Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
NPJ Precis Oncol ; 7(1): 108, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37880313

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that has a poor prognosis. TOP2A is a key enzyme in DNA replication and is a therapeutic target for breast and other cancers. TOP2A-specific Th1-promoting epitopes with optimal binding affinity to MHC II were identified using a combined scoring system. The multi-peptide TOP2A vaccine elicited a robust immunologic response in immunized mice, as demonstrated by the significant production of Th1 cytokines from immunized animals' splenocytes stimulated in vitro with TOP2A peptides. Anti-tumor efficacy of the TOP2A vaccine was demonstrated in a syngeneic TNBC mouse model, in which pre-graft preventive vaccination was associated with significantly decreased tumor growth as compared to adjuvant control. In a genetically engineered mouse (GEM) model of TNBC, vaccinated animals demonstrated a significant reduction in tumor incidence and average tumor volume compared to adjuvant control. Finally, we examined TCR sequences in CD4 tumor Infiltrating lymphocytes (TIL) from vaccinated mice and found that the TIL contained TCR sequences specific to the three vaccine peptides. These data indicate that our newly developed multi-peptide TOP2A vaccine is highly immunogenic, elicits TILs with vaccine specific TCRs, and is highly effective in preventing and intercepting TNBC development and progression in vivo.

2.
Oncologist ; 25(12): 1013-e1824, 2020 12.
Article in English | MEDLINE | ID: mdl-32510664

ABSTRACT

LESSONS LEARNED: Monotherapy with prexasertib demonstrated modest activity in BRCA wild-type, recurrent triple-negative breast cancer, highlighting the unmet need for combination treatment strategies. Neutropenia, anemia, and thrombocytopenia are common with the use of prexasertib but are manageable with supportive care measures. Prophylactic use of granulocyte colony stimulating factor should be considered to avoid dose reductions or treatment delays. Pharmacodynamic studies showed prexasertib treatment induced DNA damage in peripheral immune cells. BACKGROUND: Cell cycle checkpoint kinase 1 (CHK1) is a major G2/M cell cycle regulator in tumors with p53 dysfunction, such as triple-negative breast cancer (TNBC). We hypothesized the second-generation CHK1 inhibitor, prexasertib, would yield clinical activity in sporadic TNBC. METHODS: This single arm, phase II trial evaluated prexasertib at 105 mg/m2 IV every 2 weeks in patients with metastatic/recurrent TNBC. The primary endpoint was overall response rate (ORR). RESULTS: All nine patients enrolled were germline BRCA wild-type (BRCAwt) and had at least one prior treatment. One partial response (PR) was observed (ORR of 11.1%). Four patients experienced stable disease. The median progression-free survival (PFS) was 86 days (range 17 to 159 days). Grade 3/4 treatment-related adverse events included afebrile neutropenia (n = 8; 88.9%), anemia (n = 3; 33.3%), and thrombocytopenia (n = 1; 11.1%). Pharmacodynamic studies showed prexasertib treatment induced DNA damage in peripheral immune cells and demonstrated a decrease in activated/reinvigorated CD8 T cells; however, the one patient with a PR showed evidence of T-cell recovery. CONCLUSION: Prexasertib monotherapy had modest clinical efficacy in BRCAwt TNBC. Further studies of prexasertib in combination with other agents are needed.


Subject(s)
Triple Negative Breast Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Pilot Projects , Pyrazines , Pyrazoles , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
3.
PLoS One ; 14(7): e0219143, 2019.
Article in English | MEDLINE | ID: mdl-31260484

ABSTRACT

Receptor Tyrosine Kinase (RTK) signaling is essential for normal biological processes and disruption of this regulation can lead to tumor initiation and progression. Cbl proteins (Cbl, Cbl-b and Cbl-c) are a family of RING finger (RF) ubiquitin ligases that negatively regulate a variety of RTKs, including EGFR, MET, and RET. Recent studies have identified Cbl mutations associated with human myeloid neoplasias in approximately 5% of the cases. Cbl-c is the most recently identified human Cbl protein and is expressed exclusively in epithelial cells. We identified a novel cDNA that was isolated from a mouse mammary cancer from the C3(1) Large T Antigen transgenic model. This mutant cDNA encodes a protein that has a deletion in the RF domain of Cbl-c, thereby resembling known Cbl family mutations associated with myeoloid neoplasias. Genomic analysis of both parental and transgenic lines shows no evidence of germline mutation indicating that this mutation is likely a somatic mutation. The mutant protein enhances transformation of NIH 3T3 cells when expressed in combination with SV40 Large T antigen. Together these data are consistent with a second hit mutation. In overexpression studies, this mutant Cbl-c protein fails to mediate ubiquitination of activated EGFR and acts in a dominant negative fashion to prevent ubiquitination and downregulation of the activated EGFR by wild type Cbl proteins. Mechanistically, the mutant Cbl-c binds to the EGFR and prevents recruitment of the wild type Cbl protein. Furthermore, data mining reveals Cbl-c mutations associated with solid tumors in humans. Subsequent cell-based analysis demonstrates a similar loss of E3 function and dominant negative effects for one of these human mutations. These data suggest that like Cbl mutations in myeloid neoplasms, loss of Cbl-c function may contribute to the pathogenesis of solid tumors in murine models and in humans.


Subject(s)
Loss of Function Mutation , Neoplasms/genetics , Proto-Oncogene Proteins c-cbl/genetics , Amino Acid Sequence , Animals , Antigens, Viral, Tumor/genetics , Base Sequence , Cell Transformation, Neoplastic/genetics , Female , HEK293 Cells , Humans , Male , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , NIH 3T3 Cells , Neoplasms/metabolism , Proto-Oncogene Proteins c-cbl/chemistry , Proto-Oncogene Proteins c-cbl/metabolism , RING Finger Domains/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Signal Transduction
4.
Heliyon ; 5(1): e01128, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30705983

ABSTRACT

Previously we demonstrated that muscadine grape skin extract (MSKE), a natural product, significantly inhibited androgen-responsive prostate cancer cell growth by inducing apoptosis through the targeting of survival pathways. However, the therapeutic effect of MSKE on more aggressive androgen-independent prostate cancer remains unknown. This study examined the effects of MSKE treatment in metastatic prostate cancer using complementary PC-3 cells and xenograft model. MSKE significantly inhibited PC-3 human prostate cancer cell tumor growth in vitro and in vivo. The growth-inhibitory effect of MSKE appeared to be through the induction of cell-cycle arrest. This induction was accompanied by a reduction in the protein expression of Hsp40 and cell-cycle regulation proteins, cyclin D1 and NF-kBp65. In addition, MSKE induced p21 expression independent of wild-type p53 induced protein expression. Moreover, we demonstrate that MSKE significantly inhibited cell migration in PC-3 prostate cancer cells. Overall, these results demonstrate that MSKE inhibits prostate tumor growth and migration, and induces cell-cycle arrest by targeting Hsp40 and proteins involved in cell-cycle regulation and proliferation. This suggests that MSKE may also be explored either as a neo-adjuvant or therapeutic for castration resistant prostate cancer.

5.
Nat Commun ; 9(1): 1944, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29789598

ABSTRACT

Cancer recurrence after initial diagnosis and treatment is a major cause of breast cancer (BC) mortality, which results from the metastatic outbreak of dormant tumour cells. Alterations in the tumour microenvironment can trigger signalling pathways in dormant cells leading to their proliferation. However, processes involved in the initial and the long-term survival of disseminated dormant BC cells remain largely unknown. Here we show that autophagy is a critical mechanism for the survival of disseminated dormant BC cells. Pharmacologic or genetic inhibition of autophagy in dormant BC cells results in significantly decreased cell survival and metastatic burden in mouse and human 3D in vitro and in vivo preclinical models of dormancy. In vivo experiments identify autophagy gene autophagy-related 7 (ATG7) to be essential for autophagy activation. Mechanistically, inhibition of the autophagic flux in dormant BC cells leads to the accumulation of damaged mitochondria and reactive oxygen species (ROS), resulting in cell apoptosis.


Subject(s)
Autophagy-Related Protein 7/genetics , Autophagy/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Autophagy-Related Protein 7/antagonists & inhibitors , Autophagy-Related Protein 7/metabolism , Beclin-1/antagonists & inhibitors , Beclin-1/genetics , Beclin-1/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen Type I/pharmacology , Female , Humans , Hydroxychloroquine/pharmacology , Lymphatic Metastasis , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Recurrence , Signal Transduction
6.
Cancer Prev Res (Phila) ; 11(7): 383-392, 2018 07.
Article in English | MEDLINE | ID: mdl-29636351

ABSTRACT

Three decades ago, the Garlands postulated that vitamin D3 produced in the skin by ultraviolet radiation (UVR)-induced conversion of 7-dehydrocholesterol to pre-D3 has anticancer effects, thus triggering more than 9,500 publications on D3 and cancer. Here, we report that UVR treatment of transgenic mice of the well-established C3(1)/SV40 Tag mammary cancer model significantly inhibits both autochthonous carcinogenesis and allograft tumor growth, but in contrast neither dietary nor topical D3 influences mammary carcinogenesis in this specific mouse model. Furthermore, UVR's inhibitory effects occur irrespective of whether or not the treatment increases circulating D3 in the mice. The inhibitory effect of UVR on autochthonous tumors occurs at or before the stage of ductal carcinoma in situ. Our studies indicate clearly that UVR can exert D3-independent anticancer effects in C3(1)/SV40 Tag mice. Therefore, supplemental D3 may not mimic all possible beneficial effects of UVR, and uncovering non-D3-mediated mechanisms of UVR tumor inhibition may lead to novel strategies for cancer prevention. Cancer Prev Res; 11(7); 383-92. ©2018 AACR.


Subject(s)
Carcinogenesis/radiation effects , Carcinoma, Intraductal, Noninfiltrating/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Receptors, Estrogen/metabolism , Ultraviolet Rays , Animals , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor/transplantation , Cholecalciferol/metabolism , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Skin/metabolism , Skin/radiation effects
7.
Oncotarget ; 8(19): 30621-30643, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28430642

ABSTRACT

Effective drug development to combat metastatic disease in breast cancer would be aided by the availability of well-characterized preclinical animal models that (a) metastasize with high efficiency, (b) metastasize in a reasonable time-frame, (c) have an intact immune system, and (d) capture some of the heterogeneity of the human disease. To address these issues, we have assembled a panel of twelve mouse mammary cancer cell lines that can metastasize efficiently on implantation into syngeneic immunocompetent hosts. Genomic characterization shows that more than half of the 30 most commonly mutated genes in human breast cancer are represented within the panel. Transcriptomically, most of the models fall into the luminal A or B intrinsic molecular subtypes, despite the predominance of an aggressive, poorly-differentiated or spindled histopathology in all models. Patterns of immune cell infiltration, proliferation rates, apoptosis and angiogenesis differed significantly among models. Inherent within-model variability of the metastatic phenotype mandates large cohort sizes for intervention studies but may also capture some relevant non-genetic sources of variability. The varied molecular and phenotypic characteristics of this expanded panel of models should aid in model selection for development of antimetastatic therapies in vivo, and serve as a useful platform for predictive biomarker identification.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , Mammary Neoplasms, Experimental , Allografts , Animals , Biomarkers, Tumor , Biopsy , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Genomics/methods , Heterografts , Humans , Immunohistochemistry , Mice , Molecular Targeted Therapy , Neoplasm Metastasis , Polymorphism, Single Nucleotide
8.
Cell Death Dis ; 7(12): e2491, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906177

ABSTRACT

Reestablishing tissue organization of breast cancer cells into acini was previously shown to override their malignant phenotype. In our study, we demonstrate that alpha(v)beta(3) integrin (Int-αvß3), previously shown to play a role in cancer progression, promoted differentiation and growth arrest of organoids derived from luminal A breast cancer cells grown in their relevant three-dimensional microenvironment. These organoids differentiated into normal-like acini resembling a benign stage of breast tissue. Likewise, we demonstrate that Int-αvß3 is selectively expressed in the epithelium of the benign stage of breast tissues, and is lost during the early stages of luminal A breast cancer progression. Notably, the organoids' reversion into normal-like acini was mediated by cancer luminal progenitor-like cells expressing both EpCAMhighCD49flowCD24+ and Int-αvß3. Furthermore, downregulation of Notch4 expression and downstream signaling was shown to mediate Int-αvß3-induced reversion. Intriguingly, when luminal A breast cancer cells expressing Int-αvß3 were injected into a humanized mouse model, differentiated tumors developed when compared with that generated by control cells. Hence, our data suggest that promoting differentiation of luminal A breast cancer cells by signaling emanating from Int-αvß3 can potentially promote 'normalization' of their malignant phenotype and may prevent the malignant cells from progressing.


Subject(s)
Breast Neoplasms/pathology , Integrin alphaVbeta3/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Basement Membrane/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Embryonic Stem Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Hyperplasia , MCF-7 Cells , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoids/metabolism , Organoids/pathology , Phenotype , Proto-Oncogene Proteins/metabolism , Receptor, Notch4 , Receptors, Notch/metabolism , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Teratoma/pathology
9.
Oncotarget ; 7(31): 49322-49333, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27384994

ABSTRACT

INTRODUCTION: Multiple gene expression based prognostic biomarkers have been repeatedly identified in gastric carcinoma. However, without confirmation in an independent validation study, their clinical utility is limited. Our goal was to establish a robust database enabling the swift validation of previous and future gastric cancer survival biomarker candidates. RESULTS: The entire database incorporates 1,065 gastric carcinoma samples, gene expression data. Out of 29 established markers, higher expression of BECN1 (HR = 0.68, p = 1.5E-05), CASP3 (HR = 0.5, p = 6E-14), COX2 (HR = 0.72, p = 0.0013), CTGF (HR = 0.72, p = 0.00051), CTNNB1 (HR = 0.47, p = 4.3E-15), MET (HR = 0.63, p = 1.3E-05), and SIRT1 (HR = 0.64, p = 2.2E-07) correlated to longer OS. Higher expression of BIRC5 (HR = 1.45, p = 1E-04), CNTN1 (HR = 1.44, p = 3.5E- 05), EGFR (HR = 1.86, p = 8.5E-11), ERCC1 (HR = 1.36, p = 0.0012), HER2 (HR = 1.41, p = 0.00011), MMP2 (HR = 1.78, p = 2.6E-09), PFKB4 (HR = 1.56, p = 3.2E-07), SPHK1 (HR = 1.61, p = 3.1E-06), SP1 (HR = 1.45, p = 1.6E-05), TIMP1 (HR = 1.92, p = 2.2E- 10) and VEGF (HR = 1.53, p = 5.7E-06) were predictive for poor OS. MATERIALS AND METHODS: We integrated samples of three major cancer research centers (Berlin, Bethesda and Melbourne datasets) and publicly available datasets with available follow-up data to form a single integrated database. Subsequently, we performed a literature search for prognostic markers in gastric carcinomas (PubMed, 2012-2015) and re-validated their findings predicting first progression (FP) and overall survival (OS) using uni- and multivariate Cox proportional hazards regression analysis. CONCLUSIONS: The major advantage of our analysis is that we evaluated all genes in the same set of patients thereby making direct comparison of the markers feasible. The best performing genes include BIRC5, CASP3, CTNNB1, TIMP-1, MMP-2, SIRT, and VEGF.


Subject(s)
Biomarkers, Tumor/metabolism , Stomach Neoplasms/metabolism , Transcriptome , Caspase 3/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Oligonucleotide Array Sequence Analysis , Proportional Hazards Models , Sirtuin 1/metabolism , Survivin , Tissue Inhibitor of Metalloproteinase-1/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism
10.
Anticancer Res ; 36(6): 2649-57, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27272773

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer currently lacking targeted therapies. Our previous work demonstrated a therapeutic synergism with gemcitabine (GEM) and the CHK1 inhibitor (AZD7762) combination treatment in a TNBC cell line. We hypothesized that the response to this combination therapy would differ among heterogeneous TNBC patients and that addition of a SMAC mimetic (TL32711) could improve efficacy. MATERIALS AND METHODS: Therapeutic responses to GEM, GEM/AZD7762, and GEM/AZD7762/TL32711 combinations were investigated by XTT assays and western blotting of cell cycle and apoptosis-related proteins in ten TNBC cell lines. RESULTS: TNBC cell lines harboring low levels of endogenous CHK1, cIAP1 and cIAP2 were responsive to GEM alone, whereas cell lines demonstrating a minimal increase in phospho-S345 CHK1 after treatment were responsive to GEM/AZD7762 or GEM/AZD7762/TL32711 combination. CONCLUSION: The response of TNBC cells to particular therapies varies and will require development of predictive biomarkers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Dipeptides/administration & dosage , Indoles/administration & dosage , Thiophenes/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Urea/analogs & derivatives , Apoptosis/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , DNA Repair , Deoxycytidine/administration & dosage , Female , Humans , Phosphorylation , Triple Negative Breast Neoplasms/pathology , Urea/administration & dosage , Gemcitabine
11.
PLoS One ; 11(5): e0155262, 2016.
Article in English | MEDLINE | ID: mdl-27171183

ABSTRACT

Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is associated with a poor prognosis and for which no targeted therapies currently exist. In order to improve preclinical testing for TNBC that relies primarily on using human xenografts in immunodeficient mice, we have developed a novel immunocompetent syngeneic murine tumor transplant model for basal-like triple-negative breast cancer. The C3(1)/SV40-T/t-antigen (C3(1)/Tag) mouse mammary tumor model in the FVB/N background shares important similarities with human basal-like TNBC. However, these tumors or derived cell lines are rejected when transplanted into wt FVB/N mice, likely due to the expression of SV40 T-antigen. We have developed a sub-line of mice (designated REAR mice) that carry only one copy of the C3(1)/Tag-antigen transgene resulting from a spontaneous transgene rearrangement in the original founder line. Unlike the original C3(1)/Tag mice, REAR mice do not develop mammary tumors or other phenotypes observed in the original C3(1)/Tag transgenic mice. REAR mice are more immunologically tolerant to SV40 T-antigen driven tumors and cell lines in an FVB/N background (including prostate tumors from TRAMP mice), but are otherwise immunologically intact. This transplant model system offers the ability to synchronously implant the C3(1)/Tag tumor-derived M6 cell line or individual C3(1)/Tag tumors from various stages of tumor development into the mammary fat pads or tail veins of REAR mice. C3(1)/Tag tumors or M6 cells implanted into the mammary fat pads spontaneously metastasize at a high frequency to the lung and liver. M6 cells injected by tail vein can form brain metastases. We demonstrate that irradiated M6 tumor cells or the same cells expressing GM-CSF can act as a vaccine to retard tumor growth of implanted tumor cells in the REAR model. Preclinical studies performed in animals with an intact immune system should more authentically replicate treatment responses in human patients.


Subject(s)
Brain Neoplasms/secondary , Immunocompetence , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays , Animals , Antigens, Polyomavirus Transforming/metabolism , Cell Line, Tumor , Female , Gene Dosage , Humans , Lymphocytes/pathology , Male , Mammary Glands, Animal/pathology , Mice, Transgenic , Phenotype , Prostatic Neoplasms/pathology , Spleen/pathology , Transgenes , Tumor Burden , Vaccination
12.
Clin Epigenetics ; 8: 38, 2016.
Article in English | MEDLINE | ID: mdl-27081402

ABSTRACT

BACKGROUND: Oncogene overexpression in primary cells often triggers the induction of a cellular safeguard response promoting senescence or apoptosis. Secondary cooperating genetic events are generally required for oncogene-induced tumorigenesis to overcome these biologic obstacles. We employed comparative genomic hybridization for eight genetically engineered mouse models of mammary cancer to identify loci that might harbor genes that enhance oncogene-induced tumorigenesis. RESULTS: Unlike many other mammary tumor models, the MMTV-Myc tumors displayed few copy number variants except for amplification of distal mouse chromosome 11 in 80 % of the tumors (syntenic to human 17q23-qter often amplified in human breast cancer). Analyses of candidate genes located in this region identified JMJD6 as an epigenetic regulatory gene that cooperates with Myc to enhance tumorigenesis. It suppresses Myc-induced apoptosis under varying stress conditions through inhibition of p19ARF messenger RNA (mRNA) and protein, leading to reduced levels of p53. JMJD6 binds to the p19ARF promoter and exerts its inhibitory function through demethylation of H4R3me2a. JMJD6 overexpression in MMTV-Myc cell lines increases tumor burden, induces EMT, and greatly enhances tumor metastasis. Importantly, we demonstrate that co-expression of high levels of JMJD6 and Myc is associated with poor prognosis for human ER+ breast cancer patients. CONCLUSIONS: A novel epigenetic mechanism has been identified for how JMJD6 cooperates with Myc during oncogenic transformation. Combined high expression of Myc and JMJD6 confers a more aggressive phenotype in mouse and human tumors. Given the pleiotropic pro-tumorigenic activities of JMJD6, it may be useful as a prognostic factor and a therapeutic target for Myc-driven mammary tumorigenesis.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Mammary Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Comparative Genomic Hybridization , Female , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Neoplasm Metastasis , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
13.
Cancer Prev Res (Phila) ; 8(3): 231-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25681088

ABSTRACT

Epidemiologic studies have shown that diabetics receiving the biguanide metformin, as compared with sulfonylureas or insulin, have a lower incidence of breast cancer. Metformin increases levels of activated AMPK (AMP-activated protein kinase) and decreases circulating IGF-1; encouraging its potential use in both cancer prevention and therapeutic settings. In anticipation of clinical trials in nondiabetic women, the efficacy of metformin in nondiabetic rat and mouse mammary cancer models was evaluated. Metformin was administered by gavage or in the diet, at a human equivalent dose, in standard mammary cancer models: (i) methylnitrosourea (MNU)-induced estrogen receptor-positive (ER(+)) mammary cancers in rats, and (ii) MMTV-Neu/p53KO ER(-) (estrogen receptor-negative) mammary cancers in mice. In the MNU rat model, metformin dosing (150 or 50 mg/kg BW/d, by gavage) was ineffective in decreasing mammary cancer multiplicity, latency, or weight. Pharmacokinetic studies of metformin (150 mg/kg BW/d, by gavage) yielded plasma levels (Cmax and AUC) higher than humans taking 1.5 g/d. In rats bearing small palpable mammary cancers, short-term metformin (150 mg/kg BW/d) treatment increased levels of phospho-AMPK and phospho-p53 (Ser20), but failed to reduce Ki67 labeling or expression of proliferation-related genes. In the mouse model, dietary metformin (1,500 mg/kg diet) did not alter final cancer incidence, multiplicity, or weight. Metformin did not prevent mammary carcinogenesis in two mammary cancer models, raising questions about metformin efficacy in breast cancer in nondiabetic populations.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental/drug therapy , Metformin/pharmacology , Alkylating Agents/toxicity , Animals , Biomarkers, Tumor/genetics , Female , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Metformin/pharmacokinetics , Methylnitrosourea/toxicity , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Tumor Cells, Cultured
14.
Mol Carcinog ; 54(11): 1521-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25307412

ABSTRACT

There is a strong need for murine gastric cancer cell line models recapitulating human gastric cancers. Here, we describe two murine gastric cancer cell lines designated as NCC-S1 and NCC-S3. They were generated from gastric adenocarcinomas that formed in a Villin-cre, Smad4(F/F) , Trp53(F/F) , Cdh1(F/wt) mouse and a Pdx1-cre, Trp53(F/F) , Cdh1(F/F) mouse, respectively. Molecular profiles of both cell lines were very similar to human gastric cancer. NCC-S1M and NCC-S3M subpopulation clones were isolated from pulmonary metastasis of heterotopic allografts of NCC-S1 and NCC-S3 cells, respectively. NCC-S1M and NCC-S3M showed enhanced in vivo growth rates and metastatic potentials and exhibited epithelial-to-mesenchymal transition features. NCC-S1M cells developed orthotopic and heterotopic tumors in immunocompetent mice in predictable manner, and were useful for testing the efficacy of an immunotherapeutic agent, anti-4-1BB antibody. NCC-S1M and NCC-S3M cells demonstrated Wnt/ß-catenin pathway activation, and knockdown of Ctnnb1 reversed the metastatic phenotype of NCC-S1M. These results underscore the role of Wnt/ß-catenin pathway in metastatic phenotype of gastric cancer. Taken together, our novel metastatic gastric cancer cell lines are useful resources for drug development and metastasis research.


Subject(s)
Adenocarcinoma/genetics , Cadherins/genetics , Smad4 Protein/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Mice , Mice, SCID , Neoplasm Transplantation/methods , Wnt Signaling Pathway/genetics , beta Catenin/genetics
15.
Anticancer Res ; 34(7): 3807-10, 2014 07.
Article in English | MEDLINE | ID: mdl-24982406

ABSTRACT

BACKGROUND/AIM: The molecular mechanism for aggressive clinical behaviour related to v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2) amplification is not fully-understood. In particular, little is known about microRNAs in the human epidermal growth factor receptor 2 (HER2) signaling network. PATIENTS AND METHODS: Using microRNA microarray, the microRNA profiles of 16 HER2-positive breast carcinomas were compared with those of five luminal-type breast carcinomas. Additionally, two frozen, ERBB2-amplified gastric carcinomas were compared with their adjacent normal tissue samples. MicroRNAs that were differentially expressed according to the HER2 status in breast and gastric carcinomas were identified as the HER2 microRNA signature. RESULTS: MiR-337 and miR-302f were commonly overexpressed in HER2-postive breast and gastric cancer. MiR-139 and miR-129 were commonly underexpressed in HER2-positive breast and gastric cancer. A concordant pattern of microRNA expression was noted between discovery sets and the majority of candidate microRNAs (two out of three) in three validation sets. CONCLUSION: Our study identified novel microRNAs that were differentially expressed according to the HER2 status across different tumor types.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , MicroRNAs/genetics , Receptor, ErbB-2/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Adult , Female , Humans , Male , MicroRNAs/biosynthesis , Middle Aged , Principal Component Analysis , Receptor, ErbB-2/metabolism , Signal Transduction
16.
Mol Cancer Res ; 12(8): 1088-99, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24784840

ABSTRACT

UNLABELLED: Loss of E-cadherin (CDH1), Smad4, and p53 has been shown to play an integral role in gastric, intestinal, and breast cancer formation. Compound conditional knockout mice for Smad4, p53, and E-cadherin were generated to define and compare the roles of these genes in gastric, intestinal, and breast cancer development by crossing with Pdx-1-Cre, Villin-Cre, and MMTV-Cre transgenic mice. Interestingly, gastric adenocarcinoma was significantly more frequent in Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) mice than in Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(+/+) mice, demonstrating that Cdh1 heterozygosity accelerates the development and progression of gastric adenocarcinoma, in combination with loss of Smad4 and p53. Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) mice developed gastric adenocarcinomas without E-cadherin expression. However, intestinal and mammary adenocarcinomas with the same genetic background retained E-cadherin expression and were phenotypically similar to mice with both wild-type Cdh1 alleles. Lung metastases were identified in Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) mice, but not in the other genotypes. Nuclear ß-catenin accumulation was identified at the invasive tumor front of gastric adenocarcinomas arising in Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) mice. This phenotype was less prominent in mice with intact E-cadherin or Smad4, indicating that the inhibition of ß-catenin signaling by E-cadherin or Smad4 downregulates signaling pathways involved in metastases in Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) mice. Knockdown of ß-catenin significantly inhibited the migratory activity of Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) cell lines. Thus, loss of E-cadherin and Smad4 cooperates with p53 loss to promote the development and metastatic progression of gastric adenocarcinomas, with similarities to human gastric adenocarcinoma. IMPLICATIONS: This study demonstrates that inhibition of ß-catenin is a converging node for the antimetastatic signaling pathways driven by E-cadherin and Smad4 in Pdx-1-Cre;Smad4(F/F);Trp53(F/F);Cdh1(F) (/+) mice, providing novel insights into mechanisms for gastric cancer metastasis.


Subject(s)
Adenocarcinoma/genetics , Cadherins/genetics , Neoplasm Metastasis/genetics , Smad4 Protein/genetics , Adenocarcinoma/pathology , Animals , Mice , Neoplasm Metastasis/pathology , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , beta Catenin/genetics
17.
Pharmacogenomics ; 15(3): 375-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24533716

ABSTRACT

Gastric cancer is the second most common cause of cancer death worldwide. Recent development of targeted agents provides clinicians with additional systemic treatment options to conventional cytotoxic agents. Predictive markers are undoubtedly important for guiding the appropriate use of targeted and cytotoxic agents. Currently, however, HER2 is the only predictive biomarker validated for gastric cancer. In this review, candidate predictive markers for response to other targeted agents and cytotoxic chemotherapeutic agents are discussed.


Subject(s)
Biomarkers , Molecular Targeted Therapy , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Antineoplastic Agents/administration & dosage , Cytotoxins/administration & dosage , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
18.
J Clin Invest ; 124(1): 156-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316974

ABSTRACT

Breast cancer (BC) can recur as metastatic disease many years after primary tumor removal, suggesting that disseminated tumor cells survive for extended periods in a dormant state that is refractory to conventional therapies. We have previously shown that altering the tumor microenvironment through fibrosis with collagen and fibronectin deposition can trigger tumor cells to switch from a dormant to a proliferative state. Here, we used an in vivo preclinical model and a 3D in vitro model of dormancy to evaluate the role of Src family kinase (SFK) in regulating this dormant-to-proliferative switch. We found that pharmacological inhibition of SFK signaling or Src knockdown results in the nuclear localization of cyclin-dependent kinase inhibitor p27 and prevents the proliferative outbreak of dormant BC cells and metastatic lesion formation; however, SFK inhibition did not kill dormant cells. Dormant cell proliferation also required ERK1/2 activation. Combination treatment of cells undergoing the dormant-to-proliferative switch with the Src inhibitor (AZD0530) and MEK1/2 inhibitor (AZD6244) induced apoptosis in a large fraction of the dormant cells and delayed metastatic outgrowth, neither of which was observed with either inhibitor alone. Thus, targeting Src prevents the proliferative response of dormant cells to external stimuli, but requires MEK1/2 inhibition to suppress their survival. These data indicate that treatments targeting Src in combination with MEK1/2 may prevent BC recurrence.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Benzodioxoles/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Quinazolines/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Recurrence, Local/prevention & control , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays , src-Family Kinases/genetics , src-Family Kinases/metabolism
19.
Breast Cancer Res Treat ; 141(1): 89-99, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23933800

ABSTRACT

Lysyl oxidase-like 2 (LOXL2) is associated with invasiveness and metastasis in breast cancer. We analyzed the prognostic impact of LOXL2 for breast cancer patients and investigated the role of LOXL2 in breast cancer cell lines. Immunohistochemical study of LOXL2 expression was done in samples from 309 patients. Survival analysis was performed using log-rank test and Cox regression hazard model. After identification of LOXL2 expression in breast cancer cell lines, we performed matrigel invasion and wound-healing assays with LOXL2-silenced cell lines. In the human study, LOXL2 was expressed in 16.2 % of patients. Comparing the LOXL2-positive versus negative groups, there was a significantly higher proportion of estrogen receptor-negative patients (54.0 vs. 37.0 %, respectively; p = 0.029) and triple-negative patients (34.0 vs. 18.0 %; p = 0.022) in the positive group. In multivariate analysis for overall survival and metastasis-free survival, positive LOXL2 was demonstrated as a poor prognostic factor (HR 2.27 and 2.10, respectively). In vitro study indicated that LOXL2 silencing induces a mesenchymal-epithelial transition-like process in basal cell lines (MDA-MB-231 and BT549) associated with decreased invasive and migratory properties. These clinical and preclinical data confirm that higher LOXL2 expression is associated with invasiveness of basal-like breast cancer cells and lower survival of breast cancer patients. Our results suggest the clinical value of LOXL2 as a therapeutic target in breast cancer.


Subject(s)
Amino Acid Oxidoreductases/analysis , Breast Neoplasms/chemistry , Carcinoma/chemistry , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/analysis , Adult , Amino Acid Oxidoreductases/biosynthesis , Amino Acid Oxidoreductases/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/mortality , Carcinoma/pathology , Carcinoma in Situ/chemistry , Carcinoma in Situ/genetics , Carcinoma in Situ/mortality , Carcinoma in Situ/pathology , Cell Line, Tumor , Cell Movement , Collagen , Disease-Free Survival , Drug Combinations , Epithelial-Mesenchymal Transition , Female , Humans , In Situ Hybridization , Kaplan-Meier Estimate , Laminin , Middle Aged , Neoadjuvant Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Multiple Primary/chemistry , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/mortality , Neoplasms, Multiple Primary/pathology , Phyllodes Tumor/chemistry , Phyllodes Tumor/genetics , Phyllodes Tumor/mortality , Phyllodes Tumor/pathology , Prognosis , Proportional Hazards Models , Proteoglycans , RNA Interference , RNA, Small Interfering/pharmacology , Survival Analysis , Tissue Array Analysis , Triple Negative Breast Neoplasms/chemistry , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
20.
PLoS One ; 8(4): e61125, 2013.
Article in English | MEDLINE | ID: mdl-23577196

ABSTRACT

Transforming growth factor (ß1TGFß1) can promote proliferation in late stage cancers but acts as a tumor suppressor in normal epithelial cells and in early stage cancers. Although, the TGFß pathway has been shown to play a key role in tumorigenesis and metastasis, only a limited number of models have been developed to understand this process. Here, we present a novel model system to discern this paradoxical role of TGFß1 using the MDA-MB-231 (MB-231) cell line. The MB-231 triple-negative breast cancer cell line has been extensively characterized and has been shown to continue to proliferate and undergo epithelial-to-mesenchymal transition (EMT) upon TGFß1 stimulation. We have previously shown by microarray analysis that expression of GATA3 in MB-231 cells results in reprogramming of these cells from a basal to a luminal subtype associated with a reduction of metastasis and tumorigenesis when implanted as xenografts. We now demonstrate that GATA3 overexpression in these cells results in a reduction of TGFß1 response, reversal of EMT, and most importantly, restoration of sensitivity to the inhibitory effects on proliferation of TGFß1. Microarray analysis revealed that TGFß1 treatment resulted in reduction of several cell cycle effectors in 231-GATA3 cells but not in control cells. Furthermore, our microarray analysis revealed a significant increase of BMP5 in 231-GATA3 cells. We demonstrate that combined treatment of MB-231 control cells with TGFß1 and BMP5 results in a significant reduction of cellular proliferation. Thus, this model offers a means to further investigate potentially novel mechanisms involved in the switch in response to TGFß1 from tumor promoter to tumor suppressor through the reprogramming of a triple-negative breast cancer cell line by the GATA3 transcription factor.


Subject(s)
Breast Neoplasms/pathology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Transforming Growth Factor beta/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 5/metabolism , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...