Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
NEJM AI ; 1(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-39131700

RESUMEN

BACKGROUND: As artificial intelligence (AI) tools become widely accessible, more patients and medical professionals will turn to them for medical information. Large language models (LLMs), a subset of AI, excel in natural language processing tasks and hold considerable promise for clinical use. Fields such as oncology, in which clinical decisions are highly dependent on a continuous influx of new clinical trial data and evolving guidelines, stand to gain immensely from such advancements. It is therefore of critical importance to benchmark these models and describe their performance characteristics to guide their safe application to clinical oncology. Accordingly, the primary objectives of this work were to conduct comprehensive evaluations of LLMs in the field of oncology and to identify and characterize strategies that medical professionals can use to bolster their confidence in a model's response. METHODS: This study tested five publicly available LLMs (LLaMA 1, PaLM 2, Claude-v1, generative pretrained transformer 3.5 [GPT-3.5], and GPT-4) on a comprehensive battery of 2044 oncology questions, including topics from medical oncology, surgical oncology, radiation oncology, medical statistics, medical physics, and cancer biology. Model prompts were presented independently of each other, and each prompt was repeated three times to assess output consistency. For each response, models were instructed to provide a self-appraised confidence score (from 1 to 4). Model performance was also evaluated against a novel validation set comprising 50 oncology questions curated to eliminate any risk of overlap with the data used to train the LLMs. RESULTS: There was significant heterogeneity in performance between models (analysis of variance, P<0.001). Relative to a human benchmark (2013 and 2014 examination results), GPT-4 was the only model to perform above the 50th percentile. Overall, model performance varied as a function of subject area across all models, with worse performance observed in clinical oncology subcategories compared with foundational topics (medical statistics, medical physics, and cancer biology). Within the clinical oncology subdomain, worse performance was observed in female-predominant malignancies. A combination of model selection, prompt repetition, and confidence self-appraisal allowed for the identification of high-performing subgroups of questions with observed accuracies of 81.7 and 81.1% in the Claude-v1 and GPT-4 models, respectively. Evaluation of the novel validation question set produced similar trends in model performance while also highlighting improved performance in newer, centrally hosted models (GPT-4 Turbo and Gemini 1.0 Ultra) and local models (Mixtral 8×7B and LLaMA 2). CONCLUSIONS: Of the models tested on a standardized set of oncology questions, GPT-4 was observed to have the highest performance. Although this performance is impressive, all LLMs continue to have clinically significant error rates, including examples of overconfidence and consistent inaccuracies. Given the enthusiasm to integrate these new implementations of AI into clinical practice, continued standardized evaluations of the strengths and limitations of these products will be critical to guide both patients and medical professionals. (Funded by the National Institutes of Health Clinical Center for Research and the Intramural Research Program of the National Institutes of Health; Z99 CA999999.).

2.
Nat Genet ; 56(8): 1689-1700, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39020220

RESUMEN

The impact of variations in the three-dimensional structure of the genome has been recognized, but solid cancer tissue studies are limited. Here, we performed integrated deep Hi-C sequencing with matched whole-genome sequencing, whole-genome bisulfite sequencing, 5-hydroxymethylcytosine (5hmC) sequencing and RNA sequencing across a cohort of 80 biopsy samples from patients with metastatic castration-resistant prostate cancer. Dramatic differences were present in gene expression, 5-methylcytosine/5hmC methylation and in structural variation versus mutation rate between A and B (open and closed) chromatin compartments. A subset of tumors exhibited depleted regional chromatin contacts at the AR locus, linked to extrachromosomal circular DNA (ecDNA) and worse response to AR signaling inhibitors. We also identified topological subtypes associated with stark differences in methylation structure, gene expression and prognosis. Our data suggested that DNA interactions may predispose to structural variant formation, exemplified by the recurrent TMPRSS2-ERG fusion. This comprehensive integrated sequencing effort represents a unique clinical tumor resource.


Asunto(s)
5-Metilcitosina , Metilación de ADN , Humanos , Masculino , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Regulación Neoplásica de la Expresión Génica , Epigenómica/métodos , Metástasis de la Neoplasia/genética , Genoma Humano , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Epigénesis Genética , Receptores Androgénicos/genética , Cromatina/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas de Fusión Oncogénica/genética , ADN/genética , Secuenciación Completa del Genoma , ARN/genética , Pronóstico
3.
bioRxiv ; 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38895460

RESUMEN

Background: Prostate cancer is a heterogenous disease, but once it becomes metastatic it eventually becomes treatment resistant. One mechanism of resistance to AR-targeting therapy is lineage plasticity, where the tumor undergoes a transformation to an AR-indifferent phenotype, most studied in the context of neuroendocrine prostate cancer (NEPC). However, activation of additional de- or trans-differentiation programs, including a gastrointestinal (GI) gene expression program, has been suggested as an alternative method of resistance. In this study, we explored the previously identified GI prostate cancer phenotype (PCa-GI) in a large cohort of metastatic castration-resistant prostate cancer (mCRPC) patient biopsy samples. Methods: We analyzed a dataset of 634 mCRPC samples with batch effect corrected gene expression data from the West Coast Dream Team (WCDT), the East Coast Dream Team (ECDT), the Fred Hutchinson Cancer Research Center (FHCRC) and the Weill Cornell Medical center (WCM). Survival data was available from the WCDT and ECDT cohorts. We calculated a gene expression GI score using the sum of z-scores of genes from a published set of PCa-GI-defining genes (N=38). Survival analysis was performed using the Kaplan-Meier method and Cox proportional hazards regression with endpoint overall survival from time of biopsy to death of any cause. Results: We found that the PCa-GI score had a bimodal distribution, identifying a distinct set of tumors with an activated GI expression pattern. Approximately 35% of samples were classified as PCa-GI high, which was concordant with prior reports. Liver metastases had the highest median score but after excluding liver samples, 29% of the remaining samples were still classified as PCa-GI high, suggesting a distinct phenotype not exclusive to liver metastases. No correlation was observed between GI score and proliferation, AR signaling, or NEPC scores. Furthermore, the PCa-GI score was not associated with genomic alterations in AR, FOXA1, RB1, TP53 or PTEN. However, tumors with MYC amplifications showed significantly higher GI scores (p=0.0001). Patients with PCa-GI tumors had a shorter survival (HR=1.5 [1.1-2.1], p=0.02), but this result was not significant after adjusting for the liver as metastatic site (HR=1.2 [0.82-1.7], p=0.35). Patients with PCa-GI low samples had a better outcome after androgen receptor signaling inhibitors (ASI, abiraterone or enzalutamide) than other therapies (HR=0.37 [0.22-0.61], p=0.0001) while the benefit of ASI was smaller and non-significant for PCa-GI high samples (HR=0.55 [0.29-1.1], p=0.07). A differential pathway analysis identified FOXA2 signaling to be upregulated PCa-GI high tumors (FDR = 3.7 × 10-13). Conclusions: The PCa-GI phenotype is prevalent in clinical mCRPC samples and may represent a distinct biological entity. PCa-GI tumors may respond less to ASI and could offer a strategy to study novel therapeutic targets.

4.
Cancer Res Commun ; 4(6): 1481-1494, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38747612

RESUMEN

Cancer-associated fibroblasts (CAF) are a prominent cell type within the tumor microenvironment (TME) where they are known to promote cancer cell growth and survival, angiogenesis, drug resistance, and immunosuppression. The transmembrane prolyl protease fibroblast activation protein (FAP) is expressed on the surface of highly protumorigenic CAFs found in the stroma of nearly every cancer of epithelial origin. The widespread expression of FAP has made it an attractive therapeutic target based on the underlying hypothesis that eliminating protumorigenic CAFs will disrupt the cross-talk between components of TME resulting in cancer cell death and immune infiltration. This hypothesis, however, has never been directly proven. To eliminate FAP-expressing CAFs, we developed an antibody-drug conjugate using our anti-FAP antibody, huB12, coupled to a monomethyl auristatin E (huB12-MMAE) payload. After determining that huB12 was an effective targeting vector, we found that huB12-MMAE potently eliminated FAP-expressing cells as monocultures in vitro and significantly prolonged survival in vivo using a xenograft engineered to overexpress FAP. We investigated the effects of selectively eliminating CAFs using a layered, open microfluidic cell coculture platform, known as the Stacks. Analysis of mRNA and protein expression found that treatment with huB12-MMAE resulted in the increased secretion of the proinflammatory cytokines IL6 and IL8 by CAFs and an associated increase in expression of proinflammatory genes in cancer cells. We also detected increased secretion of CSF1, a cytokine involved in myeloid recruitment and differentiation. Our findings suggest that the mechanism of FAP-targeted therapies is through effects on the immune microenvironment and antitumor immune response. SIGNIFICANCE: The direct elimination of FAP-expressing CAFs disrupts the cross-talk with cancer cells leading to a proinflammatory response and alterations in the immune microenvironment and antitumor immune response.


Asunto(s)
Fibroblastos Asociados al Cáncer , Endopeptidasas , Inmunoconjugados , Microambiente Tumoral , Humanos , Animales , Inmunoconjugados/farmacología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Fibroblastos Asociados al Cáncer/inmunología , Ratones , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Endopeptidasas/genética , Endopeptidasas/metabolismo , Línea Celular Tumoral , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gelatinasas/metabolismo , Gelatinasas/genética , Oligopéptidos/farmacología , Femenino
5.
Commun Biol ; 7(1): 314, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480799

RESUMEN

Histopathologic diagnosis and classification of cancer plays a critical role in guiding treatment. Advances in next-generation sequencing have ushered in new complementary molecular frameworks. However, existing approaches do not independently assess both site-of-origin (e.g. prostate) and lineage (e.g. adenocarcinoma) and have minimal validation in metastatic disease, where classification is more difficult. Utilizing gradient-boosted machine learning, we developed ATLAS, a pair of separate AI Tumor Lineage and Site-of-origin models from RNA expression data on 8249 tumor samples. We assessed performance independently in 10,376 total tumor samples, including 1490 metastatic samples, achieving an accuracy of 91.4% for cancer site-of-origin and 97.1% for cancer lineage. High confidence predictions (encompassing the majority of cases) were accurate 98-99% of the time in both localized and remarkably even in metastatic samples. We also identified emergent properties of our lineage scores for tumor types on which the model was never trained (zero-shot learning). Adenocarcinoma/sarcoma lineage scores differentiated epithelioid from biphasic/sarcomatoid mesothelioma. Also, predicted lineage de-differentiation identified neuroendocrine/small cell tumors and was associated with poor outcomes across tumor types. Our platform-independent single-sample approach can be easily translated to existing RNA-seq platforms. ATLAS can complement and guide traditional histopathologic assessment in challenging situations and tumors of unknown primary.


Asunto(s)
Adenocarcinoma , Mesotelioma Maligno , Tumores Neuroendocrinos , Masculino , Humanos , Aprendizaje Automático , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética
6.
Cancer Discov ; 14(1): 158-175, 2024 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-37902550

RESUMEN

How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a guanine nucleotide-binding protein, which promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes nonhomologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard-of-care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in nonmalignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment. SIGNIFICANCE: A newly described GTP-dependent signaling axis is an unexpected link between nucleotide metabolism and DNA repair. Disrupting this pathway can overcome cancer resistance to genotoxic therapy while augmenting it can mitigate genotoxic injury of normal tissues. This article is featured in Selected Articles from This Issue, p. 5.


Asunto(s)
Glioblastoma , Transducción de Señal , Humanos , Ratones , Animales , Transducción de Señal/genética , Reparación del ADN , Daño del ADN , Guanosina Trifosfato
7.
NAR Cancer ; 5(3): zcad045, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37636316

RESUMEN

Androgen receptor (AR) inhibition is standard of care for advanced prostate cancer (PC). However, efficacy is limited by progression to castration-resistant PC (CRPC), usually due to AR re-activation via mechanisms that include AR amplification and structural rearrangement. These two classes of AR alterations often co-occur in CRPC tumors, but it is unclear whether this reflects intercellular or intracellular heterogeneity of AR. Resolving this is important for developing new therapies and predictive biomarkers. Here, we analyzed 41 CRPC tumors and 6 patient-derived xenografts (PDXs) using linked-read DNA-sequencing, and identified 7 tumors that developed complex, multiply-rearranged AR gene structures in conjunction with very high AR copy number. Analysis of PDX models by optical genome mapping and fluorescence in situ hybridization showed that AR residing on extrachromosomal DNA (ecDNA) was an underlying mechanism, and was associated with elevated levels and diversity of AR expression. This study identifies co-evolution of AR gene copy number and structural complexity via ecDNA as a mechanism associated with endocrine therapy resistance.

9.
Semin Radiat Oncol ; 33(3): 243-251, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37331779

RESUMEN

Developing radiation tumor biomarkers that can guide personalized radiotherapy clinical decision making is a critical goal in the effort towards precision cancer medicine. High-throughput molecular assays paired with modern computational techniques have the potential to identify individual tumor-specific signatures and create tools that can help understand heterogenous patient outcomes in response to radiotherapy, allowing clinicians to fully benefit from the technological advances in molecular profiling and computational biology including machine learning. However, the increasingly complex nature of the data generated from high-throughput and "omics" assays require careful selection of analytical strategies. Furthermore, the power of modern machine learning techniques to detect subtle data patterns comes with special considerations to ensure that the results are generalizable. Herein, we review the computational framework of tumor biomarker development and describe commonly used machine learning approaches and how they are applied for radiation biomarker development using molecular data, as well as challenges and emerging research trends.


Asunto(s)
Biomarcadores de Tumor , Neoplasias , Humanos , Aprendizaje Automático , Biomarcadores , Medicina de Precisión/métodos , Neoplasias/genética , Neoplasias/radioterapia , Toma de Decisiones Clínicas
10.
bioRxiv ; 2023 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-37090571

RESUMEN

How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a G protein, that promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes non-homologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard of care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in non-malignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment.

11.
Clin Cancer Res ; 29(12): 2324-2335, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36939530

RESUMEN

PURPOSE: Men with metastatic castration-resistant prostate cancer (mCRPC) frequently develop resistance to androgen receptor signaling inhibitor (ARSI) treatment; therefore, new therapies are needed. Trophoblastic cell-surface antigen (TROP-2) is a transmembrane protein identified in prostate cancer and overexpressed in multiple malignancies. TROP-2 is a therapeutic target for antibody-drug conjugates (ADC). EXPERIMENTAL DESIGN: TROP-2 gene (TACSTD2) expression and markers of treatment resistance from prostate biopsies were analyzed using data from four previously curated cohorts of mCRPC (n = 634) and the PROMOTE study (dbGaP accession phs001141.v1.p1, n = 88). EPCAM or TROP-2-positive circulating tumor cells (CTC) were captured from peripheral blood for comparison of protein (n = 15) and gene expression signatures of treatment resistance (n = 40). We assessed the efficacy of TROP-2-targeting agents in a mouse xenograft model generated from prostate cancer cell lines. RESULTS: We demonstrated that TACSTD2 is expressed in mCRPC from luminal and basal tumors but at lower levels in patients with neuroendocrine prostate cancer. Patients previously treated with ARSI showed no significant difference in TACSTD2 expression, whereas patients with detectable AR-V7 expression showed increased expression. We observed that TROP-2 can serve as a cell surface target for isolating CTCs, which may serve as a predictive biomarker for ADCs. We also demonstrated that prostate cancer cell line xenografts can be targeted specifically by labeled anti-TROP-2 agents in vivo. CONCLUSIONS: These results support further studies on TROP-2 as a therapeutic and diagnostic target for mCRPC.


Asunto(s)
Células Neoplásicas Circulantes , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Animales , Ratones , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Receptores Androgénicos/genética , Isoformas de Proteínas/genética , Células Neoplásicas Circulantes/patología , Antagonistas de Receptores Androgénicos/farmacología
12.
J Clin Invest ; 132(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36317634

RESUMEN

BackgroundNeuroendocrine prostate cancer (NEPC) is an aggressive subtype, the presence of which changes the prognosis and management of metastatic prostate cancer.MethodsWe performed analytical validation of a Circulating Tumor Cell (CTC) multiplex RNA qPCR assay to identify the limit of quantification (LOQ) in cell lines, synthetic cDNA, and patient samples. We next profiled 116 longitudinal samples from a prospectively collected institutional cohort of 17 patients with metastatic prostate cancer (7 NEPC, 10 adenocarcinoma) as well as 265 samples from 139 patients enrolled in 3 adenocarcinoma phase II trials of androgen receptor signaling inhibitors (ARSIs). We assessed a NEPC liquid biomarker via the presence of neuroendocrine markers and the absence of androgen receptor (AR) target genes.ResultsUsing the analytical validation LOQ, liquid biomarker NEPC detection in the longitudinal cohort had a per-sample sensitivity of 51.35% and a specificity of 91.14%. However, when we incorporated the serial information from multiple liquid biopsies per patient, a unique aspect of this study, the per-patient predictions were 100% accurate, with a receiver-operating-curve (ROC) AUC of 1. In the adenocarcinoma ARSI trials, the presence of neuroendocrine markers, even while AR target gene expression was retained, was a strong negative prognostic factor.ConclusionOur analytically validated CTC biomarker can detect NEPC with high diagnostic accuracy when leveraging serial samples that are only feasible using liquid biopsies. Patients with expression of NE genes while retaining AR-target gene expression may indicate the transition to neuroendocrine differentiation, with clinical characteristics consistent with this phenotype.FundingNIH (DP2 OD030734, 1UH2CA260389, R01CA247479, and P30 CA014520), Department of Defense (PC190039 and PC200334), and Prostate Cancer Foundation (Movember Foundation - PCF Challenge Award).


Asunto(s)
Adenocarcinoma , Neoplasias de la Próstata , Humanos , Masculino , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/patología , Biomarcadores , Transducción de Señal , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
13.
NPJ Genom Med ; 7(1): 58, 2022 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-36253482

RESUMEN

DNA mutations in specific genes can confer preferential benefit from drugs targeting those genes. However, other molecular perturbations can "phenocopy" pathogenic mutations, but would not be identified using standard clinical sequencing, leading to missed opportunities for other patients to benefit from targeted treatments. We hypothesized that RNA phenocopy signatures of key cancer driver gene mutations could improve our ability to predict response to targeted therapies, despite not being directly trained on drug response. To test this, we built gene expression signatures in tissue samples for specific mutations and found that phenocopy signatures broadly increased accuracy of drug response predictions in-vitro compared to DNA mutation alone, and identified additional cancer cell lines that respond well with a positive/negative predictive value on par or better than DNA mutations. We further validated our results across four clinical cohorts. Our results suggest that routine RNA sequencing of tumors to identify phenocopies in addition to standard targeted DNA sequencing would improve our ability to accurately select patients for targeted therapies in the clinic.

14.
Clin Cancer Res ; 28(24): 5396-5404, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36260524

RESUMEN

PURPOSE: Although numerous biology-driven subtypes have been described previously in metastatic castration-resistant prostate cancer (mCRPC), unsupervised molecular subtyping based on gene expression has been less studied, especially using large cohorts. Thus, we sought to identify the intrinsic molecular subtypes of mCRPC and assess molecular and clinical correlates in the largest combined cohort of mCRPC samples with gene expression data available to date. EXPERIMENTAL DESIGN: We combined and batch-effect corrected gene expression data from four mCRPC cohorts from the Fred Hutchinson Cancer Research Center (N = 157), a small-cell neuroendocrine (NE) prostate cancer (SCNC)-enriched cohort from Weill Cornell Medicine (N = 49), and cohorts from the Stand Up 2 Cancer/Prostate Cancer Foundation East Coast Dream Team (N = 266) and the West Coast Dream Team (N = 162). RESULTS: Hierarchical clustering of RNA-sequencing data from these 634 mCRPC samples identified two distinct adenocarcinoma subtypes, one of which (adeno-immune) was characterized by higher gene expression of immune pathways, higher CIBERSORTx immune scores, diminished ASI benefit, and non-lymph node metastasis tropism compared with an adeno-classic subtype. We also identified two distinct subtypes with enrichment for an NE phenotype, including an NE-liver subgroup characterized by liver metastasis tropism, PTEN loss, and APC and SPOP mutations compared with an NE-classic subgroup. CONCLUSIONS: Our results emphasize the heterogeneity of mCRPC beyond currently accepted molecular phenotypes, and suggest that future studies should consider incorporating transcriptome-wide profiling to better understand how these differences impact treatment responses and outcomes.


Asunto(s)
Adenocarcinoma , Neoplasias de la Próstata Resistentes a la Castración , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Perfilación de la Expresión Génica , Proteínas Nucleares/genética , Proteínas Represoras/genética
15.
Cancer Res ; 82(21): 3888-3902, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36251389

RESUMEN

Analysis of DNA methylation is a valuable tool to understand disease progression and is increasingly being used to create diagnostic and prognostic clinical biomarkers. While conversion of cytosine to 5-methylcytosine (5mC) commonly results in transcriptional repression, further conversion to 5-hydroxymethylcytosine (5hmC) is associated with transcriptional activation. Here we perform the first study integrating whole-genome 5hmC with DNA, 5mC, and transcriptome sequencing in clinical samples of benign, localized, and advanced prostate cancer. 5hmC is shown to mark activation of cancer drivers and downstream targets. Furthermore, 5hmC sequencing revealed profoundly altered cell states throughout the disease course, characterized by increased proliferation, oncogenic signaling, dedifferentiation, and lineage plasticity to neuroendocrine and gastrointestinal lineages. Finally, 5hmC sequencing of cell-free DNA from patients with metastatic disease proved useful as a prognostic biomarker able to identify an aggressive subtype of prostate cancer using the genes TOP2A and EZH2, previously only detectable by transcriptomic analysis of solid tumor biopsies. Overall, these findings reveal that 5hmC marks epigenomic activation in prostate cancer and identify hallmarks of prostate cancer progression with potential as biomarkers of aggressive disease. SIGNIFICANCE: In prostate cancer, 5-hydroxymethylcytosine delineates oncogene activation and stage-specific cell states and can be analyzed in liquid biopsies to detect cancer phenotypes. See related article by Wu and Attard, p. 3880.


Asunto(s)
5-Metilcitosina , Neoplasias de la Próstata , Masculino , Humanos , Próstata , Biopsia
16.
Adv Radiat Oncol ; 7(3): 100884, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35647405

RESUMEN

Purpose: Our purpose was to determine whether bone density and bone-derived radiomic metrics in combination with dosimetric variables could improve risk stratification of rib fractures after stereotactic body radiation therapy (SBRT) for early-stage non-small cell lung cancer (NSCLC). Methods and Materials: A retrospective analysis was conducted of patients with early-stage NSCLC treated with SBRT. Dosimetric data and rib radiomic data extracted using PyRadiomics were used for the analysis. A subset of patients had bone density scans that were used to create a predicted bone density score for all patients. A 10-fold cross validated approach with 10 resamples was used to find the top univariate logistic models and elastic net regression models that predicted for rib fracture. Results: A total of 192 treatment plans were included in the study with a rib fracture rate of 16.1%. A predicted bone density score was created from a multivariate model with vertebral body Hounsfield units and patient weight, with an R-squared of 0.518 compared with patient dual-energy x-ray absorptiometry T-scores. When analyzing all patients, a low predicted bone density score approached significance for increased risk of rib fracture (P = .07). On competing risk analysis, when stratifying patients based on chest wall V30 Gy and bone density score, those with a V30 Gy ≥30 cc and a low bone density score had a significantly higher risk of rib fracture compared with all other patients (P < .001), with a predicted 2-year risk of rib fracture of 28.6% (95% confidence interval, 17.2%-41.1%) and 4.9% (95% confidence interval, 2.3%-9.0%), respectively. Dosimetric variables were the primary drivers of fracture risk. A multivariate elastic net regression model including all dosimetric variables was the best predictor of rib fracture (area under the curve [AUC], 0.864). Bone density variables (AUC, 0.618) and radiomic variables (AUC, 0.617) have better predictive power than clinical variables that exclude bone density (AUC, 0.538). Conclusion: Radiomic features, including a bone density score that includes vertebral body Hounsfield units and radiomic signatures from the ribs, can be used to stratify risk of rib fracture after SBRT for NSCLC.

17.
J Clin Oncol ; 40(31): 3633-3641, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35617646

RESUMEN

PURPOSE: Liquid biopsies in metastatic renal cell carcinoma (mRCC) provide a unique approach to understand the molecular basis of treatment response and resistance. This is particularly important in the context of immunotherapies, which target key immune-tumor interactions. Unlike metastatic tissue biopsies, serial real-time profiling of mRCC is feasible with our noninvasive circulating tumor cell (CTC) approach. METHODS: We collected 457 longitudinal liquid biopsies from 104 patients with mRCC enrolled in one of two studies, either a prospective cohort or a phase II multicenter adaptive immunotherapy trial. Using a novel CTC capture and automated microscopy platform, we profiled CTC enumeration and expression of HLA I and programmed cell death-ligand 1 (PD-L1). Given their diametric immunological roles, we focused on the HLA I to PD-L1 ratio (HP ratio). RESULTS: Patients with radiographic responses showed significantly lower CTC abundances throughout treatment. Furthermore, patients whose CTC enumeration trajectory was in the highest quartile (> 0.12 CTCs/mL annually) had shorter overall survival (median 17.0 months v 21.1 months, P < .001). Throughout treatment, the HP ratio decreased in patients receiving immunotherapy but not in patients receiving tyrosine kinase inhibitors. Patients with an HP ratio trajectory in the highest quartile (≥ 0.0012 annually) displayed significantly shorter overall survival (median 18.4 months v 21.2 months, P = .003). CONCLUSION: In the first large longitudinal CTC study in mRCC to date to our knowledge, we identified the prognostic importance of CTC enumeration and the change over time of both CTC enumeration and the HP ratio. These insights into changes in both tumor burden and the molecular profile of tumor cells in response to different treatments provide potential biomarkers to predict and monitor response to immunotherapy in mRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Células Neoplásicas Circulantes , Humanos , Células Neoplásicas Circulantes/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/terapia , Antígeno B7-H1/metabolismo , Estudios Prospectivos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/terapia , Pronóstico
18.
Clin Epigenetics ; 14(1): 37, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35272673

RESUMEN

BACKGROUND: DNA methylation alterations have emerged as hallmarks of cancer and have been proposed as screening, prognostic, and predictive biomarkers. Traditional approaches for methylation analysis have relied on bisulfite conversion of DNA, which can damage DNA and is not suitable for targeted gene analysis in low-input samples. Here, we have adapted methyl-CpG-binding domain protein 2 (MBD2)-based DNA enrichment for use on a semi-automated exclusion-based sample preparation (ESP) platform for robust and scalable enrichment of methylated DNA from low-input samples, called SEEMLIS. RESULTS: We show that combining methylation-sensitive enzyme digestion with ESP-based MBD2 enrichment allows for single gene analysis with high sensitivity for GSTP1 in highly impure, heterogenous samples. We also show that ESP-based MBD2 enrichment coupled with targeted pre-amplification allows for analysis of multiple genes with sensitivities approaching the single cell level in pure samples for GSTP1 and RASSF1 and sensitivity down to 14 cells for these genes in highly impure samples. Finally, we demonstrate the potential clinical utility of SEEMLIS by successful detection of methylated gene signatures in circulating tumor cells (CTCs) from patients with prostate cancer with varying CTC number and sample purity. CONCLUSIONS: SEEMLIS is a robust assay for targeted DNA methylation analysis in low-input samples, with flexibility at multiple steps. We demonstrate the feasibility of this assay to analyze DNA methylation in prostate cancer cells using CTCs from patients with prostate cancer as a real-world example of a low-input analyte of clinical importance. In summary, this novel assay provides a platform for determining methylation signatures in rare cell populations with broad implications for research as well as clinical applications.


Asunto(s)
Metilación de ADN , Neoplasias de la Próstata , Islas de CpG , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Gutatión-S-Transferasa pi/genética , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/patología
19.
J Clin Oncol ; 40(5): 520-522, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34878806
20.
Prostate ; 82(2): 169-181, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34734426

RESUMEN

INTRODUCTION: The 2021 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Prostate Cancer Research in the 21st Century," was held virtually, from June 24-25, 2021. METHODS: The CHPCA Meeting is organized by the Prostate Cancer Foundation as a unique discussion-oriented meeting focusing on critical topics in prostate cancer research envisioned to bridge the next major advances in prostate cancer biology and treatment. The 2021 CHPCA Meeting was virtually attended by 89 investigators and included 31 talks over nine sessions. RESULTS: Major topic areas discussed at the meeting included: cancer genomics and sequencing, functional genomic approaches to studying mediators of plasticity, emerging signaling pathways in metastatic castration resistant prostate cancer, Wnt signaling biology and the challenges of targeted therapy, clonal hematopoiesis, neuroendocrine cell plasticity and antitumor immunity, cancer immunotherapy and its synergizers, and imaging the tumor microenvironment and metabolism. DISCUSSION: This meeting report summarizes the research presented at the 2021 CHPCA Meeting. We hope that publication of this knowledge will accelerate new understandings and the development of new biomarkers and treatments for prostate cancer.


Asunto(s)
Inmunoterapia/métodos , Próstata , Neoplasias de la Próstata , Congresos como Asunto , Humanos , Masculino , Próstata/diagnóstico por imagen , Próstata/inmunología , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Investigación/tendencias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA