Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Breast Cancer Res Treat ; 179(3): 631-642, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31823098

ABSTRACT

BACKGROUND/PURPOSE: TrkA overexpression occurs in over 20% of breast cancers, including triple-negative breast cancers (TNBC), and has recently been recognized as a potential driver of carcinogenesis. Recent clinical trials of pan-Trk inhibitors have demonstrated targeted activity against tumors harboring NTRK fusions, a relatively rare alteration across human cancers. Despite this success, current clinical trials have not investigated TrkA overexpression as an additional therapeutic target for pan-Trk inhibitors. Here, we evaluate the cancerous phenotypes of TrkA overexpression relative to NTRK1 fusions in human cells and assess response to pharmacologic Trk inhibition. EXPERIMENTAL DESIGN/METHODS: To evaluate the clinical utility of TrkA overexpression, a panel of TrkA overexpressing cells were developed via stable transfection of an NTRK1 vector into the non-tumorigenic breast cell lines, MCF10A and hTERT-IMEC. A panel of positive controls was generated via stable transfection with a CD74-NTRK1 fusion vector into MCF10A cells. Cells were assessed via various in vitro and in vivo analyses to determine the transformative potential and targetability of TrkA overexpression. RESULTS: TrkA overexpressing cells demonstrated transformative phenotypes similar to Trk fusions, indicating increased oncogenic potential. TrkA overexpressing cells demonstrated growth factor-independent proliferation, increased PI3Kinase and MAPKinase pathway activation, anchorage-independent growth, and increased migratory capacity. These phenotypes were abrogated by the addition of the pan-Trk inhibitor, larotrectinib. In vivo analysis demonstrated increased tumorgenicity and metastatic potential of TrkA overexpressing breast cancer cells. CONCLUSIONS: Herein, we demonstrate TrkA overexpressing cells show increased tumorgenicity and are sensitive to pan-Trk inhibitors. These data suggest that TrkA overexpression may be an additional target for pan-Trk inhibitors and provide a targeted therapy for breast cancer patients.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression , Oncogenes , Receptor, trkA/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
2.
Breast Cancer Res Treat ; 174(2): 401-412, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30560461

ABSTRACT

PURPOSE: Estrogen receptor-alpha (ER) is a therapeutic target of ER-positive (ER+) breast cancers. Although ER signaling is complex, many mediators of this pathway have been identified. Specifically, phosphorylation of ER at serine 118 affects responses to estrogen and therapeutic ligands and has been correlated with clinical outcomes in ER+ breast cancer patients. We hypothesized that a newly described germline variant (S118P) at this residue would drive cellular changes consistent with breast cancer development and/or hormone resistance. METHODS: Isogenic human breast epithelial cell line models harboring ER S118P were developed via genome editing and characterized to determine the functional effects of this variant. We also examined the frequency of ER S118P in a case-control study (N = 536) of women with and without breast cancer with a familial risk. RESULTS: In heterozygous knock-in models, the S118P variant demonstrated no significant change in proliferation, migration, MAP Kinase pathway signaling, or response to the endocrine therapies tamoxifen and fulvestrant. Further, there was no difference in the prevalence of S118P between women with and without cancer relative to population registry databases. CONCLUSIONS: This study suggests that the ER S118P variant does not affect risk for breast cancer or hormone therapy resistance. Germline screening and modification of treatments for patients harboring this variant are likely not warranted.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/epidemiology , Estrogen Receptor alpha/genetics , Germ-Line Mutation , Adult , Aged , Breast Neoplasms/genetics , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cell Proliferation , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/therapeutic use , Genetic Variation , Humans , Incidence , MCF-7 Cells , Middle Aged , Phosphorylation , Survival Analysis , Tamoxifen/therapeutic use , Treatment Outcome
3.
Proc Natl Acad Sci U S A ; 112(37): 11583-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324937

ABSTRACT

The tumor protein 53 (TP53) tumor suppressor gene is the most frequently somatically altered gene in human cancers. Here we show expression of N-Myc down-regulated gene 1 (NDRG1) is induced by p53 during physiologic low proliferative states, and mediates centrosome homeostasis, thus maintaining genome stability. When placed in physiologic low-proliferating conditions, human TP53 null cells fail to increase expression of NDRG1 compared with isogenic wild-type controls and TP53 R248W knockin cells. Overexpression and RNA interference studies demonstrate that NDRG1 regulates centrosome number and amplification. Mechanistically, NDRG1 physically associates with γ-tubulin, a key component of the centrosome, with reduced association in p53 null cells. Strikingly, TP53 homozygous loss was mutually exclusive of NDRG1 overexpression in over 96% of human cancers, supporting the broad applicability of these results. Our study elucidates a mechanism of how TP53 loss leads to abnormal centrosome numbers and genomic instability mediated by NDRG1.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/ultrastructure , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Aneuploidy , Animals , Breast/metabolism , Cell Line , Cell Proliferation , Centrosome/metabolism , Female , Genome , Heterozygote , Homeostasis , Homozygote , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Neoplasms/pathology , Phenotype , RNA Interference , Tubulin/metabolism
4.
Proc Natl Acad Sci U S A ; 112(45): E6205-14, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26508629

ABSTRACT

Recurrent human epidermal growth factor receptor 2 (HER2) missense mutations have been reported in human cancers. These mutations occur primarily in the absence of HER2 gene amplification such that most HER2-mutant tumors are classified as "negative" by FISH or immunohistochemistry assays. It remains unclear whether nonamplified HER2 missense mutations are oncogenic and whether they are targets for HER2-directed therapies that are currently approved for the treatment of HER2 gene-amplified breast cancers. Here we functionally characterize HER2 kinase and extracellular domain mutations through gene editing of the endogenous loci in HER2 nonamplified human breast epithelial cells. In in vitro and in vivo assays, the majority of HER2 missense mutations do not impart detectable oncogenic changes. However, the HER2 V777L mutation increased biochemical pathway activation and, in the context of a PIK3CA mutation, enhanced migratory features in vitro. However, the V777L mutation did not alter in vivo tumorigenicity or sensitivity to HER2-directed therapies in proliferation assays. Our results suggest the oncogenicity and potential targeting of HER2 missense mutations should be considered in the context of cooperating genetic alterations and provide previously unidentified insights into functional analysis of HER2 mutations and strategies to target them.


Subject(s)
Cell Movement/genetics , Mutation, Missense/genetics , Neoplasms/genetics , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation/physiology , Colony-Forming Units Assay , Flow Cytometry , Gene Targeting , HEK293 Cells , Humans , Immunoblotting , Lapatinib , Quinazolines , Quinolines , Thiazoles
5.
Breast Cancer Res Treat ; 162(3): 451-464, 2017 04.
Article in English | MEDLINE | ID: mdl-28190247

ABSTRACT

BACKGROUND/PURPOSE: The combined contributions of oncogenes and tumor suppressor genes toward carcinogenesis remain poorly understood. Elucidation of cancer gene cooperativity can provide new insights leading to more effective use of therapies. EXPERIMENTAL DESIGN/METHODS: We used somatic cell genome editing to introduce singly and in combination PIK3CA mutations (E545K or H1047R) with TP53 alterations (R248W or knockout), to assess any enhanced cancerous phenotypes. The non-tumorigenic human breast epithelial cell line, MCF10A, was used as the parental cell line, and resultant cells were assessed via various in vitro assays, growth as xenografts, and drug sensitivity assays using targeted agents and chemotherapies. RESULTS: Compared to single-gene-targeted cells and parental controls, cells with both a PIK3CA mutation and TP53 alteration had increased cancerous phenotypes including cell proliferation, soft agar colony formation, aberrant morphology in acinar formation assays, and genomic heterogeneity. Cells also displayed varying sensitivities to anti-neoplastic drugs, although all cells with PIK3CA mutations showed a relative increased sensitivity to paclitaxel. All cell lines remained non-tumorigenic. CONCLUSIONS: This cell line panel provides a resource for further elucidating cooperative genetic mediators of carcinogenesis and response to therapies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Mutation , Phenotype , Tumor Suppressor Protein p53/genetics , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Centromere/genetics , DNA Copy Number Variations , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Amplification , Gene Editing , Gene Knockout Techniques , Genomic Instability , Genotype , Humans , Mice , Paclitaxel/pharmacology
6.
J Natl Compr Canc Netw ; 14(12): 1495-1498, 2016 12.
Article in English | MEDLINE | ID: mdl-27956534

ABSTRACT

Next-generation sequencing (NGS) is increasingly being used in cancer care to identify both somatic tumor driver mutations that can be targeted for therapy, and heritable mutations in the germline associated with increased cancer risk. This report presents a case of a JAK2 V617F mutation falsely identified as a duodenal cancer mutation via NGS. The patient was found to have a history of polycythemia vera, a disorder with a high incidence of JAK2 somatic mutations. Buccal cell DNA showed heterozygosity for the mutation, suggesting that it was potentially germline. However, subsequent resequencing of tumor, adjacent normal tissue, and fingernail DNA confirmed the mutation was somatic, and its presence in tumor and buccal cells resulted from contaminating blood cells. This report highlights important nuances of NGS that can lead to misinterpretation of results with potential clinical implications.


Subject(s)
Adenocarcinoma/diagnosis , DNA Contamination , Duodenal Neoplasms/diagnosis , Janus Kinase 2/genetics , Polycythemia Vera/diagnosis , Abdominal Pain/etiology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Cells , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Chemotherapy, Adjuvant , Diagnosis, Differential , Duodenal Neoplasms/genetics , Duodenal Neoplasms/pathology , Duodenal Neoplasms/therapy , Duodenum/diagnostic imaging , Female , Fluorouracil/therapeutic use , Heterozygote , High-Throughput Nucleotide Sequencing , Hospice Care , Humans , Leucovorin/therapeutic use , Mouth Mucosa/cytology , Mutation , Nails , Organoplatinum Compounds/therapeutic use , Pancreaticoduodenectomy/methods , Phlebotomy , Polycythemia Vera/complications , Polycythemia Vera/genetics , Polycythemia Vera/therapy , Sequence Analysis, DNA , Tomography, X-Ray Computed
7.
Oncotarget ; 13: 373-386, 2022.
Article in English | MEDLINE | ID: mdl-35186194

ABSTRACT

Activating variants in the PEST region of NOTCH1 have been associated with aggressive phenotypes in human cancers, including triple-negative breast cancer (TNBC). Previous studies suggested that PEST domain variants in TNBC patients resulted in increased cell proliferation, invasiveness, and decreased overall survival. In this study, we assess the phenotypic transformation of activating NOTCH1 variants and their response to standard of care therapies. AAV-mediated gene targeting was used to isogenically incorporate 3 NOTCH1 variants, including a novel TNBC frameshift variant, in two non-tumorigenic breast epithelial cell lines, MCF10A and hTERT-IMEC. Two different variants at the NOTCH1 A2241 site (A2441fs and A2441T) both demonstrated increased transformative properties when compared to a non-transformative PEST domain variant (S2523L). These phenotypic changes include proliferation, migration, anchorage-independent growth, and MAPK pathway activation. In contrast to previous studies, activating NOTCH1 variants did not display sensitivity to a gamma secretase inhibitor (GSI) or resistance to chemotherapies. This study demonstrates distinct transformative phenotypes are specific to a given variant within NOTCH1 and these phenotypes do not correlate with sensitivities or resistance to chemotherapies or GSIs. Although previous studies have suggested NOTCH1 variants may be prognostic for TNBC, our study does not demonstrate prognostic ability of these variants and suggests further characterization would be required for clinical applications.


Subject(s)
Triple Negative Breast Neoplasms , Cell Line, Tumor , Cell Proliferation/genetics , Gamma Secretase Inhibitors and Modulators , Humans , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction , Standard of Care , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/therapy
8.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33529175

ABSTRACT

Intratumor heterogeneity is an important mediator of poor outcomes in many cancers, including breast cancer. Genetic subclones frequently contribute to this heterogeneity; however, their growth dynamics and interactions remain poorly understood. PIK3CA and HER2 alterations are known to coexist in breast and other cancers. Herein, we present data that describe the ability of oncogenic PIK3CA mutant cells to induce the proliferation of quiescent HER2 mutant cells through a cell contact-mediated mechanism. Interestingly, the HER2 cells proliferated to become the major subclone over PIK3CA counterparts both in vitro and in vivo. Furthermore, this phenotype was observed in both hormone receptor-positive and -negative cell lines, and was dependent on the expression of fibronectin from mutant PIK3CA cells. Analysis of human tumors demonstrated similar HER2:PIK3CA clonal dynamics and fibronectin expression. Our study provides insight into nonrandom subclonal architecture of heterogenous tumors, which may aid the understanding of tumor evolution and inform future strategies for personalized medicine.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Communication/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Coculture Techniques , Female , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Gene Frequency , Gene Knockout Techniques , Humans , Immunohistochemistry , MCF-7 Cells , Mutation , Phenotype , Receptor, ErbB-2/genetics
9.
Pract Lab Med ; 12: e00108, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30140723

ABSTRACT

BACKGROUND: Genomic testing is often limited by the exhaustible nature of human tissue and blood samples. Here we describe biotinylated amplicon sequencing (BAmSeq), a method that allows for the creation of PCR amplicon based next-generation sequencing (NGS) libraries while retaining the original source DNA. DESIGN AND METHODS: Biotinylated primers for different loci were designed to create NGS libraries using human genomic DNA from cell lines, plasma, and formalin-fixed paraffin embedded (FFPE) tissues using the BAmSeq protocol. DNA from the original template used for each BAmSeq library was recovered after separation with streptavidin magnetic beads. The recovered DNA was then used for end-point, quantitative and droplet digital PCR (ddPCR) as well as NGS using a cancer gene panel. RESULTS: Recovered DNA was analyzed and compared to the original DNA after one or two rounds of BAmSeq. Recovered DNA revealed comparable genomic distributions and mutational allelic frequencies when compared to original source DNA. Sufficient quantities of recovered DNA after BAmSeq were obtained, allowing for additional downstream applications. CONCLUSIONS: We demonstrate that BAmSeq allows original DNA template to be recovered with comparable quality and quantity to the source DNA. This recovered DNA is suitable for many downstream applications and may prevent sample exhaustion, especially when DNA quantity or source material is limiting.

10.
Clin Cancer Res ; 23(16): 4875-4884, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28424200

ABSTRACT

Purpose: Although most human cancers display a single histology, there are unusual cases where two or more distinct tissue types present within a primary tumor. One such example is metaplastic breast carcinoma, a rare but aggressive cancer with a heterogeneous histology, including squamous, chondroid, and spindle cells. Metaplastic carcinomas often contain an admixed conventional ductal invasive or in situ mammary carcinoma component, and are typically triple-negative for estrogen receptor, progesterone receptor, and HER-2 amplification/overexpression. An unanswered question is the origin of metaplastic breast cancers. While they may arise independently from their ductal components, their close juxtaposition favors a model that postulates a shared origin, either as two derivatives from the same primary cancer or one histology as an outgrowth of the other. Understanding the mechanism of development of these tumors may inform clinical decisions.Experimental Design: We performed exome sequencing for paired metaplastic and adjacent conventional invasive ductal carcinomas in 8 patients and created a pipeline to identify somatic variants and predict their functional impact, without having normal tissue. We then determined the genetic relationships between the histologically distinct compartments.Results: In each case, the tumor components have nearly identical landscapes of somatic mutation, implying that the differing histologies do not derive from genetic clonal divergence.Conclusions: A shared origin for tumors with differing histologies suggests that epigenetic or noncoding changes may mediate the metaplastic phenotype and that alternative therapeutic approaches, including epigenetic therapies, may be required for metaplastic breast cancers. Clin Cancer Res; 23(16); 4875-84. ©2017 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast/metabolism , Carcinoma, Ductal, Breast/genetics , Exome Sequencing/methods , Adult , Aged , Aged, 80 and over , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Clone Cells/metabolism , Clone Cells/pathology , DNA Copy Number Variations , Female , Humans , Metaplasia/genetics , Metaplasia/metabolism , Middle Aged , Mutation , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
11.
Genes Dis ; 3(2): 124-129, 2016 Jun.
Article in English | MEDLINE | ID: mdl-30258881

ABSTRACT

Estrogen and estrogen receptor-alpha (ER) signaling are important factors in the pathogenesis and treatment of ER-positive breast cancers. Therefore targeted therapies against ER, known as endocrine therapies, are widely used in the treatment of ER-positive breast cancers. While these therapies have shown great success, de novo and acquired resistance are common. The approach to the problem of endocrine therapy resistance is two-fold: identify the mechanisms of resistance and develop alternative treatments to overcome these mechanisms. This review focuses on the progress and integration of these two aspects of resolving endocrine therapy resistance in ER-positive breast cancer patients.

12.
Clin Cancer Res ; 22(4): 993-9, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26261103

ABSTRACT

PURPOSE: Mutations in the estrogen receptor (ER)α gene, ESR1, have been identified in breast cancer metastases after progression on endocrine therapies. Because of limitations of metastatic biopsies, the reported frequency of ESR1 mutations may be underestimated. Here, we show a high frequency of ESR1 mutations using circulating plasma tumor DNA (ptDNA) from patients with metastatic breast cancer. EXPERIMENTAL DESIGN: We retrospectively obtained plasma samples from eight patients with known ESR1 mutations and three patients with wild-type ESR1 identified by next-generation sequencing (NGS) of biopsied metastatic tissues. Three common ESR1 mutations were queried for using droplet digital PCR (ddPCR). In a prospective cohort, metastatic tissue and plasma were collected contemporaneously from eight ER-positive and four ER-negative patients. Tissue biopsies were sequenced by NGS, and ptDNA ESR1 mutations were analyzed by ddPCR. RESULTS: In the retrospective cohort, all corresponding mutations were detected in ptDNA, with two patients harboring additional ESR1 mutations not present in their metastatic tissues. In the prospective cohort, three ER-positive patients did not have adequate tissue for NGS, and no ESR1 mutations were identified in tissue biopsies from the other nine patients. In contrast, ddPCR detected seven ptDNA ESR1 mutations in 6 of 12 patients (50%). CONCLUSIONS: We show that ESR1 mutations can occur at a high frequency and suggest that blood can be used to identify additional mutations not found by sequencing of a single metastatic lesion.


Subject(s)
Breast Neoplasms/genetics , DNA, Neoplasm/blood , Estrogen Receptor alpha/genetics , Liver Neoplasms/genetics , Adult , Aged , Breast Neoplasms/blood , Breast Neoplasms/pathology , DNA Mutational Analysis , DNA, Neoplasm/genetics , Female , Gene Frequency , Humans , Liver Neoplasms/blood , Liver Neoplasms/secondary , Middle Aged , Mutation, Missense
SELECTION OF CITATIONS
SEARCH DETAIL