Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Genet ; 56(3): 371-376, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38424461

ABSTRACT

Available genetically defined cancer models are limited in genotypic and phenotypic complexity and underrepresent the heterogeneity of human cancer. Here, we describe a combinatorial genetic strategy applied to an organoid transformation assay to rapidly generate diverse, clinically relevant bladder and prostate cancer models. Importantly, the clonal architecture of the resultant tumors can be resolved using single-cell or spatially resolved next-generation sequencing to uncover polygenic drivers of cancer phenotypes.


Subject(s)
Neoplasms , Male , Humans , Genotype , Phenotype , Neoplasms/genetics , Genetic Association Studies
2.
bioRxiv ; 2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37609344

ABSTRACT

Available genetically-defined cancer models are limited in genotypic and phenotypic complexity and underrepresent the heterogeneity of human cancer. Herein, we describe a combinatorial genetic strategy applied to an organoid transformation assay to rapidly generate diverse, clinically relevant bladder and prostate cancer models. Importantly, the clonal architecture of the resultant tumors can be resolved using single-cell or spatially resolved next-generation sequencing to uncover polygenic drivers of cancer phenotypes.

3.
Nat Commun ; 14(1): 2041, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37041154

ABSTRACT

Six transmembrane epithelial antigen of the prostate 1 (STEAP1) is a cell surface antigen for therapeutic targeting in prostate cancer. Here, we report broad expression of STEAP1 relative to prostate-specific membrane antigen (PSMA) in lethal metastatic prostate cancers and the development of a STEAP1-directed chimeric antigen receptor (CAR) T cell therapy. STEAP1 CAR T cells demonstrate reactivity in low antigen density, antitumor activity across metastatic prostate cancer models, and safety in a human STEAP1 knock-in mouse model. STEAP1 antigen escape is a recurrent mechanism of treatment resistance and is associated with diminished tumor antigen processing and presentation. The application of tumor-localized interleukin-12 (IL-12) therapy in the form of a collagen binding domain (CBD)-IL-12 fusion protein combined with STEAP1 CAR T cell therapy enhances antitumor efficacy by remodeling the immunologically cold tumor microenvironment of prostate cancer and combating STEAP1 antigen escape through the engagement of host immunity and epitope spreading.


Subject(s)
Prostatic Neoplasms , Receptors, Chimeric Antigen , Male , Mice , Animals , Humans , T-Lymphocytes , Interleukin-12 , Cell Line, Tumor , Prostatic Neoplasms/pathology , Immunotherapy , Tumor Microenvironment , Antigens, Neoplasm , Oxidoreductases
5.
JCI Insight ; 8(7)2023 04 10.
Article in English | MEDLINE | ID: mdl-36821396

ABSTRACT

Prostate-specific membrane antigen (PSMA) is an important cell surface target in prostate cancer. There are limited data on the heterogeneity of PSMA tissue expression in metastatic castration-resistant prostate cancer (mCRPC). Furthermore, the mechanisms regulating PSMA expression (encoded by the FOLH1 gene) are not well understood. Here, we demonstrate that PSMA expression is heterogeneous across different metastatic sites and molecular subtypes of mCRPC. In a rapid autopsy cohort in which multiple metastatic sites per patient were sampled, we found that 13 of 52 (25%) cases had no detectable PSMA and 23 of 52 (44%) cases showed heterogeneous PSMA expression across individual metastases, with 33 (63%) cases harboring at least 1 PSMA-negative site. PSMA-negative tumors displayed distinct transcriptional profiles with expression of druggable targets such as MUC1. Loss of PSMA was associated with epigenetic changes of the FOLH1 locus, including gain of CpG methylation and loss of histone 3 lysine 27 (H3K27) acetylation. Treatment with histone deacetylase (HDAC) inhibitors reversed this epigenetic repression and restored PSMA expression in vitro and in vivo. Collectively, these data provide insights into the expression patterns and regulation of PSMA in mCRPC and suggest that epigenetic therapies - in particular, HDAC inhibitors - can be used to augment PSMA levels.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/metabolism , Treatment Outcome , Prostate-Specific Antigen , Histone Deacetylase Inhibitors
6.
Semin Cancer Biol ; 83: 136-151, 2022 08.
Article in English | MEDLINE | ID: mdl-33545340

ABSTRACT

Conrad Waddington's theory of epigenetic landscape epitomize the process of cell fate and cellular decision-making during development. Wherein the epigenetic code maintains patterns of gene expression in pluripotent and differentiated cellular states during embryonic development and differentiation. Over the years disruption or reprogramming of the epigenetic landscape has been extensively studied in the course of cancer progression. Cellular dedifferentiation being a key hallmark of cancer allow us to take cues from the biological processes involving epigenetic reprogramming in development such as the cellular differentiation and morphogenesis. Here, we discuss the role of epigenetic landscape and its modifiers in cell-fate determination, differentiation and prostate cancer progression. Lately, the emergence of RNA-modifications has also furthered our understanding of epigenetics in cancer. The overview of the epigenetic code regulating androgen signalling, and progression to aggressive neuroendocrine stage of PCa reinforces its gene regulatory functions during the development of prostate gland as well as cancer progression. Additionally, we also highlight the clinical implications of cancer cell epigenome, and discuss the recent advancements in the therapeutic strategies targeting the advanced stage disease.


Subject(s)
Prostate , Prostatic Neoplasms , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Epigenomics , Humans , Male , Prostatic Neoplasms/genetics
8.
Nat Commun ; 12(1): 5325, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493733

ABSTRACT

Distal-less homeobox-1 (DLX1) is a well-established non-invasive biomarker for prostate cancer (PCa) diagnosis, however, its mechanistic underpinnings in disease pathobiology are not known. Here, we reveal the oncogenic role of DLX1 and show that abrogating its function leads to reduced tumorigenesis and metastases. We observed that ~60% of advanced-stage and metastatic patients display higher DLX1 levels. Moreover, ~96% of TMPRSS2-ERG fusion-positive and ~70% of androgen receptor (AR)-positive patients show elevated DLX1, associated with aggressive disease and poor survival. Mechanistically, ERG coordinates with enhancer-bound AR and FOXA1 to drive transcriptional upregulation of DLX1 in ERG-positive background. However, in ERG-negative context, AR/AR-V7 and FOXA1 suffice to upregulate DLX1. Notably, inhibiting ERG/AR-mediated DLX1 transcription using BET inhibitor (BETi) or/and anti-androgen drugs reduce its expression and downstream oncogenic effects. Conclusively, this study establishes DLX1 as a direct-target of ERG/AR with an oncogenic role and demonstrates the clinical significance of BETi and anti-androgens for DLX1-positive patients.


Subject(s)
Homeodomain Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcription Factors/genetics , Transcription, Genetic , Androgen Antagonists/pharmacology , Animals , Azepines/pharmacology , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Protein Binding , Receptors, Androgen/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Survival Analysis , Transcription Factors/metabolism , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Triazoles/pharmacology , Xenograft Model Antitumor Assays
10.
Home Healthc Now ; 38(5): 261-267, 2020.
Article in English | MEDLINE | ID: mdl-32889994

ABSTRACT

The need for comprehensive palliative care is inevitable with the aging population. Incorporating home-based palliative care is a new frontier within healthcare. The purpose of this study was to embed home-based palliative care services within the visiting nursing association (VNA) at a health system in Pennsylvania, examining effect on quality of life and symptom control, and average number of hospital admission days. A convenience sample of patients with one or more chronic conditions was selected from the existing VNA census (n = 22). A series of topics were outlined for discussion at each weekly visit for the pilot length of up to 6 months, scripted by evidence-based guidelines from the ENABLE II: Charting Your Course booklet (). A pretest/posttest survey method was conducted by utilizing results of the Edmonton Symptom Assessment System (ESAS) and the National Comprehensive Cancer Network Distress Thermometer. The effectiveness of the program was assessed using Spearman correlation to compare the difference in scores to the number of weeks in the program. The average number of hospital admission days during the pilot period was compared with admission days 6 months before enrollment in the pilot using the Wilcoxon signed-rank test. A significant relationship was found between the number of weeks in the program and reduction in the total ESAS symptom scores (rho = -0.484, p = .022), indicating that a reduction in symptoms was significantly more likely the longer a patient was in the program. Percentage of patients hospitalized decreased from 86% during preintervention period to 32% while enrolled. There was a noted reduction in the average number of days patients spent in the hospital while enrolled in the pilot (z = -2.24, p = 0.025).


Subject(s)
Home Care Services , Hospice and Palliative Care Nursing , Neoplasms , Aged , Humans , Palliative Care , Quality of Life , Surveys and Questionnaires
11.
Home Healthc Now ; 38(3): 131-137, 2020.
Article in English | MEDLINE | ID: mdl-32358440

ABSTRACT

Sepsis results in 270,000 deaths annually in the United States. Despite the current healthcare focus on sepsis, there exist few postacute best-practice standards to rapidly identify health changes in home healthcare patients to prevent and reduce hospital readmissions due to sepsis. We systematically examined whether an evidence-based process and intervention triggering home healthcare clinicians to activate a Positive Sepsis Assessment would reduce the likelihood that the patient would be readmitted to the acute care hospital. Over 24 months, we tracked the rate of sepsis readmissions to acute care hospitals through the initial phase of early recognition education; assessment, review, and revision of best-practice algorithms; standardized documentation; and proactive care management, in conjunction with the patient's primary care provider. During our review of the last 12 months of data on home care patients triggering the Positive Sepsis Assessment 130 patients were identified to have potential signs of sepsis. Ninety-seven of these patients received early medical intervention in place and were not readmitted to the hospital. Our findings suggest that a multidisciplinary home healthcare team utilizing standard sepsis education and sepsis algorithm on every patient during every visit can reduce and prevent readmissions.


Subject(s)
Clinical Decision-Making , Patient Readmission/statistics & numerical data , Patient Satisfaction/statistics & numerical data , Sepsis/therapy , Humans , Risk Assessment , Risk Factors , United States
12.
Nat Commun ; 11(1): 384, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959826

ABSTRACT

Emergence of an aggressive androgen receptor (AR)-independent neuroendocrine prostate cancer (NEPC) after androgen-deprivation therapy (ADT) is well-known. Nevertheless, the majority of advanced-stage prostate cancer patients, including those with SPINK1-positive subtype, are treated with AR-antagonists. Here, we show AR and its corepressor, REST, function as transcriptional-repressors of SPINK1, and AR-antagonists alleviate this repression leading to SPINK1 upregulation. Increased SOX2 expression during NE-transdifferentiation transactivates SPINK1, a critical-player for maintenance of NE-phenotype. SPINK1 elicits epithelial-mesenchymal-transition, stemness and cellular-plasticity. Conversely, pharmacological Casein Kinase-1 inhibition stabilizes REST, which in cooperation with AR causes SPINK1 transcriptional-repression and impedes SPINK1-mediated oncogenesis. Elevated levels of SPINK1 and NEPC markers are observed in the tumors of AR-antagonists treated mice, and in a subset of NEPC patients, implicating a plausible role of SPINK1 in treatment-related NEPC. Collectively, our findings provide an explanation for the paradoxical clinical-outcomes after ADT, possibly due to SPINK1 upregulation, and offers a strategy for adjuvant therapies.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Trypsin Inhibitor, Kazal Pancreatic/metabolism , Androgen Receptor Antagonists/therapeutic use , Animals , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/metabolism , Cell Line, Tumor , Co-Repressor Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Nerve Tissue Proteins/metabolism , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , SOXB1 Transcription Factors/metabolism , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
14.
Trends Mol Med ; 25(11): 1024-1038, 2019 11.
Article in English | MEDLINE | ID: mdl-31353123

ABSTRACT

Inter- and intra-patient molecular heterogeneity of primary and metastatic prostate cancer (PCa) confers variable clinical outcome and poses a formidable challenge in disease management. High-throughput integrative genomics and functional approaches have untangled the complexity involved in this disease and revealed a spectrum of diverse aberrations prevalent in various molecular subtypes, including ETS fusion negative. Emerging evidence indicates that SPINK1 upregulation, mutations in epigenetic regulators or chromatin modifiers, and SPOP are associated with the ETS-fusion negative subtype. Additionally, patients with defects in a DNA-repair pathway respond to poly-(ADP-ribose)-polymerase (PARP) inhibition therapies. Furthermore, a new class of immunogenic subtype defined by CDK12 biallelic loss has also been identified in ETS-fusion-negative cases. This review focuses on the emerging molecular underpinnings driving key oncogenic aberrations and advancements in therapeutic strategies of this disease.


Subject(s)
Molecular Targeted Therapy/trends , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Prostatic Neoplasms , Repressor Proteins/genetics , Trypsin Inhibitor, Kazal Pancreatic/genetics , Cyclin-Dependent Kinases/genetics , DNA Repair , ETS Motif/genetics , Epigenetic Repression , Gene Expression Regulation, Neoplastic , Genomics , Humans , Loss of Heterozygosity , Male , Nuclear Proteins/metabolism , Phosphatidylethanolamine Binding Protein/pharmacology , Piperazines/therapeutic use , Poly (ADP-Ribose) Polymerase-1/drug effects , Precision Medicine/trends , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Proteomics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Pyrimidines/therapeutic use , Repressor Proteins/metabolism , Signal Transduction , Trypsin Inhibitor, Kazal Pancreatic/metabolism
15.
Clin Cancer Res ; 25(9): 2755-2768, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30587549

ABSTRACT

PURPOSE: Serine peptidase inhibitor, Kazal type-1 (SPINK1) overexpression defines the second most recurrent and aggressive prostate cancer subtype. However, the underlying molecular mechanism and pathobiology of SPINK1 in prostate cancer remains largely unknown. EXPERIMENTAL DESIGN: miRNA prediction tools were employed to examine the SPINK1-3'UTR for miRNA binding. Luciferase reporter assays were performed to confirm the SPINK1-3'UTR binding of shortlisted miR-338-5p/miR-421. Furthermore, miR-338-5p/-421-overexpressing cancer cells (SPINK1-positive) were evaluated for oncogenic properties using cell-based functional assays and a mouse xenograft model. Global gene expression profiling was performed to unravel the biological pathways altered by miR-338-5p/-421. IHC and RNA in situ hybridization were carried out on prostate cancer patients' tissue microarray for SPINK1 and EZH2 expression, respectively. Chromatin immunoprecipitation assay was performed to examine EZH2 occupancy on the miR-338-5p/-421-regulatory regions. Bisulfite sequencing and methylated DNA immunoprecipitation were performed on prostate cancer cell lines and patients' specimens. RESULTS: We established a critical role of miRNA-338-5p/-421 in posttranscriptional regulation of SPINK1. Ectopic expression of miRNA-338-5p/-421 in SPINK1-positive cells abrogates oncogenic properties including cell-cycle progression, stemness, and drug resistance, and shows reduced tumor burden and distant metastases in a mouse model. Importantly, we show that patients with SPINK1-positive prostate cancer exhibit increased EZH2 expression, suggesting its role in epigenetic silencing of miRNA-338-5p/-421. Furthermore, presence of CpG dinucleotide DNA methylation marks on the regulatory regions of miR-338-5p/-421 in SPINK1-positive prostate cancer cells and patients' specimens confirms epigenetic silencing. CONCLUSIONS: Our findings revealed that miRNA-338-5p/-421 are epigenetically silenced in SPINK1-positive prostate cancer, although restoring the expression of these miRNAs using epigenetic drugs or synthetic mimics could abrogate SPINK1-mediated oncogenesis.See related commentary by Bjartell, p. 2679.


Subject(s)
MicroRNAs , Prostatic Neoplasms/genetics , Animals , Cell Line, Tumor , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Trypsin Inhibitor, Kazal Pancreatic/genetics
16.
ACS Appl Mater Interfaces ; 9(40): 34625-34633, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-28902490

ABSTRACT

Direct delivery of anticancer drugs to nuclei of tumor cells is required to enhance the therapeutic activity, which can be achieved by a nuclear localization signal (NLS) or peptide-decorated nanovehicles. However, NLS/peptide-based approaches may create certain undesirable immunological responses and the utilized synthesis processes are generally labor intensive. To this end, we report ligand-free, enhanced intranuclear delivery of Doxorubicin (Dox) to different cancer cells via porous polydimethylsiloxane (PDMS) nanoparticles (NPs). PDMS NPs were prepared by sacrificial silica template-based approach and Dox was loaded into the pores of PDMS NPs. These Dox-loaded PDMS NPs show enhanced cytotoxicity and reduce the IC50 values by 84 and 54% for HeLa and PC-3, respectively, compared to free Dox. Further, DNA damage in HeLa cells was estimated using comet assay suggesting enhanced DNA damage (72%) with Dox-loaded PDMS NPs as compared to free Dox (12%). The therapeutic efficiency of PDMS-Dox drug delivery system was tested in prostate cancer (PC-3) xenografts in NOD/SCID mice which showed enhanced tumor reduction (∼66%) as compared to free Dox. Taken together, our PDMS-Dox delivery system shows efficient and enhanced transportation of Dox to tumor cells which can be harnessed to develop advanced chemotherapy-based approaches to treat prostate and other cancers.


Subject(s)
Nanoparticles , Animals , Antineoplastic Agents , Cell Line, Tumor , Dimethylpolysiloxanes , Doxorubicin , Drug Carriers , Drug Delivery Systems , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID
17.
Trends Cancer ; 2(3): 116-120, 2016 03.
Article in English | MEDLINE | ID: mdl-28741531

ABSTRACT

Recent molecular characterization of prostate cancer (PCa) identified novel genetic aberrations and disease subtypes. The frequencies of molecular aberrations show racial disparity. Clinical strategies and targeted therapies embracing these racial differences are required. Here we discuss ethnic differences in genetic alterations and their impact on the susceptibility, progression, and treatment of prostate cancer.


Subject(s)
Prostatic Neoplasms/ethnology , Prostatic Neoplasms/genetics , Ethnicity , Genetic Predisposition to Disease , Humans , Male , Racial Groups , Serine Endopeptidases/genetics , Transcriptional Regulator ERG/genetics , Trypsin Inhibitor, Kazal Pancreatic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...