Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cancer Res ; 84(13): 2060-2072, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-39082680

RÉSUMÉ

Patient-derived xenografts (PDX) model human intra- and intertumoral heterogeneity in the context of the intact tissue of immunocompromised mice. Histologic imaging via hematoxylin and eosin (H&E) staining is routinely performed on PDX samples, which could be harnessed for computational analysis. Prior studies of large clinical H&E image repositories have shown that deep learning analysis can identify intercellular and morphologic signals correlated with disease phenotype and therapeutic response. In this study, we developed an extensive, pan-cancer repository of >1,000 PDX and paired parental tumor H&E images. These images, curated from the PDX Development and Trial Centers Research Network Consortium, had a range of associated genomic and transcriptomic data, clinical metadata, pathologic assessments of cell composition, and, in several cases, detailed pathologic annotations of neoplastic, stromal, and necrotic regions. The amenability of these images to deep learning was highlighted through three applications: (i) development of a classifier for neoplastic, stromal, and necrotic regions; (ii) development of a predictor of xenograft-transplant lymphoproliferative disorder; and (iii) application of a published predictor of microsatellite instability. Together, this PDX Development and Trial Centers Research Network image repository provides a valuable resource for controlled digital pathology analysis, both for the evaluation of technical issues and for the development of computational image-based methods that make clinical predictions based on PDX treatment studies. Significance: A pan-cancer repository of >1,000 patient-derived xenograft hematoxylin and eosin-stained images will facilitate cancer biology investigations through histopathologic analysis and contributes important model system data that expand existing human histology repositories.


Sujet(s)
Apprentissage profond , Tumeurs , Humains , Animaux , Souris , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/imagerie diagnostique , Génomique/méthodes , Hétérogreffes , Tests d'activité antitumorale sur modèle de xénogreffe , Syndromes lymphoprolifératifs/génétique , Syndromes lymphoprolifératifs/anatomopathologie , Traitement d'image par ordinateur/méthodes
2.
Nat Cancer ; 3(2): 232-250, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35221336

RÉSUMÉ

Models that recapitulate the complexity of human tumors are urgently needed to develop more effective cancer therapies. We report a bank of human patient-derived xenografts (PDXs) and matched organoid cultures from tumors that represent the greatest unmet need: endocrine-resistant, treatment-refractory and metastatic breast cancers. We leverage matched PDXs and PDX-derived organoids (PDxO) for drug screening that is feasible and cost-effective with in vivo validation. Moreover, we demonstrate the feasibility of using these models for precision oncology in real time with clinical care in a case of triple-negative breast cancer (TNBC) with early metastatic recurrence. Our results uncovered a Food and Drug Administration (FDA)-approved drug with high efficacy against the models. Treatment with this therapy resulted in a complete response for the individual and a progression-free survival (PFS) period more than three times longer than their previous therapies. This work provides valuable methods and resources for functional precision medicine and drug development for human breast cancer.


Sujet(s)
Organoïdes , Tumeurs du sein triple-négatives , Découverte de médicament , Hétérogreffes , Humains , Médecine de précision/méthodes , Tumeurs du sein triple-négatives/traitement médicamenteux , États-Unis , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Breast Cancer Res ; 23(1): 100, 2021 10 30.
Article de Anglais | MEDLINE | ID: mdl-34717714

RÉSUMÉ

BACKGROUND: Metastatic breast cancer (MBC) is incurable, with a 5-year survival rate of 28%. In the USA, more than 42,000 patients die from MBC every year. The most common type of breast cancer is estrogen receptor-positive (ER+), and more patients die from ER+ breast cancer than from any other subtype. ER+ tumors can be successfully treated with hormone therapy, but many tumors acquire endocrine resistance, at which point treatment options are limited. There is an urgent need for model systems that better represent human ER+ MBC in vivo, where tumors can metastasize. Patient-derived xenografts (PDX) made from MBC spontaneously metastasize, but the immunodeficient host is a caveat, given the known role of the immune system in tumor progression and response to therapy. Thus, we attempted to develop an immune-humanized PDX model of ER+ MBC. METHODS: NSG-SGM3 mice were immune-humanized with CD34+ hematopoietic stem cells, followed by engraftment of human ER+ endocrine resistant MBC tumor fragments. Strategies for exogenous estrogen supplementation were compared, and immune-humanization in blood, bone marrow, spleen, and tumors was assessed by flow cytometry and tissue immunostaining. Characterization of the new model includes assessment of the human tumor microenvironment performed by immunostaining. RESULTS: We describe the development of an immune-humanized PDX model of estrogen-independent endocrine resistant ER+ MBC. Importantly, our model harbors a naturally occurring ESR1 mutation, and immune-humanization recapitulates the lymphocyte-excluded and myeloid-rich tumor microenvironment of human ER+ breast tumors. CONCLUSION: This model sets the stage for development of other clinically relevant models of human breast cancer and should allow future studies on mechanisms of endocrine resistance and tumor-immune interactions in an immune-humanized in vivo setting.


Sujet(s)
Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Résistance aux médicaments antinéoplasiques , Récepteurs des oestrogènes/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Antigènes CD34/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Récepteur alpha des oestrogènes/génétique , Oestrogènes/administration et posologie , Oestrogènes/pharmacologie , Femelle , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/métabolisme , Hétérogreffes/effets des médicaments et des substances chimiques , Hétérogreffes/métabolisme , Hétérogreffes/anatomopathologie , Humains , Souris , Souris de lignée NOD , Souris SCID , Souris transgéniques , Mutation , Récepteurs des oestrogènes/génétique , Microenvironnement tumoral/immunologie
4.
Breast Cancer Res ; 22(1): 132, 2020 11 30.
Article de Anglais | MEDLINE | ID: mdl-33256808

RÉSUMÉ

BACKGROUND: Targeted therapies for triple-negative breast cancer (TNBC) are limited; however, the epidermal growth factor receptor (EGFR) represents a potential target, as the majority of TNBC express EGFR. The purpose of these studies was to evaluate the effectiveness of two EGFR-targeted antibody-drug conjugates (ADC: ABT-414; ABBV-321) in combination with navitoclax, an antagonist of the anti-apoptotic BCL-2 and BCL-XL proteins, in order to assess the translational relevance of these combinations for TNBC. METHODS: The pre-clinical efficacy of combined treatments was evaluated in multiple patient-derived xenograft (PDX) models of TNBC. Microscopy-based dynamic BH3 profiling (DBP) was used to assess mitochondrial apoptotic signaling induced by navitoclax and/or ADC treatments, and the expression of EGFR and BCL-2/XL was analyzed in 46 triple-negative patient tumors. RESULTS: Treatment with navitoclax plus ABT-414 caused a significant reduction in tumor growth in five of seven PDXs and significant tumor regression in the highest EGFR-expressing PDX. Navitoclax plus ABBV-321, an EGFR-targeted ADC that displays more effective wild-type EGFR-targeting, elicited more significant tumor growth inhibition and regressions in the two highest EGFR-expressing models evaluated. The level of mitochondrial apoptotic signaling induced by single or combined drug treatments, as measured by DBP, correlated with the treatment responses observed in vivo. Lastly, the majority of triple-negative patient tumors were found to express EGFR and co-express BCL-XL and/or BCL-2. CONCLUSIONS: The dramatic tumor regressions achieved using combined agents in pre-clinical TNBC models underscore the abilities of BCL-2/XL antagonists to enhance the effectiveness of EGFR-targeted ADCs and highlight the clinical potential for usage of such targeted ADCs to alleviate toxicities associated with combinations of BCL-2/XL inhibitors and systemic chemotherapies.


Sujet(s)
Dérivés de l'aniline/pharmacologie , Anticorps monoclonaux humanisés/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Immunoconjugués/pharmacologie , Sulfonamides/pharmacologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Dérivés de l'aniline/usage thérapeutique , Animaux , Anticorps monoclonaux humanisés/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Région mammaire/anatomopathologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/analyse , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Femelle , Humains , Immunoconjugués/usage thérapeutique , Souris , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/métabolisme , Sulfonamides/usage thérapeutique , Tumeurs du sein triple-négatives/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe , Protéine bcl-X/antagonistes et inhibiteurs , Protéine bcl-X/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE