1.1 nM (pmol/mL, 47.4 ng/mL) in normal healthy humans and 0.35 nM (15.1 ng/mL) in naive cynomolgus. The assay sensitivity was 0.018 nM based on calibration with recombinant human EDB + FN (rhEDB + FN).


Sujet(s)
Tumeurs du sein , Fibronectines , Animaux , Tumeurs du sein/anatomopathologie , Chromatographie en phase liquide , Modèles animaux de maladie humaine , Femelle , Fibronectines/analyse , Formaldéhyde , Hétérogreffes , Humains , Souris , Inclusion en paraffine , Isoformes de protéines/métabolisme , Spectrométrie de masse en tandem
2.
Mol Cancer Ther ; 21(9): 1462-1472, 2022 09 06.
Article de Anglais | MEDLINE | ID: mdl-35793468

RÉSUMÉ

Extra domain B splice variant of fibronectin (EDB+FN) is an extracellular matrix protein (ECM) deposited by tumor-associated fibroblasts, and is associated with tumor growth, angiogenesis, and invasion. We hypothesized that EDB+FN is a safe and abundant target for therapeutic intervention with an antibody-drug conjugate (ADC). We describe the generation, pharmacology, mechanism of action, and safety profile of an ADC specific for EDB+FN (EDB-ADC). EDB+FN is broadly expressed in the stroma of pancreatic, non-small cell lung (NSCLC), breast, ovarian, head and neck cancers, whereas restricted in normal tissues. In patient-derived xenograft (PDX), cell-line xenograft (CLX), and mouse syngeneic tumor models, EDB-ADC, conjugated to auristatin Aur0101 through site-specific technology, demonstrated potent antitumor growth inhibition. Increased phospho-histone H3, a pharmacodynamic biomarker of response, was observed in tumor cells distal to the target site of tumor ECM after EDB-ADC treatment. EDB-ADC potentiated infiltration of immune cells, including CD3+ T lymphocytes into the tumor, providing rationale for the combination of EDB-ADC with immune checkpoint therapy. EDB-ADC and anti-PD-L1 combination in a syngeneic breast tumor model led to enhanced antitumor activity with sustained tumor regressions. In nonclinical safety studies in nonhuman primates, EDB-ADC had a well-tolerated safety profile without signs of either on-target toxicity or the off-target effects typically observed with ADCs that are conjugated through conventional conjugation methods. These data highlight the potential for EDB-ADC to specifically target the tumor microenvironment, provide robust therapeutic benefits against multiple tumor types, and enhance activity antitumor in combination with checkpoint blockade.


Sujet(s)
Tumeurs du sein , Immunoconjugués , Animaux , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Femelle , Fibronectines/métabolisme , Humains , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Souris , Néovascularisation pathologique/métabolisme , Microenvironnement tumoral , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Cell Rep Med ; 2(5): 100279, 2021 05 18.
Article de Anglais | MEDLINE | ID: mdl-34095881

RÉSUMÉ

Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Immunoconjugués/pharmacologie , Récepteur Notch3/métabolisme , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Humains , Immunoconjugués/métabolisme , Oncogènes/effets des médicaments et des substances chimiques , Récepteur Notch3/immunologie , Récepteurs Notch/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Mol Cancer Ther ; 19(10): 2068-2078, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32747418

RÉSUMÉ

The approval of ado-trastuzumab emtansine (T-DM1) in HER2+ metastatic breast cancer validated HER2 as a target for HER2-specific antibody-drug conjugates (ADC). Despite its demonstrated clinical efficacy, certain inherent properties within T-DM1 hamper this compound from achieving the full potential of targeting HER2-expressing solid tumors with ADCs. Here, we detail the discovery of PF-06804103, an anti-HER2 ADC designed to have a widened therapeutic window compared with T-DM1. We utilized an empirical conjugation site screening campaign to identify the engineered ĸkK183C and K290C residues as those that maximized in vivo ADC stability, efficacy, and safety for a four drug-antibody ratio (DAR) ADC with this linker-payload combination. PF-06804103 incorporates the following novel design elements: (i) a new auristatin payload with optimized pharmacodynamic properties, (ii) a cleavable linker for optimized payload release and enhanced antitumor efficacy, and (iii) an engineered cysteine site-specific conjugation approach that overcomes the traditional safety liabilities of conventional conjugates and generates a homogenous drug product with a DAR of 4. PF-06804103 shows (i) an enhanced efficacy against low HER2-expressing breast, gastric, and lung tumor models, (ii) overcomes in vitro- and in vivo-acquired T-DM1 resistance, and (iii) an improved safety profile by enhancing ADC stability, pharmacokinetic parameters, and reducing off-target toxicities. Herein, we showcase our platform approach in optimizing ADC design, resulting in the generation of the anti-HER2 ADC, PF-06804103. The design elements of identifying novel sites of conjugation employed in this study serve as a platform for developing optimized ADCs against other tumor-specific targets.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Immunoconjugués/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs de l'estomac/traitement médicamenteux , Animaux , Tumeurs du sein/anatomopathologie , Femelle , Humains , Immunoconjugués/pharmacologie , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Tumeurs de l'estomac/anatomopathologie
5.
J Vis Exp ; (157)2020 03 17.
Article de Anglais | MEDLINE | ID: mdl-32250351

RÉSUMÉ

Tumor-stroma interactions play a critical role in the development of lung squamous carcinoma (LUSC). However, understanding how these dynamic interactions contribute to tissue architectural changes observed during tumorigenesis remains challenging due to the lack of appropriate models. In this protocol, we describe the generation of a 3D coculture model using a LUSC primary cell culture known as TUM622. TUM622 cells were established from a LUSC patient-derived xenograft (PDX) and have the unique property to form acinar-like structures when seeded in a basement membrane matrix. We demonstrate that TUM622 acini in 3D coculture recapitulate key features of tissue architecture during LUSC progression as well as the dynamic interactions between LUSC cells and components of the tumor microenvironment (TME), including the extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs). We further adapt our principal 3D culturing protocol to demonstrate how this system could be utilized for various downstream analyses. Overall, this organoid model creates a biologically rich and adaptable platform that enables one to gain insight into the cell-intrinsic and extrinsic mechanisms that promote the disruption of epithelial architectures during carcinoma progression and will aid the search for new therapeutic targets and diagnostic markers.


Sujet(s)
Carcinome épidermoïde/anatomopathologie , Tumeurs du poumon/anatomopathologie , Transformation cellulaire néoplasique/métabolisme , Techniques de coculture , Évolution de la maladie , Matrice extracellulaire/anatomopathologie , Humains , Organoïdes/métabolisme , Culture de cellules primaires , Microenvironnement tumoral
6.
Proc Natl Acad Sci U S A ; 115(50): E11671-E11680, 2018 12 11.
Article de Anglais | MEDLINE | ID: mdl-30487219

RÉSUMÉ

Tumorigenesis depends on intricate interactions between genetically altered tumor cells and their surrounding microenvironment. While oncogenic drivers in lung squamous carcinoma (LUSC) have been described, the role of stroma in modulating tissue architecture, particularly cell polarity, remains unclear. Here, we report the establishment of a 3D coculture system of LUSC epithelial cells with cancer-associated fibroblasts (CAFs) and extracellular matrix that together capture key components of the tumor microenvironment (TME). Single LUSC epithelial cells develop into acinar-like structures with 0.02% efficiency, and addition of CAFs provides proper tumor-stromal interactions within an appropriate 3D architectural context. Using this model, we recapitulate key pathological changes during tumorigenesis, from hyperplasia to dysplasia and eventually invasion, in malignant LUSC spheroids that undergo phenotypic switching in response to cell intrinsic and extrinsic changes. Overexpression of SOX2 is sufficient to mediate the transition from hyperplasia to dysplasia in LUSC spheroids, while the presence of CAFs makes them invasive. Unexpectedly, CAFs suppress the activity of high SOX2 levels, restore hyperplasia, and enhance the formation of acinar-like structures. Taken together, these observations suggest that stromal factors can override cell intrinsic oncogenic changes in determining the disease phenotype, thus providing fundamental evidence for the existence of dynamic reciprocity between the nucleus and the TME of LUSC.


Sujet(s)
Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Facteurs de transcription SOX-B1/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome épidermoïde/génétique , Polarité de la cellule , Techniques de coculture , Régulation de l'expression des gènes tumoraux , Humains , Hyperplasie , Tumeurs du poumon/génétique , Modèles biologiques , Invasion tumorale/génétique , Invasion tumorale/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Facteurs de transcription SOX-B1/génétique , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/anatomopathologie , Cellules cancéreuses en culture/métabolisme , Cellules cancéreuses en culture/anatomopathologie , Microenvironnement tumoral/génétique , Régulation positive
7.
EMBO J ; 34(13): 1773-85, 2015 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-25979827

RÉSUMÉ

Lat1 (SLC7A5) is an amino acid transporter often required for tumor cell import of essential amino acids (AA) including Methionine (Met). Met is the obligate precursor of S-adenosylmethionine (SAM), the methyl donor utilized by all methyltransferases including the polycomb repressor complex (PRC2)-specific EZH2. Cell populations sorted for surface Lat1 exhibit activated EZH2, enrichment for Met-cycle intermediates, and aggressive tumor growth in mice. In agreement, EZH2 and Lat1 expression are co-regulated in models of cancer cell differentiation and co-expression is observed at the invasive front of human lung tumors. EZH2 knockdown or small-molecule inhibition leads to de-repression of RXRα resulting in reduced Lat1 expression. Our results describe a Lat1-EZH2 positive feedback loop illustrated by AA depletion or Lat1 knockdown resulting in SAM reduction and concomitant reduction in EZH2 activity. shRNA-mediated knockdown of Lat1 results in tumor growth inhibition and points to Lat1 as a potential therapeutic target.


Sujet(s)
Acides aminés/métabolisme , Épigenèse génétique/physiologie , Transporteur-1 d'acides aminés neutres à longue chaîne/physiologie , Complexe répresseur Polycomb-2/physiologie , Animaux , Transport biologique/génétique , Prolifération cellulaire/génétique , Protéine-2 homologue de l'activateur de Zeste , Femelle , Régulation de l'expression des gènes tumoraux , Cellules HEK293 , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Cellules cancéreuses en culture
8.
Oncotarget ; 6(5): 2928-38, 2015 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-25671303

RÉSUMÉ

In addition to genetic alterations, cancer cells are characterized by myriad epigenetic changes. EZH2 is a histone methyltransferase that is over-expressed and mutated in cancer. The EZH2 gain-of-function (GOF) mutations first identified in lymphomas have recently been reported in melanoma (~2%) but remain uncharacterized. We expressed multiple EZH2 GOF mutations in the A375 metastatic skin melanoma cell line and observed both increased H3K27me3 and dramatic changes in 3D culture morphology. In these cells, prominent morphological changes were accompanied by a decrease in cell contractility and an increase in collective cell migration. At the molecular level, we observed significant alteration of the axonal guidance pathway, a pathway intricately involved in the regulation of cell shape and motility. Furthermore, the aggressive 3D morphology of EZH2 GOF-expressing melanoma cells (both endogenous and ectopic) was attenuated by EZH2 catalytic inhibition. Finally, A375 cells expressing exogenous EZH2 GOF mutants formed larger tumors than control cells in mouse xenograft studies. This study not only demonstrates the first functional characterization of EZH2 GOF mutants in non-hematopoietic cells, but also provides a rationale for EZH2 catalytic inhibition in melanoma.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Forme de la cellule , Épigenèse génétique , Mélanome/génétique , Mutation , Complexe répresseur Polycomb-2/génétique , Tumeurs cutanées/génétique , Animaux , Lignée cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Forme de la cellule/effets des médicaments et des substances chimiques , Méthylation de l'ADN , Protéine-2 homologue de l'activateur de Zeste , Antienzymes/pharmacologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Jumonji Domain-Containing Histone Demethylases/métabolisme , Mélanome/traitement médicamenteux , Mélanome/enzymologie , Mélanome/anatomopathologie , Souris nude , Thérapie moléculaire ciblée , Invasion tumorale , Complexe répresseur Polycomb-2/métabolisme , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/enzymologie , Tumeurs cutanées/anatomopathologie , Charge tumorale , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Mol Cancer Ther ; 12(1): 38-47, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-23223830

RÉSUMÉ

Antibody-drug conjugates (ADC) represent a promising therapeutic modality for the clinical management of cancer. We sought to develop a novel ADC that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells (TIC), which comprise the most aggressive cell population in the tumor. We optimized an anti-5T4 ADC (A1mcMMAF) by sulfydryl-based conjugation of the humanized A1 antibody to the tubulin inhibitor monomethylauristatin F (MMAF) via a maleimidocaproyl linker. A1mcMMAF exhibited potent in vivo antitumor activity in a variety of tumor models and induced long-term regressions for up to 100 days after the last dose. Strikingly, animals showed pathologic complete response in each model with doses as low as 3 mg antibody/kg dosed every 4 days. In a non-small cell lung cancer patient-derived xenograft model, in which 5T4 is preferentially expressed on the less differentiated tumor cells, A1mcMMAF treatment resulted in sustained tumor regressions and reduced TIC frequency. These results highlight the potential of ADCs that target the most aggressive cell populations within tumors, such as TICs. In exploratory safety studies, A1mcMMAF exhibited no overt toxicities when administered to cynomolgus monkeys at doses up to 10 mg antibody/kg/cycle × 2 and displayed a half-life of 5 days. The preclinical efficacy and safety data established a promising therapeutic index that supports clinical testing of A1mcMMAF.


Sujet(s)
Anticorps monoclonaux humanisés/pharmacologie , Antinéoplasiques/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Glycoprotéines membranaires/métabolisme , Cellules souches tumorales/métabolisme , Animaux , Anticorps monoclonaux humanisés/métabolisme , Anticorps monoclonaux humanisés/toxicité , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/métabolisme , Antinéoplasiques/métabolisme , Antinéoplasiques/toxicité , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Humains , Concentration inhibitrice 50 , Macaca fascicularis , Mâle , Dose maximale tolérée , Glycoprotéines membranaires/immunologie , Souris , Souris nude , Cellules souches tumorales/effets des médicaments et des substances chimiques , Induction de rémission , Distribution tissulaire , Modulateurs de la polymérisation de la tubuline/synthèse chimique , Modulateurs de la polymérisation de la tubuline/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Cancer Res ; 71(12): 4236-46, 2011 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-21540235

RÉSUMÉ

Poorly differentiated tumors in non-small cell lung cancer (NSCLC) have been associated with shorter patient survival and shorter time to recurrence following treatment. Here, we integrate multiple experimental models with clinicopathologic analysis of patient tumors to delineate a cellular hierarchy in NSCLC. We show that the oncofetal protein 5T4 is expressed on tumor-initiating cells and associated with worse clinical outcome in NSCLC. Coexpression of 5T4 and factors involved in the epithelial-to-mesenchymal transition were observed in undifferentiated but not in differentiated tumor cells. Despite heterogeneous expression of 5T4 in NSCLC patient-derived xenografts, treatment with an anti-5T4 antibody-drug conjugate resulted in complete and sustained tumor regression. Thus, the aggressive growth of heterogeneous solid tumors can be blocked by therapeutic agents that target a subpopulation of cells near the top of the cellular hierarchy.


Sujet(s)
Antigènes néoplasiques/analyse , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Immunotoxines/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Glycoprotéines membranaires/analyse , Cellules souches tumorales/immunologie , Animaux , Antigènes CD24/analyse , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse , Humains , Antigènes CD44/analyse , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Glycoprotéines membranaires/physiologie , Souris
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...