Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Neurooncol Adv ; 6(1): vdae041, 2024.
Article in English | MEDLINE | ID: mdl-38596716

ABSTRACT

Background: Liquid biopsy based on circulating tumor DNA (ctDNA) is a novel tool in clinical oncology, however, its use has been limited in glioma to date, due to low levels of ctDNA. In this study, we aimed to demonstrate that sequencing techniques optimized for liquid biopsy in glioma patients can detect ctDNA in plasma with high sensitivity and with potential clinical utility. Methods: We investigated 10 glioma patients with tumor tissue available from at least 2 surgical operations, who had 49 longitudinally collected plasma samples available for analysis. Plasma samples were sequenced with CAPP-seq (AVENIO) and tissue samples with TSO500. Results: Glioma-derived ctDNA mutations were detected in 93.8% of plasma samples. 25% of all mutations detected were observed in plasma only. Mutations of the mismatch repair (MMR) genes MSH2 and MSH6 were the most frequent circulating gene alterations seen after temozolomide treatment and were frequently observed to appear in plasma prior to their appearance in tumor tissue at the time of surgery for recurrence. Conclusions: This pilot study suggests that plasma ctDNA in glioma is feasible and may provide sensitive and complementary information to tissue biopsy. Furthermore, plasma ctDNA detection of new MMR gene mutations not present in the initial tissue biopsy may provide an early indication of the development of chemotherapy resistance. Additional clinical validation in larger cohorts is needed.

2.
J Pathol ; 263(2): 242-256, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38578195

ABSTRACT

There are diverse phenotypes of castration-resistant prostate cancer, including neuroendocrine disease, that vary in their sensitivity to drug treatment. The efficacy of BET and CBP/p300 inhibitors in prostate cancer is attributed, at least in part, to their ability to decrease androgen receptor (AR) signalling. However, the activity of BET and CBP/p300 inhibitors in prostate cancers that lack the AR is unclear. In this study, we showed that BRD4, CBP, and p300 were co-expressed in AR-positive and AR-null prostate cancer. A combined inhibitor of these three proteins, NEO2734, reduced the growth of both AR-positive and AR-null organoids, as measured by changes in viability, size, and composition. NEO2734 treatment caused consistent transcriptional downregulation of cell cycle pathways. In neuroendocrine models, NEO2734 treatment reduced ASCL1 levels and other neuroendocrine markers, and reduced tumour growth in vivo. Collectively, these results show that epigenome-targeted inhibitors cause decreased growth and phenotype-dependent disruption of lineage regulators in neuroendocrine prostate cancer, warranting further development of compounds with this activity in the clinic. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
E1A-Associated p300 Protein , Receptors, Androgen , Signal Transduction , Male , Humans , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Animals , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mice , Xenograft Model Antitumor Assays , Bromodomain Containing Proteins , CREB-Binding Protein
3.
Genome Biol ; 25(1): 110, 2024 04 29.
Article in English | MEDLINE | ID: mdl-38685127

ABSTRACT

BACKGROUND: Metazoans inherited genes from unicellular ancestors that perform essential biological processes such as cell division, metabolism, and protein translation. Multicellularity requires careful control and coordination of these unicellular genes to maintain tissue integrity and homeostasis. Gene regulatory networks (GRNs) that arose during metazoan evolution are frequently altered in cancer, resulting in over-expression of unicellular genes. We propose that an imbalance in co-expression of unicellular (UC) and multicellular (MC) genes is a driving force in cancer. RESULTS: We combine gene co-expression analysis to infer changes to GRNs in cancer with protein sequence conservation data to distinguish genes with UC and MC origins. Co-expression networks created using RNA sequencing data from 31 tumor types and normal tissue samples are divided into modules enriched for UC genes, MC genes, or mixed UC-MC modules. The greatest differences between tumor and normal tissue co-expression networks occur within mixed UC-MC modules. MC and UC genes not commonly co-expressed in normal tissues form distinct co-expression modules seen only in tumors. The degree of rewiring of genes within mixed UC-MC modules increases with tumor grade and stage. Mixed UC-MC modules are enriched for somatic mutations in cancer genes, particularly amplifications, suggesting an important driver of the rewiring observed in tumors is copy number changes. CONCLUSIONS: Our study shows the greatest changes to gene co-expression patterns during tumor progression occur between genes of MC and UC origins, implicating the breakdown and rewiring of metazoan gene regulatory networks in cancer development and progression.


Subject(s)
Gene Regulatory Networks , Neoplasms , Neoplasms/genetics , Humans , Animals , Gene Expression Regulation, Neoplastic , Evolution, Molecular
4.
Prostate ; 84(7): 623-635, 2024 May.
Article in English | MEDLINE | ID: mdl-38450798

ABSTRACT

BACKGROUND: There are relatively few widely used models of prostate cancer compared to other common malignancies. This impedes translational prostate cancer research because the range of models does not reflect the diversity of disease seen in clinical practice. In response to this challenge, research laboratories around the world have been developing new patient-derived models of prostate cancer, including xenografts, organoids, and tumor explants. METHODS: In May 2023, we held a workshop at the Monash University Prato Campus for researchers with expertise in establishing and using a variety of patient-derived models of prostate cancer. This review summarizes our collective ideas on how patient-derived models are currently being used, the common challenges, and future opportunities for maximizing their usefulness in prostate cancer research. RESULTS: An increasing number of patient-derived models for prostate cancer are being developed. Despite their individual limitations and varying success rates, these models are valuable resources for exploring new concepts in prostate cancer biology and for preclinical testing of potential treatments. Here we focus on the need for larger collections of models that represent the changing treatment landscape of prostate cancer, robust readouts for preclinical testing, improved in vitro culture conditions, and integration of the tumor microenvironment. Additional priorities include ensuring model reproducibility, standardization, and replication, and streamlining the exchange of models and data sets among research groups. CONCLUSIONS: There are several opportunities to maximize the impact of patient-derived models on prostate cancer research. We must develop large, diverse and accessible cohorts of models and more sophisticated methods for emulating the intricacy of patient tumors. In this way, we can use the samples that are generously donated by patients to advance the outcomes of patients in the future.


Subject(s)
Prostatic Neoplasms , Male , Humans , Reproducibility of Results , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Prostate/pathology , Organoids/pathology , Heterografts , Tumor Microenvironment
5.
PLoS One ; 18(11): e0292278, 2023.
Article in English | MEDLINE | ID: mdl-37917641

ABSTRACT

Drugs targeting cyclin-dependent kinases 4 and 6 (CDK4/6) are promising new treatments for melanoma and other solid malignancies. In studies on CDK4/6 inhibitor resistance, protein arginine methyltransferase 5 (PRMT5) regulation of alternative splicing was shown to be an important downstream component of the CDK4/6 pathway. However, the full effects of inhibition of CDK4/6 on splicing events in melanoma and the extent to which they are dependent on PRMT5 has not been established. We performed full-length mRNA sequencing on CHL1 and A375 melanoma cell lines treated with the CDK4/6 inhibitor palbociclib and the PRMT5 inhibitor GSK3326595 and analysed data for differential gene expression and differential pre-mRNA splicing induced by these agents. Changes in gene expression and RNA splicing were more extensive under PRMT5 inhibition than under CDK4/6 inhibition. Although PRMT5 inhibition and CDK4/6 inhibition induced common RNA splicing events and gene expression profiles, the majority of events induced by CDK4/6 inhibition were distinct. Our findings indicate CDK4/6 has the ability to regulate alternative splicing in a manner that is distinct from PRMT5 inhibition, resulting in divergent changes in gene expression under each therapy.


Subject(s)
Alternative Splicing , Melanoma , Humans , Protein-Arginine N-Methyltransferases/metabolism , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , RNA Splicing , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism
6.
Nat Commun ; 14(1): 2697, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37188662

ABSTRACT

Spatial proteomics technologies have revealed an underappreciated link between the location of cells in tissue microenvironments and the underlying biology and clinical features, but there is significant lag in the development of downstream analysis methods and benchmarking tools. Here we present SPIAT (spatial image analysis of tissues), a spatial-platform agnostic toolkit with a suite of spatial analysis algorithms, and spaSim (spatial simulator), a simulator of tissue spatial data. SPIAT includes multiple colocalization, neighborhood and spatial heterogeneity metrics to characterize the spatial patterns of cells. Ten spatial metrics of SPIAT are benchmarked using simulated data generated with spaSim. We show how SPIAT can uncover cancer immune subtypes correlated with prognosis in cancer and characterize cell dysfunction in diabetes. Our results suggest SPIAT and spaSim as useful tools for quantifying spatial patterns, identifying and validating correlates of clinical outcomes and supporting method development.


Subject(s)
Neoplasms , Humans , Algorithms , Image Processing, Computer-Assisted/methods , Proteomics , Tumor Microenvironment
7.
Nat Rev Urol ; 20(6): 371-384, 2023 06.
Article in English | MEDLINE | ID: mdl-36650259

ABSTRACT

Patient-derived xenografts (PDXs) are generated by engrafting human tumours into mice. Serially transplantable PDXs are used to study tumour biology and test therapeutics, linking the laboratory to the clinic. Although few prostate cancer PDXs are available in large repositories, over 330 prostate cancer PDXs have been established, spanning broad clinical stages, genotypes and phenotypes. Nevertheless, more PDXs are needed to reflect patient diversity, and to study new treatments and emerging mechanisms of resistance. We can maximize the use of PDXs by exchanging models and datasets, and by depositing PDXs into biorepositories, but we must address the impediments to accessing PDXs, such as institutional, ethical and legal agreements. Through collaboration, researchers will gain greater access to PDXs representing diverse features of prostate cancer.


Subject(s)
Prostatic Neoplasms , Male , Humans , Mice , Animals , Heterografts , Xenograft Model Antitumor Assays , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Prostate/pathology , Genotype , Disease Models, Animal
8.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35764368

ABSTRACT

BACKGROUND: Aberrations in homologous recombination repair (HRR) genes are emerging as important biomarkers for personalized treatment in prostate cancer (PCa). HRR deficiency (HRD) could affect the tumor immune microenvironment (TIME), potentially contributing to differential responses to poly ADP-ribose polymerase (PARP) inhibitors and immune checkpoint inhibitors. Spatial distribution of immune cells in a range of cancers identifies novel disease subtypes and is related to prognosis. In this study we aimed to determine the differences in the TIME of PCa with and without germline (g) HRR mutations. METHODS: We performed gene expression analysis, multiplex immunohistochemistry of T and B cells and quantitative spatial analysis of PCa samples from 36 patients with gHRD and 26 patients with sporadic PCa. Samples were archival tumor tissue from radical prostatectomies with the exception of one biopsy. Results were validated in several independent cohorts. RESULTS: Although the composition of the T cell and B cells was similar in the tumor areas of gHRD-mutated and sporadic tumors, the spatial profiles differed between these cohorts. We describe two T-cell spatial profiles across primary PCa, a clustered immune spatial (CIS) profile characterized by dense clusters of CD4+ T cells closely interacting with PD-L1+ cells, and a free immune spatial (FIS) profile of CD8+ cells in close proximity to tumor cells. gHRD tumors had a more T-cell inflamed microenvironment than sporadic tumors. The CIS profile was mainly observed in sporadic tumors, whereas a FIS profile was enriched in gHRD tumors. A FIS profile was associated with lower Gleason scores, smaller tumors and longer time to biochemical recurrence and metastasis. CONCLUSIONS: gHRD-mutated tumors have a distinct immune microenvironment compared with sporadic tumors. Spatial profiling of T-cells provides additional information beyond T-cell density and is associated with time to biochemical recurrence, time to metastasis, tumor size and Gleason scores.


Subject(s)
Germ-Line Mutation , Prostatic Neoplasms , Humans , Immune Checkpoint Inhibitors , Male , Prostatic Neoplasms/genetics , Recombinational DNA Repair , Tumor Microenvironment/genetics
9.
BMC Gastroenterol ; 22(1): 197, 2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35448971

ABSTRACT

BACKGROUND: Intestinal metaplasia (IM) is considered a key pivot point in the Correa model of gastric cancer (GC). It is histologically subtyped into the complete and incomplete subtypes, the latter being associated with a greater risk of progression. However, the clinical utility of IM subtyping remains unclear, partially due to the absence of reliable defining biomarkers. METHODS: Based on gene expression data and existing literature, we selected CD10 and Das1 as candidate biomarkers to distinguish complete and incomplete IM glands in tissues from patients without GC (IM-GC) and patients with GC (IM + GC). Immunohistochemical staining of individually subtyped IM glands was scored after blinding by two researchers using tissue belonging to both IM-GC and IM + GC patients. Whole tissue Das1 staining was further assessed using digital image quantification (cellSens Dimension, Olympus). RESULTS: Across both cohorts CD10 stained the IM brush border and was shown to have a high sensitivity (87.5% and 94.9% in IM-GC and IM + GC patients respectively) and specificity (100.0% and 96.7% respectively) with an overall AUROC of 0.944 for complete IM glands. By contrast Das1 stained mainly goblet cells and the apical membrane of epithelial cells, mostly of incomplete IM glands with a low sensitivity (28.6% and 29.3% in IM-GC and IM + GC patients respectively) but high specificity (98.3% and 85.1% respectively) and an overall AUROC of 0.603 for incomplete IM glands. A combined logistic regression model showed a significant increase in AUROC for detecting complete IM glands (0.955 vs 0.970). Whole tissue digital quantification of Das1 staining showed a significant association with incomplete IM compared to complete IM, both in IM-GC and in IM + GC patients (p = 0.016 and p = 0.009 respectively, Mann-Whitney test and unpaired t test used). Additionally, complete IM in IM + GC patients exhibited significantly more Das1 staining than in IM-GC patients (p = 0.019, Mann-Whitney test). CONCLUSIONS: These findings suggest that CD10 is an outstanding biomarker for complete IM and Das1 may be useful as a secondary biomarker for IM glands at greater risk of progression irrespective of IM subtype. Overall, the clinical use of these biomarkers could lead to improved patient stratification and targeted surveillance.


Subject(s)
Precancerous Conditions , Stomach Neoplasms , Biomarkers , Gastric Mucosa/pathology , Humans , Immunohistochemistry , Metaplasia/pathology , Precancerous Conditions/pathology , Stomach Neoplasms/pathology
10.
Nat Commun ; 13(1): 1100, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35232962

ABSTRACT

Despite the success of therapies targeting oncogenes in cancer, clinical outcomes are limited by residual disease that ultimately results in relapse. This residual disease is often characterized by non-genetic adaptive resistance, that in melanoma is characterised by altered metabolism. Here, we examine how targeted therapy reprograms metabolism in BRAF-mutant melanoma cells using a genome-wide RNA interference (RNAi) screen and global gene expression profiling. Using this systematic approach we demonstrate post-transcriptional regulation of metabolism following BRAF inhibition, involving selective mRNA transport and translation. As proof of concept we demonstrate the RNA processing kinase U2AF homology motif kinase 1 (UHMK1) associates with mRNAs encoding metabolism proteins and selectively controls their transport and translation during adaptation to BRAF-targeted therapy. UHMK1 inactivation induces cell death by disrupting therapy induced metabolic reprogramming, and importantly, delays resistance to BRAF and MEK combination therapy in multiple in vivo models. We propose selective mRNA processing and translation by UHMK1 constitutes a mechanism of non-genetic resistance to targeted therapy in melanoma by controlling metabolic plasticity induced by therapy.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Molecular Targeted Therapy , Mutation , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , RNA, Messenger/therapeutic use
11.
Mol Cancer Ther ; 20(11): 2140-2150, 2021 11.
Article in English | MEDLINE | ID: mdl-34413130

ABSTRACT

Monotherapy with PARP inhibitors is effective for the subset of castrate-resistant prostate cancer (CRPC) with defects in homologous recombination (HR) DNA repair. New treatments are required for the remaining tumors, and an emerging strategy is to combine PARP inhibitors with other therapies that induce DNA damage. Here we tested whether PARP inhibitors are effective for HR-proficient CRPC, including androgen receptor (AR)-null tumors, when used in combination with CX-5461, a small molecule that inhibits RNA polymerase I transcription and activates the DNA damage response, and has antitumor activity in early phase I trials. The combination of CX-5461 and talazoparib significantly decreased in vivo growth of patient-derived xenografts of HR-proficient CRPC, including AR-positive, AR-null, and neuroendocrine tumors. CX-5461 and talazoparib synergistically inhibited the growth of organoids and cell lines, and significantly increased the levels of DNA damage. Decreased tumor growth after combination therapy was maintained for 2 weeks without treatment, significantly increasing host survival. Therefore, combination treatment with CX-5461 and talazoparib is effective for HR-proficient tumors that are not suitable for monotherapy with PARP inhibitors, including AR-null CRPC. This expands the spectrum of CRPC that is sensitive to PARP inhibition.


Subject(s)
Benzothiazoles/therapeutic use , DNA Damage/genetics , Naphthyridines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Benzothiazoles/pharmacology , Humans , Male , Mice , Naphthyridines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
12.
Nat Commun ; 12(1): 5049, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413304

ABSTRACT

Preclinical testing is a crucial step in evaluating cancer therapeutics. We aimed to establish a significant resource of patient-derived xenografts (PDXs) of prostate cancer for rapid and systematic evaluation of candidate therapies. The PDX collection comprises 59 tumors collected from 30 patients between 2012-2020, coinciding with availability of abiraterone and enzalutamide. The PDXs represent the clinico-pathological and genomic spectrum of prostate cancer, from treatment-naïve primary tumors to castration-resistant metastases. Inter- and intra-tumor heterogeneity in adenocarcinoma and neuroendocrine phenotypes is evident from bulk and single-cell RNA sequencing data. Organoids can be cultured from PDXs, providing further capabilities for preclinical studies. Using a 1 x 1 x 1 design, we rapidly identify tumors with exceptional responses to combination treatments. To govern the distribution of PDXs, we formed the Melbourne Urological Research Alliance (MURAL). This PDX collection is a substantial resource, expanding the capacity to test and prioritize effective treatments for prospective clinical trials in prostate cancer.


Subject(s)
Drug Evaluation, Preclinical/methods , Organoids/pathology , Prostatic Neoplasms/pathology , Animals , Disease Models, Animal , Genome , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Metastasis , Organoids/metabolism , Prospective Studies , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tissue Banks , Transcriptome , Xenograft Model Antitumor Assays
13.
Cancers (Basel) ; 13(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799802

ABSTRACT

Mast cells (MCs) are important cellular components of the tumor microenvironment and are significantly associated with poor patient outcomes in prostate cancer and other solid cancers. The promotion of tumor progression partly involves heterotypic interactions between MCs and cancer-associated fibroblasts (CAFs), which combine to potentiate a pro-tumor extracellular matrix and promote epithelial cell invasion and migration. Thus far, the interactions between MCs and CAFs remain poorly understood. To identify molecular changes that may alter resident MC function in the prostate tumor microenvironment, we profiled the transcriptome of human prostate MCs isolated from patient-matched non-tumor and tumor-associated regions of fresh radical prostatectomy tissue. Transcriptomic profiling revealed a distinct gene expression profile of MCs isolated from prostate tumor regions, including the downregulation of SAMD14, a putative tumor suppressor gene. Proteomic profiling revealed that overexpression of SAMD14 in HMC-1 altered the secretion of proteins associated with immune regulation and extracellular matrix processes. To assess MC biological function within a model of the prostate tumor microenvironment, HMC-1-SAMD14+ conditioned media was added to co-cultures of primary prostatic CAFs and prostate epithelium. HMC-1-SAMD14+ secretions were shown to reduce the deposition and alignment of matrix produced by CAFs and suppress pro-tumorigenic prostate epithelial morphology. Overall, our data present the first profile of human MCs derived from prostate cancer patient specimens and identifies MC-derived SAMD14 as an important mediator of MC phenotype and function within the prostate tumor microenvironment.

14.
J Pathol ; 254(2): 121-134, 2021 06.
Article in English | MEDLINE | ID: mdl-33620092

ABSTRACT

Amplifications of the androgen receptor (AR) occur in up to 80% of men with castration-resistant prostate cancer (CRPC). Recent studies highlighted that these amplifications not only span the AR gene but usually encompass a distal enhancer. This represents a newly recognised, non-coding mechanism of resistance to AR-directed therapies, including enzalutamide. To study disease progression before and after AR amplification, we used tumour samples from a castrate-sensitive primary tumour and castrate-resistant metastasis of the same patient. For subsequent functional and genomic studies, we established serially transplantable patient-derived xenografts (PDXs). Whole genome sequencing showed that alterations associated with poor prognosis, such as TP53 and PTEN loss, existed before androgen deprivation therapy, followed by co-amplification of the AR gene and enhancer after the development of metastatic CRPC. The PDX of the primary tumour, without the AR amplification, was sensitive to AR-directed treatments, including castration, enzalutamide, and apalutamide. The PDX of the metastasis, with the AR amplification, had higher AR and AR-V7 expression in castrate conditions, and was resistant to castration, apalutamide, and enzalutamide in vivo. Treatment with a BET inhibitor outperformed the AR-directed therapies for the metastasis, resulting in tumour regression for some, but not all, grafts. Therefore, this study provides novel matched PDXs to test potential treatments that target the overabundance of AR in tumours with AR enhancer amplifications. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antineoplastic Agents/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Androgen Antagonists/pharmacology , Androgens/metabolism , Animals , Benzamides/pharmacology , Disease Models, Animal , Disease Progression , Heterografts , Humans , Male , Mice , Nitriles/pharmacology , Orchiectomy , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Thiohydantoins/pharmacology , Whole Genome Sequencing
15.
EMBO J ; 39(21): e105111, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32945574

ABSTRACT

Elevated ribosome biogenesis in oncogene-driven cancers is commonly targeted by DNA-damaging cytotoxic drugs. Our previous first-in-human trial of CX-5461, a novel, less genotoxic agent that specifically inhibits ribosome biogenesis via suppression of RNA polymerase I (Pol I) transcription, revealed single-agent efficacy in refractory blood cancers. Despite this clinical response, patients were not cured. In parallel, we demonstrated a marked improvement in the in vivo efficacy of CX-5461 in combination with PI3K/AKT/mTORC1 pathway inhibitors. Here, we reveal the molecular basis for this improved efficacy observed in vivo, which is associated with specific suppression of translation of mRNAs encoding regulators of cellular metabolism. Importantly, acquired resistance to this cotreatment is driven by translational rewiring that results in dysregulated cellular metabolism and induction of a cAMP-dependent pathway critical for the survival of blood cancers including lymphoma and acute myeloid leukemia. Our studies thus identify key molecular mechanisms underpinning the response of blood cancers to selective inhibition of ribosome biogenesis and define metabolic vulnerabilities that will facilitate the rational design of more effective regimens for Pol I-directed therapies.


Subject(s)
Neoplasms/metabolism , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Ribosomes/metabolism , Transcription, Genetic/drug effects , Animals , Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Naphthyridines/pharmacology , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors , RNA Polymerase I/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal , Ribosomes/drug effects , Transcriptome
16.
Biotechniques ; 69(2): 133-140, 2020 08.
Article in English | MEDLINE | ID: mdl-32654508

ABSTRACT

Plasma circulating tumor DNA (ctDNA) analysis has emerged as a minimally invasive means to perform molecular tumor typing. Here we developed a custom ultra-sensitive ctDNA next-generation sequencing assay using molecular barcoding technology and off-the-shelf reagents combined with bioinformatics tools for enhanced ctDNA analysis. Assay performance was assessed via a spike-in experiment and the technique was applied to analyze 41 plasma samples from men with advanced prostate cancer. Orthogonal validation was performed using a commercial assay. Sensitivity and specificity of 93 and 99.5% were recorded for ultra-rare somatic variants (<1%), with high concordance observed between the in-house and commercial assays. The optimized protocol dramatically improved the efficiency of the assay and enabled the detection of low-frequency somatic variants from plasma cell-free DNA (cfDNA).


Subject(s)
Circulating Tumor DNA/blood , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Cell-Free Nucleic Acids/blood , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Liquid Biopsy/methods , Liquid Biopsy/standards , Male , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Sensitivity and Specificity , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
17.
Science ; 368(6495): 1127-1131, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32499442

ABSTRACT

In microorganisms, evolutionarily conserved mechanisms facilitate adaptation to harsh conditions through stress-induced mutagenesis (SIM). Analogous processes may underpin progression and therapeutic failure in human cancer. We describe SIM in multiple in vitro and in vivo models of human cancers under nongenotoxic drug selection, paradoxically enhancing adaptation at a competing intrinsic fitness cost. A genome-wide approach identified the mechanistic target of rapamycin (MTOR) as a stress-sensing rheostat mediating SIM across multiple cancer types and conditions. These observations are consistent with a two-phase model for drug resistance, in which an initially rapid expansion of genetic diversity is counterbalanced by an intrinsic fitness penalty, subsequently normalizing to complete adaptation under the new conditions. This model suggests synthetic lethal strategies to minimize resistance to anticancer therapy.


Subject(s)
Adaptation, Physiological/genetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Mutagenesis , Neoplasms/drug therapy , Neoplasms/genetics , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , DNA Repair/genetics , Genetic Fitness , Genome-Wide Association Study , Humans , Selection, Genetic , Signal Transduction , TOR Serine-Threonine Kinases/genetics
18.
NPJ Breast Cancer ; 6: 9, 2020.
Article in English | MEDLINE | ID: mdl-32195332

ABSTRACT

Intraductal papillomas (IDP) are challenging breast findings because of their variable risk of progression to malignancy. The molecular events driving IDP development and genomic features of malignant progression are poorly understood. In this study, genome-wide CNA and/or targeted mutation analysis was performed on 44 cases of IDP, of which 20 cases had coexisting ductal carcinoma in situ (DCIS), papillary DCIS or invasive ductal carcinoma (IDC). CNA were rare in pure IDP, but 69% carried an activating PIK3CA mutation. Among the synchronous IDP cases, 55% (11/20) were clonally related to the synchronous DCIS and/or IDC, only one of which had papillary histology. In contrast to pure IDP, PIK3CA mutations were absent from clonal cases. CNAs in any of chromosomes 1, 16 or 11 were significantly enriched in clonal IDP lesions compared to pure and non-clonal IDP. The observation that 55% of IDP are clonal to DCIS/IDC indicates that IDP can be a direct precursor for breast carcinoma, not limited to the papillary type. The absence of PIK3CA mutations and presence of CNAs in IDP could be used clinically to identify patients at high risk of progression to carcinoma.

19.
J Prosthet Dent ; 124(6): 674-681, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31952858

ABSTRACT

STATEMENT OF PROBLEM: Wearers of mandibular complete dentures (CDs) often complain of retention and stability problems resulting in poor masticatory function. Evidence suggests that a mandibular overdenture (MOD) stabilized by 2 implants represents the treatment of choice to improve stability and masticatory function. Measurements are needed of the improvement in masticatory function after providing mandibular implant-stabilized overdentures. PURPOSE: The purpose of this prospective clinical study was to evaluate the changes in masticatory function from baseline (T0) to 3 months (T1) and 3 years (T2) in participants with MODs and to assess the effect of baseline mandibular bone height and volume on masticatory function after 3 years. MATERIAL AND METHODS: Participants were assessed for masticatory function by using masticatory performance involving paraffin wax cubes as an objective measure and by using masticatory ability involving a questionnaire as a subjective measure. Edentulous individuals presenting for replacement dentures were provided with conventional mucosa-supported prostheses and evaluated for masticatory function after a 3-month settling-in period (baseline measure). Before implant placement, baseline measures of bone height and volume were recorded from cone beam computed tomography (CBCT) images. The prostheses were then converted to implant-stabilized mandibular overdentures while any maxillary prostheses remained supported by the mucosa. Masticatory function was reassessed at 3 months and 3 years after insertion of the mandibular overdentures, and the mean changes from baseline were analyzed with the Wilcoxon signed-rank test. The effect of variables on masticatory function was determined by using multivariate linear regression analyses. RESULTS: A total of 23 participants were included in the study, with only 1 participant not completing the 3-year assessment. Significant improvement was observed in the masticatory performance (mixing ability index) (P<.01) and masticatory ability score (P<.001) from baseline to 3 months and baseline to 3 years. Bone height and volume had no significant effect on the improvement of masticatory function after conversion to an implant-stabilized mandibular overdenture. CONCLUSIONS: Masticatory function significantly improved after 3 months and was maintained over 3 years in participants with implant-stabilized mandibular overdentures. However, baseline bone height and volume had no significant effect on these changes in masticatory function after 3 years.


Subject(s)
Dental Implants , Denture, Overlay , Dental Prosthesis, Implant-Supported , Denture, Complete, Lower , Humans , Mandible/diagnostic imaging , Mastication , Patient Satisfaction , Prospective Studies
20.
Prostate ; 79(11): 1326-1337, 2019 08.
Article in English | MEDLINE | ID: mdl-31212368

ABSTRACT

BACKGROUND: Serially transplantable patient-derived xenografts (PDXs) are invaluable preclinical models for studying tumor biology and evaluating therapeutic agents. As these models are challenging to establish from prostate cancer specimens, the ability to preserve them through cryopreservation has several advantages for ongoing research. Despite this, there is still uncertainty about the ability to cryopreserve PDXs of prostate cancer. This study compared three different cryopreservation protocols to identify a method that can be used to reproducibly cryopreserve a diverse cohort of prostate cancer PDX models. METHODS: One serially transplantable prostate cancer PDX from the Melbourne Urological Research Alliance cohort was used to compare three cryopreservation protocols: slow freezing in fetal calf serum (FCS) with 10% dimethyl sulfoxide (DMSO), FCS with 10% DMSO supplemented with the Rho-associated kinase (ROCK) inhibitor Y-27632 and vitrification. The efficiency of the slow freezing protocols was then assessed in 17 additional prostate cancer PDXs. Following cryopreservation, PDXs were re-established in host mice that were either intact and supplemented with testosterone or castrated. Graft take rate, tumor growth, histological features, and transcriptome profiles before and after cryopreservation were compared. RESULTS: Slow freezing maintained the viability and histological features of prostate cancer PDXs, and the addition of a ROCK inhibitor increased their growth following cryopreservation. Using the slow freezing method, we re-established 100% of PDXs grown in either testosterone-supplemented or castrated host mice. Importantly, the long-term tumor growth rate and transcriptome profile were maintained following cryopreservation. CONCLUSION: This study has identified a protocol to reliably cryopreserve and re-establish a diverse cohort of serially transplantable PDXs of prostate cancer. This study has the potential to significantly improve the practicality of maintaining PDX models. Cryopreservation may also increase the accessibility of these important resources and provide new opportunities for preclinical studies on a broader spectrum of prostate tumors.


Subject(s)
Cryopreservation/methods , Heterografts , Neoplasm Transplantation/methods , Prostatic Neoplasms/pathology , Animals , Disease Models, Animal , Humans , Male , Mice , Neoplasm Transplantation/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...