Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
BMC Cancer ; 22(1): 675, 2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35725412

ABSTRACT

BACKGROUND: Somatic alterations in the cancer genome, some of which are associated with changes in gene expression, have been characterized in multiple studies across diverse cancer types. However, less is known about germline variants that influence tumor biology by shaping the cancer transcriptome. METHODS: We performed expression quantitative trait loci (eQTL) analyses using multi-dimensional data from The Cancer Genome Atlas to explore the role of germline variation in mediating the cancer transcriptome. After accounting for associations between somatic alterations and gene expression, we determined the contribution of inherited variants to the cancer transcriptome relative to that of somatic variants. Finally, we performed an interaction analysis using estimates of tumor cellularity to identify cell type-restricted eQTLs. RESULTS: The proportion of genes with at least one eQTL varied between cancer types, ranging between 0.8% in melanoma to 28.5% in thyroid cancer and was correlated more strongly with intratumor heterogeneity than with somatic alteration rates. Although contributions to variance in gene expression was low for most genes, some eQTLs accounted for more than 30% of expression of proximal genes. We identified cell type-restricted eQTLs in genes known to be cancer drivers including LPP and EZH2 that were associated with disease-specific mortality in TCGA but not associated with disease risk in published GWAS. Together, our results highlight the need to consider germline variation in interpreting cancer biology beyond risk prediction.


Subject(s)
Genome-Wide Association Study , Melanoma , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Polymorphism, Single Nucleotide , Transcriptome
2.
Clin Cancer Res ; 28(5): 870-881, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34862243

ABSTRACT

PURPOSE: This phase I, dose-escalation study investigated the recommended dose for expansion (RDE) of siremadlin, a p53-MDM2 inhibitor, in patients with wild-type TP53 advanced solid or hematologic cancers. PATIENTS AND METHODS: Initial dosing regimens were: 1A (day 1; 21-day cycle; dose 12.5-350 mg) and 2A (days 1-14; 28-day cycle; dose 1-20 mg). Alternative regimens included 1B (days 1 and 8; 28-day cycle) and 2C (days 1-7; 28-day cycle). The primary endpoint was incidence of dose-limiting toxicities (DLT) during cycle 1. RESULTS: Overall, 115 patients with solid tumors and 93 with hematologic malignancies received treatment. DLTs occurred in 8/92 patients with solid tumors and 10/53 patients with hematologic malignancies. In solid tumors, an RDE of 120 mg was defined in 1B. In hematologic tumors, RDEs were defined in 1A: 250 mg, 1B: 120 mg, and 2C: 45 mg. More patients with hematologic malignancies compared with solid tumors experienced grade 3/4 treatment-related adverse events (71% vs. 45%), most commonly resulting from myelosuppression. These were more frequent and severe in patients with hematologic malignancies; 22 patients exhibited tumor lysis syndrome. Overall response rates at the RDEs were 10.3% [95% confidence interval (CI), 2.2-27.4] in solid tumors and 4.2% (95% CI, 0.1-21.1), 20% (95% CI, 4.3-48.1), and 22.2% (95% CI, 8.6-42.3) in acute myeloid leukemia (AML) in 1B, 1A, and 2C, respectively. CONCLUSIONS: A common safety profile was identified and preliminary activity was noted, particularly in AML. Comprehensive investigation of dosing regimens yielded recommended doses/regimens for future combination studies.


Subject(s)
Hematologic Neoplasms , Leukemia, Myeloid, Acute , Neoplasms , Dose-Response Relationship, Drug , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Humans , Imidazoles , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Maximum Tolerated Dose , Neoplasms/drug therapy , Pyrimidines , Pyrroles , Tumor Suppressor Protein p53/genetics
3.
Nat Commun ; 12(1): 3199, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045463

ABSTRACT

In patients with metastatic cancer, spatial heterogeneity of somatic alterations may lead to incomplete assessment of a cancer's mutational profile when analyzing a single tumor biopsy. In this study, we perform sequencing of cell-free DNA (cfDNA) and distinct metastatic tissue samples from ten rapid autopsy cases with pre-treated metastatic cancer. We show that levels of heterogeneity in genetic biomarkers vary between patients but that gene expression signatures representative of the tumor microenvironment are more consistent. Across nine patients with plasma samples available, we are able to detect 62/62 truncal and 47/121 non-truncal point mutations in cfDNA. We observe that mutation clonality in cfDNA is correlated with the number of metastatic lesions in which the mutation is detected and use this result to derive a clonality threshold to classify truncal and non-truncal driver alterations with reasonable specificity. In contrast, mutation truncality is more often incorrectly assigned when studying single tissue samples. Our results demonstrate the utility of a single cfDNA sample relative to that of single tissue samples when treating patients with metastatic cancer.


Subject(s)
Autopsy/methods , Circulating Tumor DNA/genetics , DNA Mutational Analysis/methods , Neoplasms/diagnosis , Tumor Microenvironment/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Chemoradiotherapy, Adjuvant , Cohort Studies , DNA Copy Number Variations , Female , Genetic Heterogeneity , Humans , Male , Neoadjuvant Therapy , Neoplasms/blood , Neoplasms/pathology , Neoplasms/therapy , Point Mutation , RNA-Seq , Reference Values , Sensitivity and Specificity , Spatial Analysis , Time Factors , Exome Sequencing
5.
Cell Rep ; 27(9): 2690-2708.e10, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31141692

ABSTRACT

The detailed molecular characterization of lethal cancers is a prerequisite to understanding resistance to therapy and escape from cancer immunoediting. We performed extensive multi-platform profiling of multi-regional metastases in autopsies from 10 patients with therapy-resistant breast cancer. The integrated genomic and immune landscapes show that metastases propagate and evolve as communities of clones, reveal their predicted neo-antigen landscapes, and show that they can accumulate HLA loss of heterozygosity (LOH). The data further identify variable tumor microenvironments and reveal, through analyses of T cell receptor repertoires, that adaptive immune responses appear to co-evolve with the metastatic genomes. These findings reveal in fine detail the landscapes of lethal metastatic breast cancer.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Genomics/methods , Mutation , Breast Neoplasms/secondary , Female , Gene Expression Profiling , Humans , Loss of Heterozygosity , Neoplasm Metastasis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Exome Sequencing
6.
Nature ; 567(7748): 399-404, 2019 03.
Article in English | MEDLINE | ID: mdl-30867590

ABSTRACT

The rates and routes of lethal systemic spread in breast cancer are poorly understood owing to a lack of molecularly characterized patient cohorts with long-term, detailed follow-up data. Long-term follow-up is especially important for those with oestrogen-receptor (ER)-positive breast cancers, which can recur up to two decades after initial diagnosis1-6. It is therefore essential to identify patients who have a high risk of late relapse7-9. Here we present a statistical framework that models distinct disease stages (locoregional recurrence, distant recurrence, breast-cancer-related death and death from other causes) and competing risks of mortality from breast cancer, while yielding individual risk-of-recurrence predictions. We apply this model to 3,240 patients with breast cancer, including 1,980 for whom molecular data are available, and delineate spatiotemporal patterns of relapse across different categories of molecular information (namely immunohistochemical subtypes; PAM50 subtypes, which are based on gene-expression patterns10,11; and integrative or IntClust subtypes, which are based on patterns of genomic copy-number alterations and gene expression12,13). We identify four late-recurring integrative subtypes, comprising about one quarter (26%) of tumours that are both positive for ER and negative for human epidermal growth factor receptor 2, each with characteristic tumour-driving alterations in genomic copy number and a high risk of recurrence (mean 47-62%) up to 20 years after diagnosis. We also define a subgroup of triple-negative breast cancers in which cancer rarely recurs after five years, and a separate subgroup in which patients remain at risk. Use of the integrative subtypes improves the prediction of late, distant relapse beyond what is possible with clinical covariates (nodal status, tumour size, tumour grade and immunohistochemical subtype). These findings highlight opportunities for improved patient stratification and biomarker-driven clinical trials.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/genetics , Neoplasm Recurrence, Local/classification , Neoplasm Recurrence, Local/genetics , Receptors, Estrogen/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease Progression , Female , Humans , Models, Biological , Neoplasm Metastasis/genetics , Neoplasm Recurrence, Local/pathology , Organ Specificity , Prognosis , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Estrogen/analysis , Receptors, Estrogen/deficiency , Time Factors , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
7.
Breast Cancer Res ; 19(1): 99, 2017 Aug 25.
Article in English | MEDLINE | ID: mdl-28851423

ABSTRACT

BACKGROUND: Patients with BRCA1-like tumors correlate with improved response to DNA double-strand break-inducing therapy. A gene expression-based classifier was developed to distinguish between BRCA1-like and non-BRCA1-like tumors. We hypothesized that these tumors may also be more sensitive to PARP inhibitors than standard treatments. METHODS: A diagnostic gene expression signature (BRCA1ness) was developed using a centroid model with 128 triple-negative breast cancer samples from the EU FP7 RATHER project. This BRCA1ness signature was then tested in HER2-negative patients (n = 116) from the I-SPY 2 TRIAL who received an oral PARP inhibitor veliparib in combination with carboplatin (V-C), or standard chemotherapy alone. We assessed the association between BRCA1ness and pathologic complete response in the V-C and control arms alone using Fisher's exact test, and the relative performance between arms (biomarker × treatment interaction, likelihood ratio p < 0.05) using a logistic model and adjusting for hormone receptor status (HR). RESULTS: We developed a gene expression signature to identify BRCA1-like status. In the I-SPY 2 neoadjuvant setting the BRCA1ness signature associated significantly with response to V-C (p = 0.03), but not in the control arm (p = 0.45). We identified a significant interaction between BRCA1ness and V-C (p = 0.023) after correcting for HR. CONCLUSIONS: A genomic-based BRCA1-like signature was successfully translated to an expression-based signature (BRC1Aness). In the I-SPY 2 neoadjuvant setting, we determined that the BRCA1ness signature is capable of predicting benefit of V-C added to standard chemotherapy compared to standard chemotherapy alone. TRIAL REGISTRATION: I-SPY 2 TRIAL beginning December 31, 2009: Neoadjuvant and Personalized Adaptive Novel Agents to Treat Breast Cancer (I-SPY 2), NCT01042379 .


Subject(s)
BRCA1 Protein/genetics , Biomarkers, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Cluster Analysis , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Neoadjuvant Therapy , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Sensitivity and Specificity , Treatment Outcome , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
9.
Nat Commun ; 7: 11479, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27161491

ABSTRACT

The genomic landscape of breast cancer is complex, and inter- and intra-tumour heterogeneity are important challenges in treating the disease. In this study, we sequence 173 genes in 2,433 primary breast tumours that have copy number aberration (CNA), gene expression and long-term clinical follow-up data. We identify 40 mutation-driver (Mut-driver) genes, and determine associations between mutations, driver CNA profiles, clinical-pathological parameters and survival. We assess the clonal states of Mut-driver mutations, and estimate levels of intra-tumour heterogeneity using mutant-allele fractions. Associations between PIK3CA mutations and reduced survival are identified in three subgroups of ER-positive cancer (defined by amplification of 17q23, 11q13-14 or 8q24). High levels of intra-tumour heterogeneity are in general associated with a worse outcome, but highly aggressive tumours with 11q13-14 amplification have low levels of intra-tumour heterogeneity. These results emphasize the importance of genome-based stratification of breast cancer, and have important implications for designing therapeutic strategies.


Subject(s)
Breast Neoplasms/genetics , Mutation , Adult , Aged , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , DNA Copy Number Variations , Female , Genes, Tumor Suppressor , Genetic Association Studies , Humans , Kaplan-Meier Estimate , Middle Aged , Prognosis , Proportional Hazards Models , Transcriptome
10.
Sci Rep ; 6: 18517, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26729235

ABSTRACT

Invasive lobular carcinoma (ILC) is the second most frequently occurring histological breast cancer subtype after invasive ductal carcinoma (IDC), accounting for around 10% of all breast cancers. The molecular processes that drive the development of ILC are still largely unknown. We have performed a comprehensive genomic, transcriptomic and proteomic analysis of a large ILC patient cohort and present here an integrated molecular portrait of ILC. Mutations in CDH1 and in the PI3K pathway are the most frequent molecular alterations in ILC. We identified two main subtypes of ILCs: (i) an immune related subtype with mRNA up-regulation of PD-L1, PD-1 and CTLA-4 and greater sensitivity to DNA-damaging agents in representative cell line models; (ii) a hormone related subtype, associated with Epithelial to Mesenchymal Transition (EMT), and gain of chromosomes 1q and 8q and loss of chromosome 11q. Using the somatic mutation rate and eIF4B protein level, we identified three groups with different clinical outcomes, including a group with extremely good prognosis. We provide a comprehensive overview of the molecular alterations driving ILC and have explored links with therapy response. This molecular characterization may help to tailor treatment of ILC through the application of specific targeted, chemo- and/or immune-therapies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Genomics , Proteome , Transcriptome , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Carcinoma, Lobular/diagnosis , Cluster Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Genomics/methods , Humans , Immunohistochemistry , Mutation Rate , Polymorphism, Single Nucleotide , Prognosis , Proteomics , Reproducibility of Results , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Phys Biol ; 12(6): 066001, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26403334

ABSTRACT

While the majority of cells in an organism are static and remain relatively immobile in their tissue, migrating cells occur commonly during developmental processes and are crucial for a functioning immune response. The mode of migration has been described in terms of various types of random walks. To understand the details of the migratory behaviour we rely on mathematical models and their calibration to experimental data. Here we propose an approximate Bayesian inference scheme to calibrate a class of random walk models characterized by a specific, parametric particle re-orientation mechanism to observed trajectory data. We elaborate the concept of transition matrices (TMs) to detect random walk patterns and determine a statistic to quantify these TM to make them applicable for inference schemes. We apply the developed pipeline to in vivo trajectory data of macrophages and neutrophils, extracted from zebrafish that had undergone tail transection. We find that macrophage and neutrophils exhibit very distinct biased persistent random walk patterns, where the strengths of the persistence and bias are spatio-temporally regulated. Furthermore, the movement of macrophages is far less persistent than that of neutrophils in response to wounding.


Subject(s)
Cell Movement , Macrophages/physiology , Neutrophils/physiology , Zebrafish/physiology , Animals , Bayes Theorem , Leukocytes/physiology , Models, Biological
12.
Mol Oncol ; 9(8): 1528-38, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26004083

ABSTRACT

Triple negative (TN) breast cancers make up some 15% of all breast cancers. Approximately 10-15% are mutant for the tumor suppressor, BRCA1. BRCA1 is required for homologous recombination-mediated DNA repair and deficiency results in genomic instability. BRCA1-mutated tumors have a specific pattern of genomic copy number aberrations that can be used to classify tumors as BRCA1-like or non-BRCA1-like. BRCA1 mutation, promoter methylation, BRCA1-like status and genome-wide expression data was determined for 112 TN breast cancer samples with long-term follow-up. Mutation status for 21 known DNA repair genes and PIK3CA was assessed. Gene expression and mutation frequency in BRCA1-like and non-BRCA1-like tumors were compared. Multivariate survival analysis was performed using the Cox proportional hazards model. BRCA1 germline mutation was identified in 10% of patients and 15% of tumors were BRCA1 promoter methylated. Fifty-five percent of tumors classified as BRCA1-like. The functions of genes significantly up-regulated in BRCA1-like tumors included cell cycle and DNA recombination and repair. TP53 was found to be frequently mutated in BRCA1-like (P < 0.05), while PIK3CA was frequently mutated in non-BRCA1-like tumors (P < 0.05). A significant association with worse prognosis was evident for patients with BRCA1-like tumors (adjusted HR = 3.32, 95% CI = 1.30-8.48, P = 0.01). TN tumors can be further divided into two major subgroups, BRCA1-like and non-BRCA1-like with different mutation and expression patterns and prognoses. Based on these molecular patterns, subgroups may be more sensitive to specific targeted agents such as PI3K or PARP inhibitors.


Subject(s)
Genes, BRCA1 , Molecular Targeted Therapy/trends , Transcriptome , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Comparative Genomic Hybridization , DNA Methylation , DNA Mutational Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Microarray Analysis , Middle Aged , Promoter Regions, Genetic , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...