Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
1.
Prostate ; 84(11): 1033-1046, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38708958

RÉSUMÉ

BACKGROUND: Preclinical models recapitulating the metastatic phenotypes are essential for developing the next-generation therapies for metastatic prostate cancer (mPC). We aimed to establish a cohort of clinically relevant mPC models, particularly androgen receptor positive (AR+) bone metastasis models, from LuCaP patient-derived xenografts (PDX) that reflect the heterogeneity and complexity of mPC. METHODS: PDX tumors were dissociated into single cells, modified to express luciferase, and were inoculated into NSG mice via intracardiac injection. The progression of metastases was monitored by bioluminescent imaging. Histological phenotypes of metastases were characterized by immunohistochemistry and immunofluorescence staining. Castration responses were further investigated in two AR-positive models. RESULTS: Our PDX-derived metastasis (PDM) model collection comprises three AR+ adenocarcinomas (ARPC) and one AR- neuroendocrine carcinoma (NEPC). All ARPC models developed bone metastases with either an osteoblastic, osteolytic, or mixed phenotype, while the NEPC model mainly developed brain metastasis. Different mechanisms of castration resistance were observed in two AR+ PDM models with distinct genotypes, such as combined loss of TP53 and RB1 in one model and expression of AR splice variant 7 (AR-V7) expression in another model. Intriguingly, the castration-resistant tumors displayed inter- and intra-tumor as well as organ-specific heterogeneity in lineage specification. CONCLUSION: Genetically diverse PDM models provide a clinically relevant system for biomarker identification and personalized medicine in metastatic castration-resistant prostate cancer.


Sujet(s)
Tumeurs osseuses , Modèles animaux de maladie humaine , Tumeurs de la prostate , Récepteurs aux androgènes , Mâle , Tumeurs osseuses/secondaire , Tumeurs osseuses/métabolisme , Animaux , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Humains , Souris , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Adénocarcinome/anatomopathologie , Adénocarcinome/secondaire , Adénocarcinome/métabolisme , Adénocarcinome/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Carcinome neuroendocrine/anatomopathologie , Carcinome neuroendocrine/métabolisme , Carcinome neuroendocrine/génétique
2.
medRxiv ; 2024 Feb 11.
Article de Anglais | MEDLINE | ID: mdl-38370835

RÉSUMÉ

Patients diagnosed with localized high-risk prostate cancer have higher rates of recurrence, and the introduction of neoadjuvant intensive hormonal therapies seeks to treat occult micrometastatic disease by their addition to definitive treatment. Sufficient profiling of baseline disease has remained a challenge in enabling the in-depth assessment of phenotypes associated with exceptional vs. poor pathologic responses after treatment. In this study, we report comprehensive and integrative gene expression profiling of 37 locally advanced prostate tumors prior to six months of androgen deprivation therapy (ADT) plus the androgen receptor (AR) inhibitor enzalutamide prior to radical prostatectomy. A robust transcriptional program associated with HER2 activity was positively associated with poor outcome and opposed AR activity, even after adjusting for common genomic alterations in prostate cancer including PTEN loss and expression of the TMPRSS2:ERG fusion. Patients experiencing exceptional pathologic responses demonstrated lower levels of HER2 and phospho-HER2 by immunohistochemistry of biopsy tissues. The inverse correlation of AR and HER2 activity was found to be a universal feature of all aggressive prostate tumors, validated by transcriptional profiling an external cohort of 121 patients and immunostaining of tumors from 84 additional patients. Importantly, the AR activity-low, HER2 activity-high cells that resist ADT are a pre-existing subset of cells that can be targeted by HER2 inhibition alone or in combination with enzalutamide. In summary, we show that prostate tumors adopt an AR activity-low prior to antiandrogen exposure that can be exploited by treatment with HER2 inhibitors.

3.
Clin Cancer Res ; 28(16): 3509-3525, 2022 08 15.
Article de Anglais | MEDLINE | ID: mdl-35695870

RÉSUMÉ

PURPOSE: Therapies targeting the androgen receptor (AR) have improved the outcome for patients with castration-sensitive prostate cancer (CSPC). Expression of the constitutively active AR splice variant-7 (AR-V7) has shown clinical utility as a predictive biomarker of AR-targeted therapy resistance in castration-resistant prostate cancer (CRPC), but its importance in CSPC remains understudied. EXPERIMENTAL DESIGN: We assessed different approaches to quantify AR-V7 mRNA and protein in prostate cancer cell lines, patient-derived xenograft (PDX) models, publicly available cohorts, and independent institutional clinical cohorts, to identify reliable approaches for detecting AR-V7 mRNA and protein and its association with clinical outcome. RESULTS: In CSPC and CRPC cohorts, AR-V7 mRNA was much less abundant when detected using reads across splice boundaries than when considering isoform-specific exonic reads. The RM7 AR-V7 antibody had increased sensitivity and specificity for AR-V7 protein detection by immunohistochemistry (IHC) in CRPC cohorts but rarely identified AR-V7 protein reactivity in CSPC cohorts, when compared with the EPR15656 AR-V7 antibody. Using multiple CRPC PDX models, we demonstrated that AR-V7 expression was exquisitely sensitive to hormonal manipulation. In CSPC institutional cohorts, AR-V7 protein quantification by either assay was associated neither with time to development of castration resistance nor with overall survival, and intense neoadjuvant androgen-deprivation therapy did not lead to significant AR-V7 mRNA or staining following treatment. Neither pre- nor posttreatment AR-V7 levels were associated with volumes of residual disease after therapy. CONCLUSIONS: This study demonstrates that further analytical validation and clinical qualification are required before AR-V7 can be considered for clinical use in CSPC as a predictive biomarker.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Récepteurs aux androgènes , Antagonistes des androgènes/usage thérapeutique , Marqueurs biologiques , Castration , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , ARN messager/génétique , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme
4.
J Urol ; 208(1): 90-99, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35227084

RÉSUMÉ

PURPOSE: Neoadjuvant intense androgen deprivation therapy (iADT) can exert a wide range of histological responses, which in turn are reflected in the final prostatectomy specimen. Accurate identification and measurement of residual tumor volumes are critical for tracking and stratifying patient outcomes. MATERIALS AND METHODS: The goal of this current study was to evaluate the ability of antibodies against prostate-specific membrane antigen (PSMA) to specifically detect residual tumor in a cohort of 35 patients treated with iADT plus enzalutamide for 6 months prior to radical prostatectomy. RESULTS: Residual carcinoma was detected in 31 patients, and PSMA reacted positively with tumor in all cases. PSMA staining was 96% sensitive for tumor, with approximately 82% of benign regions showing no reactivity. By contrast, PSMA positively reacted with 72% of benign regions in a control cohort of 37 untreated cases, resulting in 28% specificity for tumor. PSMA further identified highly dedifferentiated prostate carcinomas including tumors with evidence of neuroendocrine differentiation. CONCLUSIONS: We propose that anti-PSMA immunostaining be a standardized marker for identifying residual cancer in the setting of iADT.


Sujet(s)
Tumeurs de la prostate , Antagonistes des androgènes/usage thérapeutique , Androgènes , Humains , Mâle , Traitement néoadjuvant , Maladie résiduelle , Prostate/anatomopathologie , Antigène spécifique de la prostate , Prostatectomie , Tumeurs de la prostate/diagnostic , Tumeurs de la prostate/traitement médicamenteux
5.
Eur Urol ; 80(6): 746-757, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-33785256

RÉSUMÉ

BACKGROUND: Patients diagnosed with high risk localized prostate cancer have variable outcomes following surgery. Trials of intense neoadjuvant androgen deprivation therapy (NADT) have shown lower rates of recurrence among patients with minimal residual disease after treatment. The molecular features that distinguish exceptional responders from poor responders are not known. OBJECTIVE: To identify genomic and histologic features associated with treatment resistance at baseline. DESIGN, SETTING, AND PARTICIPANTS: Targeted biopsies were obtained from 37 men with intermediate- to high-risk prostate cancer before receiving 6 mo of ADT plus enzalutamide. Biopsy tissues were used for whole-exome sequencing and immunohistochemistry (IHC). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: We assessed the relationship of molecular features with final pathologic response using a cutpoint of 0.05 cm3 for residual cancer burden to compare exceptional responders to incomplete and nonresponders. We assessed intratumoral heterogeneity at the tissue and genomic level, and compared the volume of residual disease to the Shannon diversity index for each tumor. We generated multivariate models of resistance based on three molecular features and one histologic feature, with and without multiparametric magnetic resonance imaging estimates of baseline tumor volume. RESULTS AND LIMITATIONS: Loss of chromosome 10q (containing PTEN) and alterations to TP53 were predictive of poor response, as were the expression of nuclear ERG on IHC and the presence of intraductal carcinoma of the prostate. Patients with incompletely and nonresponding tumors harbored greater tumor diversity as estimated via phylogenetic tree reconstruction from DNA sequencing and analysis of IHC staining. Our four-factor binary model (area under the receiver operating characteristic curve [AUC] 0.89) to predict poor response correlated with greater diversity in our cohort and a validation cohort of 57 Gleason score 8-10 prostate cancers from The Cancer Genome Atlas. When baseline tumor volume was added to the model, it distinguished poor response to NADT with an AUC of 0.98. Prospective use of this model requires further retrospective validation with biopsies from additional trials. CONCLUSIONS: A subset of prostate cancers exhibit greater histologic and genomic diversity at the time of diagnosis, and these localized tumors have greater fitness to resist therapy. PATIENT SUMMARY: Some prostate cancer tumors do not respond well to a hormonal treatment called androgen deprivation therapy (ADT). We used tumor volume and four other parameters to develop a model to identify tumors that will not respond well to ADT. Treatments other than ADT should be considered for these patients.


Sujet(s)
Antagonistes des androgènes , Tumeurs de la prostate , Antagonistes des androgènes/effets indésirables , Androgènes , Humains , Mâle , Phylogenèse , Études prospectives , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Études rétrospectives
6.
Clin Cancer Res ; 27(2): 429-437, 2021 01 15.
Article de Anglais | MEDLINE | ID: mdl-33023952

RÉSUMÉ

PURPOSE: For high-risk prostate cancer, standard treatment options include radical prostatectomy (RP) or radiotherapy plus androgen deprivation therapy (ADT). Despite definitive therapy, many patients will have disease recurrence. Imaging has the potential to better define characteristics of response and resistance. In this study, we evaluated prostate multiparametric MRI (mpMRI) before and after neoadjuvant enzalutamide plus ADT. PATIENTS AND METHODS: Men with localized intermediate- or high-risk prostate cancer underwent a baseline mpMRI and mpMRI-targeted biopsy followed by a second mpMRI after 6 months of enzalutamide and ADT prior to RP. Specimens were sectioned in the same plane as mpMRI using patient-specific 3D-printed molds to permit mpMRI-targeted biopsies to be compared with the same lesion from the RP. Specimens were analyzed for imaging and histologic correlates of response. RESULTS: Of 39 patients enrolled, 36 completed imaging and RP. Most patients (92%) had high-risk disease. Fifty-eight lesions were detected on baseline mpMRI, of which 40 (69%) remained measurable at 6-month follow-up imaging. Fifty-five of 59 lesions (93%) demonstrated >50% volume reduction on posttreatment mpMRI. Three of 59 lesions (5%) demonstrated growth in size at follow-up imaging, with two lesions increasing more than 3-fold in volume. On whole-mount pathology, 15 patients demonstrated minimal residual disease (MRD) of <0.05 cc or pathologic complete response. Low initial mpMRI relative tumor burden was most predictive of MRD on final pathology. CONCLUSIONS: Low relative lesion volume at baseline mpMRI was predictive of pathologic response. A subset of patients had limited response. Selection of patients based on these metrics may improve outcomes in high-risk disease.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Imagerie par résonance magnétique multiparamétrique/méthodes , Tumeurs de la prostate/imagerie diagnostique , Tumeurs de la prostate/traitement médicamenteux , Sujet âgé , Antagonistes des androgènes/administration et posologie , Antagonistes des androgènes/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Benzamides/administration et posologie , Benzamides/effets indésirables , Fatigue/induit chimiquement , Bouffées de chaleur/induit chimiquement , Humains , Mâle , Adulte d'âge moyen , Traitement néoadjuvant/effets indésirables , Traitement néoadjuvant/méthodes , Nitriles/administration et posologie , Nitriles/effets indésirables , /méthodes , /statistiques et données numériques , 3-Phényl-2-thiohydantoïne/administration et posologie , 3-Phényl-2-thiohydantoïne/effets indésirables , Prostate/imagerie diagnostique , Prostate/effets des médicaments et des substances chimiques , Prostate/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Facteurs de risque , Charge tumorale/effets des médicaments et des substances chimiques
7.
Nat Commun ; 11(1): 837, 2020 02 13.
Article de Anglais | MEDLINE | ID: mdl-32054861

RÉSUMÉ

Localized prostate cancers are genetically variable and frequently multifocal, comprising spatially distinct regions with multiple independently-evolving clones. To date there is no understanding of whether this variability can influence management decisions for patients with prostate tumors. Here, we present a single case from a clinical trial of neoadjuvant intense androgen deprivation therapy. A patient was diagnosed with a large semi-contiguous tumor by imaging, histologically composed of a large Gleason score 9 tumor with an adjacent Gleason score 7 nodule. DNA sequencing demonstrates these are two independent tumors, as only the Gleason 9 tumor harbors single-copy losses of PTEN and TP53. The PTEN/TP53-deficient tumor demonstrates treatment resistance, selecting for subclones with mutations to the remaining copies of PTEN and TP53, while the Gleason 7 PTEN-intact tumor is almost entirely ablated. These findings indicate that spatiogenetic variability is a major confounder for personalized treatment of patients with prostate cancer.


Sujet(s)
Tumeurs primitives multiples/traitement médicamenteux , Tumeurs de la prostate/traitement médicamenteux , Sujet âgé , Antagonistes des androgènes/usage thérapeutique , Résistance aux médicaments antinéoplasiques , Délétion de gène , Humains , Mâle , Mutation , Traitement néoadjuvant , Grading des tumeurs , Tumeurs primitives multiples/génétique , Tumeurs primitives multiples/anatomopathologie , Phosphohydrolase PTEN/déficit , Phosphohydrolase PTEN/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Analyse de séquence d'ADN , Protéine p53 suppresseur de tumeur/déficit , Protéine p53 suppresseur de tumeur/génétique
8.
ChemSusChem ; 13(1): 267-273, 2020 Jan 09.
Article de Anglais | MEDLINE | ID: mdl-31549489

RÉSUMÉ

Solvent selection guides are crucial in chemical process design and development. Lignin from lignocellulosic biomass is a potentially attractive feedstock for sustainable chemical feedstocks. One approach would use a solvent to recover lignin prior to the traditional pulping process to make cellulose fibers: lignin value prior to pulping (LVPP). A solvent selection methodology for LVPP is presented herein that may be expanded for any proposed solvent for this process. Four screening categories are elucidated, providing metrics for 30 solvents across multiple molecular functional groups. Through performance, hazards and environment, cost and availability, and process-economics screens, the initial list of solvents is reduced to two top-tier candidates, 1,6 hexamethylenediamine and diethanolamine. 1-Methylpiperazine also emerged as a potential candidate. This solvent-selection methodology streamlines experimentation and provides promising candidates for LVPP. In addition to creating a tailored solvent selection guide, valuable biomass pretreatment data that may be utilized in different renewable applications are also presented.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...