Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
J Clin Oncol ; 40(18): 2036-2047, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35263119

ABSTRACT

PURPOSE: Tubo-ovarian cancer (TOC) is a sentinel cancer for BRCA1 and BRCA2 pathogenic variants (PVs). Identification of a PV in the first member of a family at increased genetic risk (the proband) provides opportunities for cancer prevention in other at-risk family members. Although Australian testing rates are now high, PVs in patients with TOC whose diagnosis predated revised testing guidelines might have been missed. We assessed the feasibility of detecting PVs in this population to enable genetic risk reduction in relatives. PATIENTS AND METHODS: In this pilot study, deceased probands were ascertained from research cohort studies, identification by a relative, and gynecologic oncology clinics. DNA was extracted from archival tissue or stored blood for panel sequencing of 10 risk-associated genes. Testing of deceased probands ascertained through clinic records was performed with a consent waiver. RESULTS: We identified 85 PVs in 84 of 787 (11%) probands. Familial contacts of 39 of 60 (65%) deceased probands with an identified recipient (60 of 84; 71%) have received a written notification of results, with follow-up verbal contact made in 85% (33 of 39). A minority of families (n = 4) were already aware of the PV. For many (29 of 33; 88%), the genetic result provided new information and referral to a genetic service was accepted in most cases (66%; 19 of 29). Those who declined referral (4 of 29) were all male next of kin whose family member had died more than 10 years before. CONCLUSION: We overcame ethical and logistic challenges to demonstrate that retrospective genetic testing to identify PVs in previously untested deceased probands with TOC is feasible. Understanding reasons for a family member's decision to accept or decline a referral will be important for guiding future TRACEBACK projects.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Australia , Breast Neoplasms/genetics , Carcinoma, Ovarian Epithelial/genetics , Family , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Male , Ovarian Neoplasms/genetics , Ovarian Neoplasms/prevention & control , Pilot Projects , Retrospective Studies
4.
Cancer Med ; 9(13): 4791-4807, 2020 07.
Article in English | MEDLINE | ID: mdl-32383556

ABSTRACT

We investigated whether a unique immune response was instigated with the development of oral tongue squamous cell carcinomas (OTSCC), with/without nodal involvement, with/without recurrent metastatic disease, or within tumor involved nodes. One hundred and ten formalin-fixed paraffin-embedded samples were collected from a retrospective cohort of 67 OTSCC patients and 10 non-cancerous tongue samples. Targets including CD4, CD8, FOXP3, PD-L1, and PD-1 were analyzed by immunohistochemistry. The Nanostring PanCancer Immune Profiling Panel was used for gene expression profiling. Data were externally validated in the The Cancer Genome Atlas (TCGA) head and neck (HNSCC), melanoma and lung squamous cell carcinoma (LSCC) cohorts. A 24-immune gene signature was identified that discriminated more aggressive OTSCC cases, and although not prognostic in HNSCC was associated with survival in other TCGA cohorts (improved survival for melanoma, P < .001 and worse survival for LSCC, P = .038). OTSCC exhibited concordant gene and immunohistochemical (IHC) features characterized by a TH-2 biased, proinflammatory profile with upregulated B cell and neutrophil gene activity and increased CD4, FOXP3, and PD-L1 expression (P < .001 for all by IHC). Compared to less advanced disease, nodal involvement and recurrent OTSCC did not induce a different immune response although recurrent disease was characterized by significantly higher PD-L1 expression (P = .004 by SP263, P = .013 by 22C3, P = .004 for gene expression). Identification of a gene signature associated with different prognostic effects in other cancers highlights common pathways of immune dysregulation that are impacted by the tumor origin. The significant immunosuppressive signaling in OTSCC indicates primary failure of immune system to control carcinogenesis emphasizing the need for early, combination therapeutic approaches.


Subject(s)
Squamous Cell Carcinoma of Head and Neck/immunology , Tongue Neoplasms/immunology , Aged , B7-H1 Antigen/analysis , CD4 Antigens/analysis , CD8 Antigens/analysis , Female , Forkhead Transcription Factors/analysis , Gene Expression , Gene Expression Profiling , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lymph Nodes/pathology , Male , Melanoma/immunology , Melanoma/mortality , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/analysis , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Tongue/immunology , Tongue Neoplasms/genetics , Tongue Neoplasms/mortality , Tongue Neoplasms/pathology
5.
Biotechniques ; 68(1): 48-51, 2020 01.
Article in English | MEDLINE | ID: mdl-31825238

ABSTRACT

Tumor DNA sequencing results can have important clinical implications. However, its use is often limited by low DNA input, owing to small tumor biopsy size. To help overcome this limitation we have developed a simple improvement to a commonly used next-generation sequencing (NGS) capture-based library preparation method using formalin-fixed paraffin-embedded-derived tumor DNA. By using on-bead PCR for pre-capture library generation we show that library yields are dramatically increased, resulting in decreased sample failure rates. Improved yields allowed for a reduction in PCR cycles, which translated into improved sequencing parameters without affecting variant calling. This methodology should be applicable to any NGS system in which input DNA is a limiting factor.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/methods , Polymerase Chain Reaction/methods , Humans , Neoplasms/genetics , Polymerase Chain Reaction/instrumentation
6.
Clin Cancer Res ; 25(13): 3962-3973, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30967419

ABSTRACT

PURPOSE: Although ovarian clear cell carcinomas (OCCC) are commonly resistant to platinum-based chemotherapy, good clinical outcomes are observed in a subset of patients. The explanation for this is unknown but may be due to misclassification of high-grade serous ovarian cancer (HGSOC) as OCCC or mixed histology. EXPERIMENTAL DESIGN: To discover potential biomarkers of survival benefit following platinum-based chemotherapy, we ascertained a cohort of 68 Japanese and Australian patients in whom progression-free survival (PFS) and overall survival (OS) could be assessed. We performed IHC reclassification of tumors, and targeted sequencing and immunohistochemistry of known driver genes. Exome sequencing was performed in 10 patients who had either unusually long survival (N = 5) or had a very short time to progression (N = 5). RESULTS: The majority of mixed OCCC (N = 6, 85.7%) and a small proportion of pure OCCC (N = 3, 4.9%) were reclassified as likely HGSOC. However, the PFS and OS of patients with misclassified samples were similar to that of patients with pathologically validated OCCC. Absent HNF1B expression was significantly correlated with longer PFS and OS (P = 0.0194 and 0.0395, respectively). Mutations in ARID1A, PIK3CA, PPP2R1A, and TP53 were frequent, but did not explain length of PFS and OS. An exploratory exome analysis of patients with favorable and unfavorable outcomes did not identify novel outcome-associated driver mutations. CONCLUSIONS: Survival benefit following chemotherapy in OCCC was not associated with pathological misclassification of tumor histotype. HNF1B loss may help identify the subset of patients with OCCC with a more favorable outcome.


Subject(s)
Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/etiology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Diagnostic Errors , Female , Humans , Immunohistochemistry , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/etiology , Prognosis , Treatment Outcome
7.
Cancer Res ; 77(16): 4268-4278, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28646021

ABSTRACT

Low-grade serous ovarian carcinomas (LGSC) are associated with a poor response to chemotherapy and are molecularly characterized by RAS pathway activation. Using exome and whole genome sequencing, we identified recurrent mutations in the protein translational regulator EIF1AX and in NF1, USP9X, KRAS, BRAF, and NRAS RAS pathway mutations were mutually exclusive; however, we found significant co-occurrence of mutations in NRAS and EIF1AX Missense EIF1AX mutations were clustered at the N-terminus of the protein in a region associated with its role in ensuring translational initiation fidelity. Coexpression of mutant NRAS and EIF1AX proteins promoted proliferation and clonogenic survival in LGSC cells, providing the first example of co-occurring, growth-promoting mutational events in ovarian cancer. Cancer Res; 77(16); 4268-78. ©2017 AACR.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Eukaryotic Initiation Factor-1/genetics , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Mutation , Ovarian Neoplasms/genetics , Cell Line, Tumor , Cystadenocarcinoma, Serous/pathology , Eukaryotic Initiation Factor-1/biosynthesis , Female , Gene Knockdown Techniques , Humans , Mutagenesis, Site-Directed , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/pathology
8.
Mod Pathol ; 30(7): 952-963, 2017 07.
Article in English | MEDLINE | ID: mdl-28338653

ABSTRACT

The spectrum of genomic alterations in ductal carcinoma in situ (DCIS) is relatively unexplored, but is likely to provide useful insights into its biology, its progression to invasive carcinoma and the risk of recurrence. DCIS (n=20) with a range of phenotypes was assessed by massively parallel sequencing for mutations and copy number alterations and variants validated by Sanger sequencing. PIK3CA mutations were identified in 11/20 (55%), TP53 mutations in 6/20 (30%), and GATA3 mutations in 9/20 (45%). Screening an additional 91 cases for GATA3 mutations identified a final frequency of 27% (30/111), with a high proportion of missense variants (8/30). TP53 mutations were exclusive to high grade DCIS and more frequent in PR-negative tumors compared with PR-positive tumors (P=0.037). TP53 mutant tumors also had a significantly higher fraction of the genome altered by copy number than wild-type tumors (P=0.005), including a significant positive association with amplification or gain of ERBB2 (P<0.05). The association between TP53 mutation and ERBB2 amplification was confirmed in a wider DCIS cohort using p53 immunohistochemistry as a surrogate marker for TP53 mutations (P=0.03). RUNX1 mutations and MAP2K4 copy number loss were novel findings in DCIS. Frequent copy number alterations included gains on 1q, 8q, 17q, and 20q and losses on 8p, 11q, 16q, and 17p. Patterns of genomic alterations observed in DCIS were similar to those previously reported for invasive breast cancers, with all DCIS having at least one bona fide breast cancer driver event. However, an increase in GATA3 mutations and fewer copy number changes were noted in DCIS compared with invasive carcinomas. The role of such alterations as prognostic and predictive biomarkers in DCIS is an avenue for further investigation.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Mutation , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , DNA Copy Number Variations , Female , GATA3 Transcription Factor/genetics , Humans , Middle Aged , Receptor, ErbB-2/genetics , Tumor Suppressor Protein p53/genetics
9.
Genome Med ; 8(1): 121, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27846907

ABSTRACT

Unlocking clinically translatable genomic information, including copy number alterations (CNA), from formalin-fixed paraffin-embedded (FFPE) tissue is challenging due to low yields and degraded DNA. We describe a robust, cost-effective low-coverage whole genome sequencing (LC WGS) method for CNA detection using 5 ng of FFPE-derived DNA. CN profiles using 100 ng or 5 ng input DNA were highly concordant and comparable with molecular inversion probe (MIP) array profiles. LC WGS improved CN profiles of samples that performed poorly using MIP arrays. Our technique enables identification of driver and prognostic CNAs in archival patient samples previously deemed unsuitable for genomic analysis due to DNA limitations.


Subject(s)
DNA/analysis , Formaldehyde/chemistry , Gene Dosage , Breast Neoplasms/pathology , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cohort Studies , Cost-Benefit Analysis , DNA Copy Number Variations , DNA Repair , Female , Gene Library , Genome, Human , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Oligonucleotide Array Sequence Analysis , Paraffin/chemistry , Polymorphism, Single Nucleotide , Prognosis , Sequence Analysis, DNA
10.
PLoS One ; 10(9): e0136817, 2015.
Article in English | MEDLINE | ID: mdl-26334628

ABSTRACT

The development of antigen-targeted therapeutics is dependent on the preferential expression of tumor-associated antigens (TAA) at targetable levels on the tumor. Tumor-associated antigens can be generated de novo or can arise from altered expression of normal basal proteins, such as the up-regulation of human epidermal growth factor receptor 2 (Her2/ErbB2). To properly assess the development of Her2 therapeutics in an immune tolerant model, we previously generated a transgenic mouse model in which expression of the human Her2 protein was present in both the brain and mammary tissue. This mouse model has facilitated the development of Her2 targeted therapies in a clinically relevant and suitable model. While heterozygous Her2+/- mice appear to develop in a similar manner to wild type mice (Her2-/-), it has proven difficult to generate homozygous Her2+/+ mice, potentially due to embryonic lethality. In this study, we performed whole genome sequencing to determine if the integration site of the Her2 transgene was responsible for this lethality. Indeed, we report that the Her2 transgene had integrated into the Pds5b (precocious dissociation of sisters) gene on chromosome 5, as a 162 copy concatemer. Furthermore, our findings demonstrate that Her2+/+ mice, similar to Pds5b-/- mice, are embryonic lethal and confirm the necessity for Pds5b in embryonic development. This study confirms the value of whole genome sequencing in determining the integration site of transgenes to gain insight into associated phenotypes.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Lethal , Genes, erbB-2 , Homozygote , Transcription Factors/genetics , Animals , Cell Line, Tumor , Chromosome Mapping , Exons , Fetal Death , Humans , Mice , Mice, Transgenic , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...