Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Nucleic Acids Res ; 52(13): 7740-7760, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38932701

RESUMEN

Androgen receptor- (AR-) indifference is a mechanism of resistance to hormonal therapy in prostate cancer (PC). Here we demonstrate that ONECUT2 (OC2) activates resistance through multiple drivers associated with adenocarcinoma, stem-like and neuroendocrine (NE) variants. Direct OC2 gene targets include the glucocorticoid receptor (GR; NR3C1) and the NE splicing factor SRRM4, which are key drivers of lineage plasticity. Thus, OC2, despite its previously described NEPC driver function, can indirectly activate a portion of the AR cistrome through epigenetic activation of GR. Mechanisms by which OC2 regulates gene expression include promoter binding, enhancement of genome-wide chromatin accessibility, and super-enhancer reprogramming. Pharmacologic inhibition of OC2 suppresses lineage plasticity reprogramming induced by the AR signaling inhibitor enzalutamide. These results demonstrate that OC2 activation promotes a range of drug resistance mechanisms associated with treatment-emergent lineage variation in PC and support enhanced efforts to therapeutically target OC2 as a means of suppressing treatment-resistant disease.


Asunto(s)
Adenocarcinoma , Benzamidas , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Nitrilos , Neoplasias de la Próstata , Receptores Androgénicos , Receptores de Glucocorticoides , Masculino , Humanos , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/metabolismo , Adenocarcinoma/tratamiento farmacológico , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Benzamidas/farmacología , Línea Celular Tumoral , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Feniltiohidantoína/análogos & derivados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Epigénesis Genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/tratamiento farmacológico , Animales , Linaje de la Célula/genética , Ratones
3.
medRxiv ; 2024 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-38370835

RESUMEN

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.

4.
Am J Surg Pathol ; 48(4): 387-394, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38270560

RESUMEN

A well-differentiated papillary mesothelial tumor (WDPMT) and malignant mesothelioma are 2 well-recognized entities arising from the testis tunica vaginalis. Another mesothelial lesion exclusively seen at this site is mesothelioma of uncertain malignant potential (MUMP)-a lesion reminiscent of WDPMT yet demonstrating variable proportions of more complex architectural patterns that could be confused with invasion. MUMP was first described in 2010 with a total of 11 cases reported to date. Herein, we describe 19 additional patients who underwent hydrocelectomy, excision, and/or orchiectomy. Novel morphologic patterns found in addition to the 2010 series include spindle cells, keloidal-type collagen, and multicystic architecture lined by bland mesothelial cells. Clinical follow-up in 9 patients for more than 1 year (1.5 to 22.5 y, median 4.5 y) revealed no evidence of disease recurrence or metastases. Despite greater architectural complexity, MUMP has (1) bland cytology; (2) merging in with WDPMT areas; (3) low mitotic rate and Ki-67 nuclear labeling index; (4) retention of MTAP and BAP1 expression; and (5) benign clinical follow-up. If these cases were malignant mesotheliomas, one would have expected at least some of the patients to demonstrate disease recurrence/progression without adjuvant therapy within the available follow-up time, particularly with limited resection in most patients. Thus, we propose that "mesothelioma of uncertain malignant potential" be renamed as "complex mesothelial tumor of the tunica vaginalis." Using the term "complex" draws a contrast with the simple cuboidal lining and simple papillary architecture seen in WDPMT. Also, labeling the lesion as "tumor" removes the stigmata of "uncertain malignant potential" and "mesothelioma" that are alarming to patients and clinicians, and potentially could unduly lead to more extensive surgery in an attempt at "complete" resection. At the same time, not definitively labeling the lesion as benign allows recommendations for follow-up.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Neoplasias Testiculares , Masculino , Humanos , Neoplasias Testiculares/patología , Recurrencia Local de Neoplasia , Mesotelioma/cirugía , Mesotelioma/patología
5.
Clin Genitourin Cancer ; 22(2): 347-353, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38195301

RESUMEN

BACKGROUND: CD70 is commonly overexpressed in renal cell carcinoma and is minimally expressed in normal human tissue, making it a potential therapeutic target for patients with advanced renal cell carcinoma. The expression frequency of CD70 in metastatic renal cell carcinoma is not well established. MATERIALS AND METHODS: We assessed CD70 immunohistochemistry in 391 primary renal tumors and 72 metastatic renal cell carcinomas on a tissue microarray including 26 sets of paired primary and metastatic tumors. RESULTS: CD70 was frequently overexpressed in clear cell carcinoma, with a significantly lower expression rate in papillary renal cell carcinoma (P < .0001). No expression of CD70 was detected in other types of renal tumors and normal renal parenchyma. In clear cell renal cell carcinoma, CD70 expression was significantly correlated with hypoxia pathway proteins, corroborating with a recent study suggesting that CD70 is a downstream target gene of hypoxia-inducible factor. While higher expression levels were observed in males and non-Caucasians, CD70 expression was not associated with tumor grade, sarcomatoid differentiation, stage, or cancer-specific survival. Further, analysis of 26 paired primary and metastatic tumors from same individuals revealed a concordance rate of 85%. CONCLUSION: Our findings validated CD70 as a promising therapeutic target for patients with metastatic clear cell renal cell carcinoma. The utility of primary tumor tissue as surrogate samples for metastatic clear cell carcinoma awaits future CD70-targeted clinical trials.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Masculino , Humanos , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Riñón/patología , Hipoxia , Biomarcadores de Tumor , Ligando CD27/metabolismo
6.
Quant Imaging Med Surg ; 13(12): 8230-8246, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38106321

RESUMEN

Background: Deep learning has recently shown great potential in medical image reconstruction tasks. For positron emission tomography (PET) images, the direct reconstruction from raw data to radioactivity images using deep learning without any constraint may lead to the production of nonexistent structures. The aim of this study was to specifically develop and test a flexibly deep learning-based reconstruction network guided by any form of prior knowledge to achieve high quality and high reliability reconstruction. Methods: We developed a novel prior information-guided reconstruction network (PIGRN) with a dual-channel generator and a 2-scale discriminator based on a conditional generative adversarial network (cGAN). Besides the raw data channel, an additional channel is provided in the generator for prior information (PI) to guide the training phase. The PI can be reconstructed images obtained via conventional methods, nuclear medical images from other modalities, attenuation correction maps from time-of-flight-PET (TOF-PET) data, or any other physical parameters. For this study, the reconstructed images generated by filtered back projection (FBP) were chosen as the input of the additional channel. To improve the image quality, a 2-scale discriminator was adopted which can focus on both the coarse and fine field of the reconstruction images. Experiments were carried out on both a simulation dataset and a real Sprague Dawley (SD) rat dataset. Results: Two classic deep learning-based reconstruction networks, including U-Net and Deep-PET, were compared in our study. Compared with these two methods, our method could provide much higher quality PET image reconstruction in the study of the simulation dataset. The peak signal-to-noise ratio (PSNR) value reached 31.8498, and the structure similarity index measure (SSIM) value reached 0.9754. The real study on SD rats indicated that the proposed network also has strong generalization ability. Conclusions: The flexible PIGRN based on cGAN for PET images combines both raw data and PI. The results of comparison experiments and a generalization experiment based on simulation and SD rat datasets demonstrated that the proposed PIGRN has the ability to improve image quality and has strong generalization ability.

7.
bioRxiv ; 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37905039

RESUMEN

Androgen receptor- (AR-) indifference is a mechanism of resistance to hormonal therapy in prostate cancer (PC). Here we demonstrate that the HOX/CUT transcription factor ONECUT2 (OC2) activates resistance through multiple drivers associated with adenocarcinoma, stem-like and neuroendocrine (NE) variants. Direct OC2 targets include the glucocorticoid receptor and the NE splicing factor SRRM4, among others. OC2 regulates gene expression by promoter binding, enhancement of chromatin accessibility, and formation of novel super-enhancers. OC2 also activates glucuronidation genes that irreversibly disable androgen, thereby evoking phenotypic heterogeneity indirectly by hormone depletion. Pharmacologic inhibition of OC2 suppresses lineage plasticity reprogramming induced by the AR signaling inhibitor enzalutamide. These results demonstrate that OC2 activation promotes a range of drug resistance mechanisms associated with treatment-emergent lineage variation in PC. Our findings support enhanced efforts to therapeutically target this protein as a means of suppressing treatment-resistant disease.

8.
Cell Rep ; 42(10): 113221, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37815914

RESUMEN

Advanced prostate cancers are treated with therapies targeting the androgen receptor (AR) signaling pathway. While many tumors initially respond to AR inhibition, nearly all develop resistance. It is critical to understand how prostate tumor cells respond to AR inhibition in order to exploit therapy-induced phenotypes prior to the outgrowth of treatment-resistant disease. Here, we comprehensively characterize the effects of AR blockade on prostate cancer metabolism using transcriptomics, metabolomics, and bioenergetics approaches. The metabolic response to AR inhibition is defined by reduced glycolysis, robust elongation of mitochondria, and increased reliance on mitochondrial oxidative metabolism. We establish DRP1 activity and MYC signaling as mediators of AR-blockade-induced metabolic phenotypes. Rescuing DRP1 phosphorylation after AR inhibition restores mitochondrial fission, while rescuing MYC restores glycolytic activity and prevents sensitivity to complex I inhibition. Our study provides insight into the regulation of treatment-induced metabolic phenotypes and vulnerabilities in prostate cancer.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Humanos , Masculino , Andrógenos/metabolismo , Línea Celular Tumoral , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Transducción de Señal
9.
Urol Case Rep ; 45: 102218, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36248777

RESUMEN

Fibrous hamartoma of infancy (FHI) is a rare, benign soft tissue lesion observed in infants characterized histologically by triphasic appearance of bland fibroblastic fascicles, mature adipose tissue, and nodules of primitive myxoid mesenchyme. Preoperative and intraoperative recognition of FHI presents a significant diagnostic challenge due to nonspecific imaging findings and its histologic similarities to alternate benign and malignant entities. Management requires complete local excision and clinical follow-up to monitor for recurrence. Here, we present the diagnosis, management, and two-year follow-up of a 13-month-old boy with a scrotal FHI in addition to a comprehensive literature review of this entity.

10.
Oncotarget ; 13: 1054-1067, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36128328

RESUMEN

Loss-of-function mutations in genes encoding the Krebs cycle enzymes Fumarate Hydratase (FH) and Succinate Dehydrogenase (SDH) induce accumulation of fumarate and succinate, respectively and predispose patients to hereditary cancer syndromes including the development of aggressive renal cell carcinoma (RCC). Fumarate and succinate competitively inhibit αKG-dependent dioxygenases, including Lysine-specific demethylase 4A/B (KDM4A/B), leading to suppression of the homologous recombination (HR) DNA repair pathway. In this study, we have developed new syngeneic Fh1- and Sdhb-deficient murine models of RCC, which demonstrate the expected accumulation of fumarate and succinate, alterations in the transcriptomic and methylation profile, and an increase in unresolved DNA double-strand breaks (DSBs). The efficacy of poly ADP-ribose polymerase inhibitors (PARPis) and temozolomide (TMZ), alone and in combination, was evaluated both in vitro and in vivo. Combination treatment with PARPi and TMZ results in marked in vitro cytotoxicity in Fh1- and Sdhb-deficient cells. In vivo, treatment with standard dosing of the PARP inhibitor BGB-290 and low-dose TMZ significantly inhibits tumor growth without a significant increase in toxicity. These findings provide the basis for a novel therapeutic strategy exploiting HR deficiency in FH and SDH-deficient RCC with combined PARP inhibition and low-dose alkylating chemotherapy.


Asunto(s)
Carcinoma de Células Renales , Dioxigenasas , Neoplasias Renales , Adenosina Difosfato Ribosa , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Ciclo del Ácido Cítrico , ADN , Fumarato Hidratasa/genética , Fumaratos , Humanos , Histona Demetilasas con Dominio de Jumonji , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Lisina , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Succinato Deshidrogenasa/genética , Succinatos , Temozolomida/farmacología
11.
Cancer Gene Ther ; 29(10): 1463-1476, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35393570

RESUMEN

ERG translocations are commonly involved in the initiation of prostate neoplasia, yet previous experimental approaches have not addressed mechanisms of oncogenic inception. Here, in a genetically engineered mouse model, combining TMPRSS2-driven ERG with KrasG12D led to invasive prostate adenocarcinomas, while ERG or KrasG12D alone were non-oncogenic. In primary prostate luminal epithelial cells, following inducible oncogenic Kras expression or Pten depletion, TMPRSS2-ERG suppressed oncogene-induced senescence, independent of TP53 induction and RB1 inhibition. Oncogenic KRAS and TMPRSS2-ERG synergized to promote tumorigenesis and metastasis of primary luminal cells. The presence of TMPRSS2-ERG compared to a wild-type background was associated with a stemness phenotype and with relatively increased RAS-induced differential gene expression for MYC and mTOR-regulated pathways, including protein translation and lipogenesis. In addition, mTOR inhibitors abrogated ERG-dependent senescence resistance. These studies reveal a previously unappreciated function whereby ERG expression primes preneoplastic cells for the accumulation of additional gene mutations by suppression of oncogene-induced senescence.


Asunto(s)
Neoplasias de la Próstata , Proteínas Proto-Oncogénicas p21(ras) , Animales , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Proteínas de Fusión Oncogénica/genética , Oncogenes , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
12.
Ann Diagn Pathol ; 58: 151923, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35180608

RESUMEN

Yolk sac differentiation occurs in somatic neoplasms of the gastrointestinal and gynecologic tracts; it has rarely been reported in urothelial carcinoma. Here, we report three cases of yolk sac differentiation in sarcomatoid urothelial carcinoma. The epithelioid component of the sarcomatoid urothelial carcinoma showed divergent differentiation, including squamous, conventional glandular, small cell carcinoma, and yolk sac components. The sarcomatoid component showed malignant spindle cells admixed with focal chondroid and rhabdoid elements. In all three cases, the yolk sac areas were admixed with the sarcomatoid component and showed a glandular pattern, with vacuolated, eosinophilic cytoplasm. These areas were positive for SALL4, variably positive for glypican 3 and AFP, and negative for the conventional urothelial markers GATA3, p63, and 34ßE12. Yolk sac differentiation is an extremely rare occurrence in sarcomatoid urothelial carcinoma.


Asunto(s)
Carcinoma de Células Transicionales , Neoplasias de la Vejiga Urinaria , Biomarcadores de Tumor , Carcinoma de Células Transicionales/diagnóstico , Carcinoma de Células Transicionales/patología , Femenino , Humanos , Neoplasias de la Vejiga Urinaria/patología , Urotelio/patología , Saco Vitelino/patología
13.
Cancer Res ; 82(8): 1518-1533, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35131873

RESUMEN

Wnt signaling driven by genomic alterations in genes including APC and CTNNB, which encodes ß-catenin, have been implicated in prostate cancer development and progression to metastatic castration-resistant prostate cancer (mCRPC). However, nongenomic drivers and downstream effectors of Wnt signaling in prostate cancer and the therapeutic potential of targeting this pathway in prostate cancer have not been fully established. Here we analyzed Wnt/ß-catenin signaling in prostate cancer and identified effectors distinct from those found in other tissues, including aryl hydrocarbon receptor and RUNX1, which are linked to stem cell maintenance, and ROR1, a noncanonical Wnt5a coreceptor. Wnt/ß-catenin signaling-mediated increases in ROR1 enhanced noncanonical responses to Wnt5a. Regarding upstream drivers, APC genomic loss, but not its epigenetic downregulation commonly observed in prostate cancer, was strongly associated with Wnt/ß-catenin pathway activation in clinical samples. Tumor cell upregulation of the Wnt transporter Wntless (WLS) was strongly associated with Wnt/ß-catenin pathway activity in primary prostate cancer but also associated with both canonical and noncanonical Wnt signaling in mCRPC. IHC confirmed tumor cell WLS expression in primary prostate cancer and mCRPC, and patient-derived prostate cancer xenografts expressing WLS were responsive to treatment with Wnt synthesis inhibitor ETC-1922159. These findings reveal that Wnt/ß-catenin signaling in prostate cancer drives stem cell maintenance and invasion and primes for noncanonical Wnt signaling through ROR1. They further show that autocrine Wnt production is a nongenomic driver of canonical and noncanonical Wnt signaling in prostate cancer, which can be targeted with Wnt synthesis inhibitors to suppress tumor growth. SIGNIFICANCE: This work provides fundamental insights into Wnt signaling and prostate cancer cell biology and indicates that a subset of prostate cancer driven by autocrine Wnt signaling is sensitive to Wnt synthesis inhibitors.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Vía de Señalización Wnt , Comunicación Autocrina , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
14.
Case Rep Urol ; 2022: 3353268, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35132367

RESUMEN

Cancer is a significant cause of morbidity and mortality in recipients of renal transplantation. The vast majority develop from recipient origins, whereas donor-derived malignancies are exceedingly rare. We report 2 cases of poorly differentiated donor-derived urothelial carcinoma (UC) in renal transplantation recipients. The first patient underwent a living-related-donor renal transplantation 24 years prior and presented with back pain, hematuria, and rising creatinine and was found to have a 14 cm mass in the renal allograft with regional lymphadenopathy and liver metastases. Pathology showed UC with small-cell differentiation. The second patient presented with hematuria and rising creatinine and was initially found to have muscle invasive bladder cancer seven years after a deceased donor renal transplantation. Nine months after radical cystectomy, a large 9 cm mass was found on his allograft, for which radical nephrectomy and excision of prior ileal conduit was performed. Pathology showed UC with sarcomatoid differentiation. Short tandem repeat (STR) genotyping confirmed donor-derived origins. Both patient tumors expressed PD-L1 suggesting an additional therapeutic avenue for these rare tumors.

15.
JCI Insight ; 7(4)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35050902

RESUMEN

BACKGROUNDProstate cancer is multifocal with distinct molecular subtypes. The utility of genomic subtyping has been challenged due to inter- and intrafocal heterogeneity. We sought to characterize the subtype-defining molecular alterations of primary prostate cancer across all tumor foci within radical prostatectomy (RP) specimens and determine the prevalence of collision tumors.METHODSFrom the Early Detection Research Network cohort, we identified 333 prospectively collected RPs from 2010 to 2014 and assessed ETS-related gene (ERG), serine peptidase inhibitor Kazal type 1 (SPINK1), phosphatase and tensin homolog (PTEN), and speckle type BTB/POZ protein (SPOP) molecular status. We utilized dual ERG/SPINK1 immunohistochemistry and fluorescence in situ hybridization to confirm ERG rearrangements and characterize PTEN deletion, as well as high-resolution melting curve analysis and Sanger sequencing to determine SPOP mutation status.RESULTSBased on index focus alone, ERG, SPINK1, PTEN, and SPOP alterations were identified in 47.5%, 10.8%, 14.3%, and 5.1% of RP specimens, respectively. In 233 multifocal RPs with ERG/SPINK1 status in all foci, 139 (59.7%) had discordant molecular alterations between foci. Collision tumors, as defined by discrepant ERG/SPINK1 status within a single focus, were identified in 29 (9.4%) RP specimens.CONCLUSIONInterfocal molecular heterogeneity was identified in about 60% of multifocal RP specimens, and collision tumors were present in about 10%. We present this phenomenon as a model for the intrafocal heterogeneity observed in previous studies and propose that future genomic studies screen for collision tumors to better characterize molecular heterogeneity.FUNDINGEarly Detection Research Network US National Cancer Institute (NCI) 5U01 CA111275-09, Center for Translational Pathology at Weill Cornell Medicine (WCM) Department of Pathology and Laboratory Medicine, US NCI (WCM SPORE in Prostate Cancer, P50CA211024-01), R37CA215040, Damon Runyon Cancer Research Foundation, US MetLife Foundation Family Clinical Investigator Award, Norwegian Cancer Society (grant 208197), and South-Eastern Norway Regional Health Authority (grant 2019016 and 2020063).


Asunto(s)
Mutación , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/genética , ARN Neoplásico/genética , Proteínas Represoras/genética , Inhibidor de Tripsina Pancreática de Kazal/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Análisis Mutacional de ADN , Reordenamiento Génico , Humanos , Inmunohistoquímica , Masculino , Proteínas Nucleares/biosíntesis , Fosfohidrolasa PTEN/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Represoras/biosíntesis , Estudios Retrospectivos , Inhibidor de Tripsina Pancreática de Kazal/biosíntesis , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
16.
Prostate Cancer Prostatic Dis ; 25(3): 539-546, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35075215

RESUMEN

BACKGROUND: The antiprostate cancer effects of dietary ω-3 fatty acids (FAs) were previously found to be dependent on host G-protein coupled receptor 120 (GPR120). Using an orthotopic tumor model and an ex-vivo model of bone marrow derived M2-like macrophages, we sought to determine if ω-3 FAs inhibit angiogenesis and activate T-cells, and if these effects are dependent on GPR120. METHODS: Gausia luciferase labeled MycCaP prostate cancer cells (MycCaP-Gluc) were injected into the anterior prostate lobe of FVB mice. After established tumors were confirmed by blood luminescence, mice were fed an ω-3 or ω-6 diet. Five weeks after tumor injection, tumor weight, immune cell infiltration and markers of angiogenesis were determined. An ex-vivo co-culture model of bone marrow derived M2-like macrophages from wild-type or GPR120 knockout mice with MycCap prostate cancer cells was used to determine if docosahexanoic acid (DHA, ω-3 FA) inhibition of angiogenesis and T-cell activation is dependent on macrophage GPR120. RESULTS: Feeding an ω-3 diet significantly reduced orthotopic MycCaP-Gluc tumor growth relative to an ω-6 diet. Tumors from the ω-3 group had decreased M2-like macrophage infiltration and decreased expression of angiogenesis factors. DHA significantly inhibited M2 macrophage-induced endothelial tube formation and reversed M2 macrophage-induced T-cell suppression, and these DHA effects were mediated, in part, by M2 macrophage GPR120. CONCLUSION: Omega-3 FAs delayed orthotopic tumor growth, inhibited M2-like macrophage tumor infiltration, and inhibited M2-like macrophage-induced angiogenesis and T-cell suppression. Given the central role of M2-like macrophages in prostate cancer progression, GPR120-dependent ω-3 FA inhibition of M2-like macrophages may play an important role in prostate cancer therapeutics.


Asunto(s)
Ácidos Grasos Omega-3 , Neoplasias de la Próstata , Receptores Acoplados a Proteínas G , Animales , Dieta , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-3/farmacología , Humanos , Activación de Linfocitos , Masculino , Ratones , Próstata/patología , Neoplasias de la Próstata/patología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Linfocitos T , Macrófagos Asociados a Tumores
17.
Cancers (Basel) ; 13(19)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34638229

RESUMEN

Magnetic resonance imaging (MRI) has enabled non-invasive cancer diagnosis, monitoring, and management in common clinical settings. However, inadequate quantitative analyses in MRI continue to limit its full potential and these often have an impact on clinicians' judgments. Magnetic resonance fingerprinting (MRF) has recently been introduced to acquire multiple quantitative parameters simultaneously in a reasonable timeframe. Initial retrospective studies have demonstrated the feasibility of using MRF for various cancer characterizations. Further trials with larger cohorts are still needed to explore the repeatability and reproducibility of the data acquired by MRF. At the moment, technical difficulties such as undesirable processing time or lack of motion robustness are limiting further implementations of MRF in clinical oncology. This review summarises the latest findings and technology developments for the use of MRF in cancer management and suggests possible future implications of MRF in characterizing tumour heterogeneity and response assessment.

19.
Clin Cancer Res ; 27(17): 4836-4847, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34168052

RESUMEN

PURPOSE: A subset of primary prostate cancer expresses programmed death-ligand 1 (PD-L1), but whether they have a unique tumor immune microenvironment or genomic features is unclear. EXPERIMENTAL DESIGN: We selected PD-L1-positive high-grade and/or high-risk primary prostate cancer, characterized tumor-infiltrating lymphocytes with multiplex immunofluorescence, and identified genomic alterations in immunogenic and nonimmunogenic tumor foci. RESULTS: One quarter of aggressive localized prostate cancer cases (29/115) had tumor PD-L1 expression more than 5%. This correlated with increased density of CD8+ T cells, a large fraction coexpressing PD-1, versus absent PD-1 expression on sparse CD8 T cells in unselected cases. Most CD8+PD-1+ cells did not express terminal exhaustion markers (TIM3 or LAG3), while a subset expressed TCF1. Consistent with these CD8+PD-1+TCF1+ cells being progenitors, they were found in antigen-presenting cell niches in close proximity to MHC-II+ cells. CD8 T-cell density in immunogenic prostate cancer and renal cell carcinoma (RCC) was nearly identical. Shallow RB1 and BRCA2 losses, and deep deletions of CHD1, were prevalent, the latter being strongly associated with a dendritic cell gene set in The Cancer Genome Atlas. Tumor mutation burden was variable; neither high microsatellite instability nor CDK12 alterations were present. CONCLUSIONS: A subset of localized prostate cancer is immunogenic, manifested by PD-L1 expression and CD8+ T-cell content comparable with RCC. The CD8+ T cells include effector cells and exhausted progenitor cells, which may be expanded by immune checkpoint inhibitors (ICI). Genomic losses of RB1, BRCA2, and CHD1 may be drivers of this phenotype. These findings indicate that immunotherapies may be effective in biomarker-selected subpopulations of patients with localized prostate cancer.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias de la Próstata , Antígeno B7-H1/genética , Genes Supresores de Tumor , Humanos , Linfocitos Infiltrantes de Tumor , Masculino , Fenotipo , Neoplasias de la Próstata/genética , Microambiente Tumoral/genética
20.
Mol Oncol ; 15(7): 1901-1920, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33932081

RESUMEN

Androgen receptor (AR) is the principal molecule in prostate cancer (PCa) etiology and therapy. AR re-activation still remains a major challenge during treatment of castration-resistant prostate cancer (CRPC) tumors that relapse after castration therapies. Recent reports have indicated the enrichment of Ser81-phosphorylated AR (pS81) in the nucleus of CRPC cells, and CDK1 and CDK9 as the kinases phosphorylating AR at S81. In the current study we showed that pS81 is preferentially localized in the nucleus in both rapid biopsy metastatic CRPC samples and PCa xenografts, and nuclear pS81 localization is correlated with AR transactivation in tumor xenografts. Chromatin immunoprecipitation (ChIP) analysis demonstrated an alignment of S81 phosphorylation and AR-mediated transactivation with the chromatin locus openness. Moreover, pS81-specific ChIP-Seq showed a disproportional occupancy of pS81 on AR-activated promoters, while 3C-ChIP assays further indicated an enrichment of pS81 at the PSA enhancer-promoter loop, a known AR activating hub. In the latter, CDK9 was shown to modulate the transactivation of the AR and RNA Pol II. Indeed, ChIP and re-ChIP assays also confirmed that AR-dependent activation of the PSA enhancer and promoter mediated by pS81 was coupled with activation of Pol II and the pTEFb complex. Mechanistically, we determined that CDK1 and CDK9 sustained the pS81 AR modification in the soluble and chromatin-bound fractions of PCa cells, respectively. Finally, we demonstrated that CDK1 activity was maintained throughout the cell cycle, and that CDK1 inhibitors restored androgen sensitivity in CRPC tumor cells. Based on these findings, CDK1 and CDK9 could be targeted as pS81 kinases in patients with CRPC, either alone or in conjunction with direct AR antagonists.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Humanos , Masculino , Fosforilación , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA