Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Breast Cancer Res ; 24(1): 40, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35681213

ABSTRACT

BACKGROUND: The Bcl-3 protein is an atypical member of the inhibitor of -κB family that has dual roles as a transcriptional repressor and a coactivator for dimers of NF-κB p50 and p52. Bcl-3 is expressed in mammary adenocarcinomas and can promote tumorigenesis and survival signaling and has a key role in tumor metastasis. In this study, we have investigated the role of Bcl-3 in the normal mammary gland and impact on tumor pathology. METHODS: We utilized bcl-3-/- mice to study mammary gland structure in virgins and during gestation, lactation and early involution. Expression of involution-associated genes and proteins and putative Bcl-3 target genes was examined by qRT-PCR and immunoblot analysis. Cell autonomous branching morphogenesis and collagen I invasion properties of bcl-3-/- organoids were tested in 3D hydrogel cultures. The role of Bcl-3 in tumorigenesis and tumor pathology was also assessed using a stochastic carcinogen-induced mammary tumor model. RESULTS: Bcl-3-/- mammary glands demonstrated reduced branching complexity in virgin and pregnant mice. This defect was recapitulated in vitro where significant defects in bud formation were observed in bcl-3-/- mammary organoid cultures. Bcl-3-/- organoids showed a striking defect in protrusive collective fibrillary collagen I invasion associated with reduced expression of Fzd1 and Twist2. Virgin and pregnant bcl-3-/- glands showed increased apoptosis and rapid increases in lysosomal cell death and apoptosis after forced weaning compared to WT mice. Bcl-2 and Id3 are strongly induced in WT but not bcl-3-/- glands in early involution. Tumors in WT mice were predominately adenocarcinomas with NF-κB activation, while bcl-3-/- lesions were largely squamous lacking NF-κB and with low Bcl-2 expression. CONCLUSIONS: Collectively, our results demonstrate that Bcl-3 has a key function in mammary gland branching morphogenesis, in part by regulation of genes involved in extracellular matrix invasion. Markedly reduced levels of pro-survival proteins expression in bcl-3 null compared to WT glands 24 h post-weaning indicate that Bcl-3 has a role in moderating the rate of early phase involution. Lastly, a reduced incidence of bcl-3-/- mammary adenocarcinomas versus squamous lesions indicates that Bcl-3 supports the progression of epithelial but not metaplastic cancers.


Subject(s)
Adenocarcinoma , B-Cell Lymphoma 3 Protein , Breast Neoplasms , Carcinoma, Squamous Cell , Mammary Glands, Animal , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , B-Cell Lymphoma 3 Protein/metabolism , Breast Neoplasms/pathology , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/pathology , Collagen/metabolism , Epithelial Cells/metabolism , Female , Lactation , Mammary Glands, Animal/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Pregnancy , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
2.
Exp Cell Res ; 371(1): 83-91, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30059665

ABSTRACT

SIRT1 is a protein deacetylase with a broad range of biological functions, many of which are known to be important in carcinogenesis, however much of the literature regarding the role of SIRT1 in cancer remains conflicting. In this study we assessed the effect of SIRT1 on the initiation and progression of thymic T cell lymphomas. We employed mouse strains in which SIRT1 activity was absent or could be reversibly modulated in conjunction with thymic lymphoma induction using either the N-nitroso-N-methylurea (NMU) carcinogenesis or the nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) transgene. Decreased SIRT1 activity reduced the development of thymic lymphomas in the NMU-treated mice but was permissive for the formation of lung adenomas. Conversely, in the NPM-ALK transgenic mice, decreased SIRT1 activity had a modest promoting effect in the development of thymic lymphomas. The results of the work presented here add to the growing body of evidence that sirt1 is neither an outright oncogene nor a tumor suppressor. These opposing results in two models of the same disease suggest that the influence of sirt1 on carcinogenesis may lie in a role in tumor surveillance.


Subject(s)
Adenocarcinoma of Lung/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell/genetics , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Sirtuin 1/genetics , Thymus Neoplasms/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/etiology , Adenocarcinoma of Lung/mortality , Administration, Oral , Animals , Antineoplastic Agents, Hormonal/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/mortality , Male , Methylnitrosourea/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/metabolism , Organ Specificity , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Sirtuin 1/metabolism , Survival Analysis , Tamoxifen/pharmacology , Thymus Gland/drug effects , Thymus Gland/metabolism , Thymus Gland/pathology , Thymus Neoplasms/drug therapy , Thymus Neoplasms/etiology , Thymus Neoplasms/mortality , Transfection
3.
J Mammary Gland Biol Neoplasia ; 23(3): 109-123, 2018 09.
Article in English | MEDLINE | ID: mdl-29876871

ABSTRACT

Cellular inhibitor of apoptosis proteins-1 and -2 (cIAP1/2) are integral to regulation of apoptosis and signaling by the tumor necrosis factor (TNF) and related family of receptors. The expression of cIAP2 in tissues is typically low and considered functionally redundant with cIAP1, however cIAP2 can be activated by a variety of cellular stresses. Members of the TNFR family and their ligands have essential roles in mammary gland biology. We have found that cIAP2-/- virgin mammary glands have reduced ductal branching and delayed lobuloalveogenesis in early pregnancy. Post-lactational involution involves two phases where the first phase is reversible and is mediated, in part, by TNFR family ligands. In cIAP2-/- mice mammary glands appeared engorged at mid-lactation accompanied by enhanced autophagic flux and decreased cIAP1 protein expression. Severely stretched myoepithelium was associated with BIM-EL expression and other indicators of anoikis. Within 24 h after forced or natural weaning, cIAP2-/- glands had nearly completed involution. The TNF-related weak inducer of apoptosis (Tweak) which results in degradation of cIAP1 through its receptor, Fn14, began to increase in late lactation and was significantly increased in cIAP2-/- relative to WT mice by 12 h post weaning accompanied by decreased cIAP1 protein expression. Carcinogen/progesterone-induced mammary tumorigenesis was significantly delayed in cIAP2-/- mice and tumors contained high numbers of apoptotic cells. We conclude that cIAP2 has a critical role in the mammary gland wherein it prevents rapid involution induced by milk stasis-induced stress associated with Tweak activation and contributes to the survival of mammary tumor cells.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Carcinogenesis/metabolism , Lactation/metabolism , Mammary Glands, Animal/metabolism , Signal Transduction/physiology , Animals , Apoptosis , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy , Weaning
4.
Cytotherapy ; 20(1): 108-125, 2018 01.
Article in English | MEDLINE | ID: mdl-29056548

ABSTRACT

BACKGROUND AIMS: Bronchopulmonary dysplasia (BPD), a chronic lung disease characterized by disrupted lung growth, is the most common complication in extreme premature infants. BPD leads to persistent pulmonary disease later in life. Alveolar epithelial type 2 cells (AEC2s), a subset of which represent distal lung progenitor cells (LPCs), promote normal lung growth and repair. AEC2 depletion may contribute to persistent lung injury in BPD. We hypothesized that induced pluripotent stem cell (iPSC)-derived AECs prevent lung damage in experimental oxygen-induced BPD. METHODS: Mouse AECs (mAECs), miPSCs/mouse embryonic stem sells, human umbilical cord mesenchymal stromal cells (hUCMSCs), human (h)iPSCs, hiPSC-derived LPCs and hiPSC-derived AECs were delivered intratracheally to hyperoxia-exposed newborn mice. Cells were pre-labeled with a red fluorescent dye for in vivo tracking. RESULTS: Airway delivery of primary mAECs and undifferentiated murine pluripotent cells prevented hyperoxia-induced impairment in lung function and alveolar growth in neonatal mice. Similar to hUCMSC therapy, undifferentiated hiPSCs also preserved lung function and alveolar growth in hyperoxia-exposed neonatal NOD/SCID mice. Long-term assessment of hiPSC administration revealed local teratoma formation and cellular infiltration in various organs. To develop a clinically relevant cell therapy, we used a highly efficient method to differentiate hiPSCs into a homogenous population of AEC2s. Airway delivery of hiPSC-derived AEC2s and hiPSC-derived LPCs, improved lung function and structure and resulted in long-term engraftment without evidence of tumor formation. CONCLUSIONS: hiPSC-derived AEC2 therapy appears effective and safe in this model and warrants further exploration as a therapeutic option for BPD and other lung diseases characterized by AEC injury.


Subject(s)
Alveolar Epithelial Cells/cytology , Hyperoxia/complications , Induced Pluripotent Stem Cells/cytology , Lung Injury/etiology , Lung Injury/therapy , Animals , Animals, Newborn , Cell Differentiation , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/ultrastructure , Lung Injury/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Oxygen , Teratoma/pathology
5.
Nature ; 477(7362): 99-102, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21886163

ABSTRACT

The efficacy and safety of biological molecules in cancer therapy, such as peptides and small interfering RNAs (siRNAs), could be markedly increased if high concentrations could be achieved and amplified selectively in tumour tissues versus normal tissues after intravenous administration. This has not been achievable so far in humans. We hypothesized that a poxvirus, which evolved for blood-borne systemic spread in mammals, could be engineered for cancer-selective replication and used as a vehicle for the intravenous delivery and expression of transgenes in tumours. JX-594 is an oncolytic poxvirus engineered for replication, transgene expression and amplification in cancer cells harbouring activation of the epidermal growth factor receptor (EGFR)/Ras pathway, followed by cell lysis and anticancer immunity. Here we show in a clinical trial that JX-594 selectively infects, replicates and expresses transgene products in cancer tissue after intravenous infusion, in a dose-related fashion. Normal tissues were not affected clinically. This platform technology opens up the possibility of multifunctional products that selectively express high concentrations of several complementary therapeutic and imaging molecules in metastatic solid tumours in humans.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Poxviridae/physiology , Adult , Aged , Aged, 80 and over , DNA, Viral/blood , Female , Gene Expression Regulation, Enzymologic , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/pathology , Neoplasms/surgery , Neoplasms/virology , Organisms, Genetically Modified/physiology , Transgenes/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
6.
Mol Ther ; 23(1): 202-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25292189

ABSTRACT

Oncolytic viral therapy utilizes a tumor-selective replicating virus which preferentially infects and destroys cancer cells and triggers antitumor immunity. The Western Reserve strain of vaccinia virus (VV) is the most virulent strain of VV in animal models and has been engineered for tumor selectivity through two targeted gene deletions (vvDD). We performed the first-in-human phase 1, intratumoral dose escalation clinical trial of vvDD in 16 patients with advanced solid tumors. In addition to safety, we evaluated signs of vvDD replication and spread to distant tumors, pharmacokinetics and pharmacodynamics, clinical and immune responses to vvDD. Dose escalation proceeded without dose-limiting toxicities to a maximum feasible dose of 3 × 10(9) pfu. vvDD replication in tumors was reproducible. vvDD genomes and/or infectious particles were recovered from injected (n = 5 patients) and noninjected (n = 2 patients) tumors. At the two highest doses, vvDD genomes were detected acutely in blood in all patients while delayed re-emergence of vvDD genomes in blood was detected in two patients. Fifteen of 16 patients exhibited late symptoms, consistent with ongoing vvDD replication. In summary, intratumoral injection of the oncolytic vaccinia vvDD was well-tolerated in patients and resulted in selective infection of injected and noninjected tumors and antitumor activity.


Subject(s)
Breast Neoplasms/therapy , Colonic Neoplasms/therapy , Melanoma/therapy , Pancreatic Neoplasms/therapy , Skin Neoplasms/therapy , Vaccinia virus/immunology , Virus Replication/genetics , Aged , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dose-Response Relationship, Immunologic , Female , Gene Deletion , Humans , Injections, Intralesional , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/growth & development , Oncolytic Viruses/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Vaccinia virus/genetics , Vaccinia virus/growth & development
7.
Breast Cancer Res ; 17: 7, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25592291

ABSTRACT

INTRODUCTION: Periostin (Postn) is a secreted cell adhesion protein that activates signaling pathways to promote cancer cell survival, angiogenesis, invasion, and metastasis. Interestingly, Postn is frequently overexpressed in numerous human cancers, including breast, lung, colon, pancreatic, and ovarian cancer. METHODS: Using transgenic mice expressing the Neu oncogene in the mammary epithelium crossed into Postn-deficient animals, we have assessed the effect of Postn gene deletion on Neu-driven mammary tumorigenesis. RESULTS: Although Postn is exclusively expressed in the stromal fibroblasts of the mammary gland, Postn deletion does not affect mammary gland outgrowth during development or pregnancy. Furthermore, we find that loss of Postn in the mammary epithelium does not alter breast tumor initiation or growth in mouse mammary tumor virus (MMTV)-Neu expressing mice but results in an apocrine-like tumor phenotype. Surprisingly, we find that tumors derived from Postn-null animals express low levels of Notch protein and Hey1 mRNA but increased expression of androgen receptor (AR) and AR target genes. We show that tumor cells derived from wild-type animals do not proliferate when transplanted in a Postn-null environment but that this growth defect is rescued by the overexpression of active Notch or the AR target gene prolactin-induced protein (PIP/GCDFP-15). CONCLUSIONS: Together our data suggest that loss of Postn in an ErbB2/Neu/HER2 overexpression model results in apocrine-like tumors that activate an AR-dependent pathway. This may have important implications for the treatment of breast cancers involving the therapeutic targeting of periostin or Notch signaling.


Subject(s)
Cell Adhesion Molecules/deficiency , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, Notch1/metabolism , Receptors, Androgen/metabolism , Sweat Gland Neoplasms/genetics , Sweat Gland Neoplasms/metabolism , Animals , Apocrine Glands/metabolism , Apocrine Glands/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Gene Expression , Genotype , Humans , Immunohistochemistry , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Sweat Gland Neoplasms/mortality , Sweat Gland Neoplasms/pathology , Tumor Burden
8.
Mol Ther ; 20(6): 1148-57, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22273579

ABSTRACT

Replicating viruses for the treatment of cancer have a number of advantages over traditional therapeutic modalities. They are highly targeted, self-amplifying, and have the added potential to act as both gene-therapy delivery vehicles and oncolytic agents. Parapoxvirus ovis or Orf virus (ORFV) is the prototypic species of the Parapoxvirus genus, causing a benign disease in its natural ungulate host. ORFV possesses a number of unique properties that make it an ideal viral backbone for the development of a cancer therapeutic: it is safe in humans, has the ability to cause repeat infections even in the presence of antibody, and it induces a potent T(h)-1-dominated immune response. Here, we show that live replicating ORFV induces an antitumor immune response in multiple syngeneic mouse models of cancer that is mediated largely by the potent activation of both cytokine-secreting, and tumoricidal natural killer (NK) cells. We have also highlighted the clinical potential of the virus by demonstration of human cancer cell oncolysis including efficacy in an A549 xenograft model of cancer.


Subject(s)
Genetic Vectors/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Orf virus/immunology , Animals , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Genetic Therapy , Genetic Vectors/adverse effects , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Lung/immunology , Lung/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/genetics , Oncolytic Viruses/genetics , Orf virus/genetics , Spleen/immunology , Spleen/metabolism , Tumor Burden , Virus Replication , Xenograft Model Antitumor Assays
9.
Mol Ther ; 19(10): 1913-22, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21772252

ABSTRACT

JX-594 is a targeted and granulocyte macrophage-colony stimulating factor (GM-CSF)-expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In order to study the mechanisms-of-action (MOA) of JX-594 in humans, a mechanistic proof-of-concept clinical trial was performed at a low dose equivalent to ≤10% of the maximum-tolerated dose (MTD) in other clinical trials. Ten patients with previously treated stage IV melanoma were enrolled. Tumors were injected weekly for up to nine total treatments. Blood samples and tumor biopsies were analyzed for evidence of transgene activity, virus replication, and immune stimulation. The ß-galactosidase (ß-gal) transgene was expressed in all patients as evidenced by antibody induction. Six patients had significant induction of GM-CSF-responsive white blood cell (WBC) subsets such as neutrophils (25-300% increase). JX-594 replication and subsequent shedding into blood was detectable in five patients after cycles 1-9. Tumor biopsies demonstrated JX-594 replication, perivascular lymphocytic infiltration, and diffuse tumor necrosis. Mild flu-like symptoms were the most common adverse events. In sum, JX-594 replication, oncolysis, and expression of both transgenes were demonstrated; replication was still evident after multiple cycles. These findings have implications for further clinical development of JX-594 and other transgene-armed oncolytic viruses.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma/therapy , Oncolytic Virotherapy , Poxviridae/genetics , Adult , Aged , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Poxviridae/physiology , Transgenes
10.
Mol Ther ; 19(5): 886-94, 2011 May.
Article in English | MEDLINE | ID: mdl-21364541

ABSTRACT

Oncolytic viruses (OVs) have been engineered or selected for cancer cell-specific infection however, we have found that following intravenous administration of vesicular stomatitis virus (VSV), tumor cell killing rapidly extends far beyond the initial sites of infection. We show here for the first time that VSV directly infects and destroys tumor vasculature in vivo but leaves normal vasculature intact. Three-dimensional (3D) reconstruction of infected tumors revealed that the majority of the tumor mass lacks significant blood flow in contrast to uninfected tumors, which exhibit relatively uniform perfusion. VSV replication in tumor neovasculature and spread within the tumor mass, initiates an inflammatory reaction including a neutrophil-dependent initiation of microclots within tumor blood vessels. Within 6 hours of intravenous administration of VSV and continuing for at least 24 hours, we observed the initiation of blood clots within the tumor vasculature whereas normal vasculature remained clot free. Blocking blood clot formation with thrombin inhibitors prevented tumor vascular collapse. Our results demonstrate that the therapeutic activity of an OV can go far beyond simple infection and lysis of malignant cells.


Subject(s)
Adenocarcinoma/blood supply , Adenocarcinoma/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Vesicular stomatitis Indiana virus , Adenocarcinoma/genetics , Animals , Blood Coagulation , Cell Line, Tumor , Cell Proliferation , Mice , Mice, Inbred BALB C , Neutrophils , Thrombin/antagonists & inhibitors
11.
J Biophotonics ; 15(2): e202100198, 2022 02.
Article in English | MEDLINE | ID: mdl-34837331

ABSTRACT

Up to 70% of ovarian cancer patients are diagnosed with advanced-stage disease and the degree of cytoreduction is an important survival prognostic factor. The aim of this study was to evaluate if Raman spectroscopy could detect cancer from different organs within the abdominopelvic region, including the ovaries. A Raman spectroscopy probe was used to interrogate specimens from a cohort of nine patients undergoing cytoreductive surgery, including four ovarian cancer patients and three patients with endometrial cancer. A feature-selection algorithm was developed to determine which spectral bands contributed to cancer detection and a machine-learning model was trained. The model could detect cancer using only eight spectral bands. The receiver-operating-characteristic curve had an area-under-the-curve of 0.96, corresponding to an accuracy, a sensitivity and a specificity of 90%, 93% and 88%, respectively. These results provide evidence multispectral Raman spectroscopy could be developed to detect ovarian cancer intraoperatively.


Subject(s)
Endometrial Neoplasms , Ovarian Neoplasms , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/surgery , Female , Humans , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/surgery , ROC Curve , Spectrum Analysis, Raman/methods
12.
Transl Oncol ; 14(2): 100996, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33341488

ABSTRACT

BACKGROUND: Signal Transducer and Activator of Transcription-3 (STAT3) mediates cellular functions. We assessed the IHC expression of phosphorylated STAT3 (pSTAT3) in paired primary tumors and liver metastases in patients with advanced stage colorectal cancer (CRC). METHODS: We included patients with tissue blocks available from both the primary CRC and a surgically resected liver metastasis. The IHC pSTAT3 expression agreement was measured using Cohen's kappa statistic. RESULTS: The study included 103 patients, 55% male, median age was 64. 43% tumors originated in rectum, and 63% of the primary tumors were synchronous. Expression of pSTAT3 was 76% in liver metastases and 71% in primary tumors. A difference in pSTAT3 staining between the primary tumor and liver metastases was noted in 64%. There was lost expression of pSTAT3 in the liver metastases in 28% and gained expression in 36% of cases compared to the primary. The kappa statistic comparing agreement between staining patterns of the primary tumors and liver metastases was a "less-than-chance", at -0.02. Median survival was 4.9 years, with no difference in survival outcomes by pSTAT3 expression in the primary tumor or liver metastases. DISCUSSION: STAT3 is not a prognostic marker in the selective setting of metastatic CRC to liver, but it may remain a potential therapeutic target given most liver metastases expressed pSTAT3. Discordant pSTAT3 expression in between primary tumors and paired liver metastases suggests that use of this class of drug to treat liver predominant metastatic colorectal cancer in a biomarker-driven approach may require confirmatory liver tumor biopsy.

13.
Lancet Oncol ; 9(6): 533-42, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18495536

ABSTRACT

BACKGROUND: JX-594 is a targeted oncolytic poxvirus designed to selectively replicate in and destroy cancer cells with cell-cycle abnormalities and epidermal growth factor receptor (EGFR)-ras pathway activation. Direct oncolysis plus granulocyte-macrophage colony-stimulating factor (GM-CSF) expression also stimulates shutdown of tumour vasculature and antitumoral immunity. We aimed to assess intratumoral injection of JX-594 in patients with refractory primary or metastatic liver cancer. METHODS: Between Jan 4, 2006, and July 4, 2007, 14 patients with histologically confirmed refractory primary or metastatic liver tumours (up to 10.9 cm total diameter) that were amenable to image-guided intratumoral injections were enrolled into this non-comparative, open-label, phase I dose-escalation trial (standard 3x3 design; two to six patients for each dose with 12-18 estimated total patients). Patients received one of four doses of intratumoral JX-594 (10(8) plaque-forming units [pfu], 3x10(8) pfu, 10(9) pfu, or 3x10(9) pfu) every 3 weeks at Dong-A University Hospital (Busan, South Korea). Patients were monitored after treatment for at least 48 h in hospital and for at least 4 weeks as out-patients. Adverse event-monitoring according to the National Cancer Institute Common Toxicity Criteria (version 3) and standard laboratory toxicity grading for haematology, liver and renal function, coagulation studies, serum chemistry, and urinalysis were done. The primary aims were to ascertain the maximum-tolerated dose (MTD) and safety of JX-594 treatment. Data were also collected on pharmacokinetics, pharmacodynamics, and efficacy. Analysis was per protocol. This study is registered with ClinicalTrials.gov, number NCT00629759. FINDINGS: Of 22 patients with liver tumours who were assessed for eligibility, eight patients did not meet inclusion criteria. Therefore, 14 patients, including those with hepatocellular, colorectal, melanoma, and lung cancer, were enrolled. Patients were heavily pretreated (5.6 previous treatments, SD 2.8, range 2.0-12.0) and had large tumours (7.0 cm diameter, SD 2.7, range 1.8-10.9). Patients received a mean of 3.4 (SD 2.2, range 1.0-8.0) cycles of JX-594. All patients were evaluable for toxicity. All patients experienced grade I-III flu-like symptoms, and four had transient grade I-III dose-related thrombocytopenia. Grade III hyperbilirubinaemia was dose-limiting in both patients at the highest dose; the MTD was therefore 1x10(9) pfu. JX-594 replication-dependent dissemination in blood was shown, with resultant infection of non-injected tumour sites. GM-CSF expression resulted in grade I-III increases in neutrophil counts in four of six patients at the MTD. Tumour responses were shown in injected and non-injected tumours. Ten patients were radiographically evaluable for objective responses; non-evaluable patients had contraindications to contrast medium (n=2) or no post-treatment scans (n=2). According to Response Evaluation Criteria in Solid Tumors (RECIST), three patients had partial response, six had stable disease, and one had progressive disease. INTERPRETATION: Intratumoral injection of JX-594 into primary or metastatic liver tumours was generally well-tolerated. Direct hyperbilirubinaemia was the dose-limiting toxicity. Safety was acceptable in the context of JX-594 replication, GM-CSF expression, systemic dissemination, and JX-594 had anti-tumoral effects against several refractory carcinomas. Phase II trials are now underway.


Subject(s)
Liver Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Vaccinia virus , Adult , Aged , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Hyperbilirubinemia/etiology , Immunohistochemistry , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/virology , Male , Middle Aged , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/growth & development , Oncolytic Viruses/metabolism , Positron-Emission Tomography , Poxviridae Infections/etiology , Time Factors , Tomography, X-Ray Computed , Treatment Failure , Treatment Outcome , Vaccinia virus/genetics , Vaccinia virus/growth & development , Vaccinia virus/metabolism , Virus Replication
14.
N Engl J Med ; 353(2): 133-44, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-16014883

ABSTRACT

BACKGROUND: A clinical trial that compared erlotinib with a placebo for non-small-cell lung cancer demonstrated a survival benefit for erlotinib. We used tumor-biopsy samples from participants in this trial to investigate whether responsiveness to erlotinib and its impact on survival were associated with expression by the tumor of epidermal growth factor receptor (EGFR) and EGFR gene amplification and mutations. METHODS: EGFR expression was evaluated immunohistochemically in non-small-cell lung cancer specimens from 325 of 731 patients in the trial; 197 samples were analyzed for EGFR mutations; and 221 samples were analyzed for the number of EGFR genes. RESULTS: In univariate analyses, survival was longer in the erlotinib group than in the placebo group when EGFR was expressed (hazard ratio for death, 0.68; P=0.02) or there was a high number of copies of EGFR (hazard ratio, 0.44; P=0.008). In multivariate analyses, adenocarcinoma (P=0.01), never having smoked (P<0.001), and expression of EGFR (P=0.03) were associated with an objective response. In multivariate analysis, survival after treatment with erlotinib was not influenced by the status of EGFR expression, the number of EGFR copies, or EGFR mutation. CONCLUSIONS: Among patients with non-small-cell lung cancer who receive erlotinib, the presence of an EGFR mutation may increase responsiveness to the agent, but it is not indicative of a survival benefit.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Analysis of Variance , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , DNA Mutational Analysis , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Gene Expression , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinazolines/adverse effects , Survival Analysis
15.
Oncotarget ; 8(5): 8559-8573, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28051998

ABSTRACT

A defining feature of the brain cancer glioblastoma is its highly invasive nature. When glioblastoma cells are isolated from patients using serum free conditions, they accurately recapitulate this invasive behaviour in animal models. The Rac subclass of Rho GTPases has been shown to promote invasive behaviour in glioblastoma cells isolated in this manner. However the guanine nucleotide exchange factors responsible for activating Rac in this context have not been characterized previously. PREX1 is a Rac guanine nucleotide exchange factor that is synergistically activated by binding of G protein αγ subunits and the phosphoinositide 3-kinase pathway second messenger phosphatidylinositol 3,4,5 trisphosphate. This makes it of particular interest in glioblastoma, as the phosphoinositide 3-kinase pathway is aberrantly activated by mutation in almost all cases. We show that PREX1 is expressed in glioblastoma cells isolated under serum-free conditions and in patient biopsies. PREX1 promotes the motility and invasion of glioblastoma cells, promoting Rac-mediated activation of p21-associated kinases and atypical PKC, which have established roles in cell motility. Glioblastoma cell motility was inhibited by either inhibition of phosphoinositide 3-kinase or inhibition of G protein ßγ subunits. Motility was also inhibited by the generic dopamine receptor inhibitor haloperidol or a combination of the selective dopamine receptor D2 and D4 inhibitors L-741,626 and L-745,870. This establishes a role for dopamine receptor signaling via G protein ßγ subunits in glioblastoma invasion and shows that phosphoinositide 3-kinase mutations in glioblastoma require a context of basal G protein-coupled receptor activity in order to promote this invasion.


Subject(s)
Brain Neoplasms/enzymology , Cell Movement , Glioblastoma/enzymology , Guanine Nucleotide Exchange Factors/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptors, Dopamine/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Female , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Guanine Nucleotide Exchange Factors/genetics , Humans , Male , Neoplasm Invasiveness , Phosphatidylinositol Phosphates/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Dopamine/drug effects , Time Factors , Up-Regulation , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
16.
Clin Cancer Res ; 9(7): 2457-64, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12855618

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) appears to play an important role in the pathogenesis of colorectal cancer. We have performed a Phase I/II study of the EGFR tyrosine kinase inhibitor ZD1839 in metastatic colorectal cancer patients in which serial biopsies were taken pre- and posttreatment to assess biological activity. EXPERIMENTAL DESIGN: Paired biopsies were obtained from colorectal cancer patients before and after treatment. Proliferation and apoptosis were assessed using Ki67 immunohistochemistry and terminal deoxynucleotidyl transferase-mediated nick end labeling assays, respectively. Immunohistochemistry for EGFR, activated EGFR, phosphorylated Akt, phosphorylated ERK, p27(Kip1), and beta-catenin was also performed. RESULTS: Posttreatment samples showed a statistically significant reduction in the cancer cell proliferation index (mean proliferation index pretreatment 31%; posttreatment 21%; P = 0.047). The mean cancer cell apoptosis index also increased from 6 to 12% in posttreatment samples, although this difference did not achieve statistical significance. All pretreatment samples showed strong staining for EGFR. Loss of immunohistochemical staining for activated EGFR, phosphorylated Akt, and phosphorylated ERK in cancer cells was observed in some patients after treatment. p27(Kip1) was absent in the cancer cells of most pretreatment biopsies; two patients showed a marked increase in staining for nuclear p27(Kip1) after treatment with ZD1839. These two patients also showed large increases in apoptotic index. CONCLUSIONS: ZD1839 inhibits EGFR signaling and proliferation in the cancer cells of patients with metastatic colorectal cancer. ZD1839 may also induce cancer cell apoptosis in a subset of colorectal cancer patients via up-regulation of p27(Kip1).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics , Apoptosis , Cell Cycle Proteins/biosynthesis , Cell Division , Cyclin-Dependent Kinase Inhibitor p27 , Cytoskeletal Proteins/metabolism , Epidermal Growth Factor/antagonists & inhibitors , Gefitinib , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Metastasis , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Time Factors , Trans-Activators/metabolism , Tumor Suppressor Proteins/biosynthesis , beta Catenin
17.
Oncotarget ; 6(5): 3013-32, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25605249

ABSTRACT

Recent evidence points to the protein arginine methyltransferase (PRMT) family of enzymes playing critical roles in cancer. PRMT7 has been identified in several gene expression studies to be associated with increased metastasis and decreased survival in breast cancer patients. However, this has not been extensively studied. Here we report that PRMT7 expression is significantly upregulated in both primary breast tumour tissues and in breast cancer lymph node metastases. We have demonstrated that reducing PRMT7 levels in invasive breast cancer cells using RNA interference significantly decreased cell invasion in vitro and metastasis in vivo. Conversely, overexpression of PRMT7 in non-aggressive MCF7 cells enhanced their invasiveness. Furthermore, we show that PRMT7 induces the expression of matrix metalloproteinase 9 (MMP9), a well-known mediator of breast cancer metastasis. Importantly, we significantly rescued invasion of aggressive breast cancer cells depleted of PRMT7 by the exogenous expression of MMP9. Our results demonstrate that upregulation of PRMT7 in breast cancer may have a significant role in promoting cell invasion through the regulation of MMP9. This identifies PRMT7 as a novel and potentially significant biomarker and therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Cell Movement , Matrix Metalloproteinase 9/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lymph Nodes/enzymology , Lymph Nodes/pathology , Lymphatic Metastasis , Matrix Metalloproteinase 9/genetics , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Protein-Arginine N-Methyltransferases/genetics , RNA Interference , Signal Transduction , Transfection , Up-Regulation
18.
Cell Cycle ; 14(18): 2938-48, 2015.
Article in English | MEDLINE | ID: mdl-26208522

ABSTRACT

Cellular senescence is a tumor suppressor mechanism where cells enter a permanent growth arrest following cellular stress. Oncogene-induced senescence (OIS) is induced in non-malignant cells following the expression of an oncogene or inactivation of a tumor suppressor. Previously, we have shown that protein kinase C iota (PKCι) depletion induces cellular senescence in glioblastoma cells in the absence of a detectable DNA damage response. Here we demonstrate that senescent glioblastoma cells exhibit an aberrant centrosome morphology. This was observed in basal levels of senescence, in p21-induced senescence, and in PKCι depletion-induced senescence. In addition, senescent glioblastoma cells are polyploid, Ki-67 negative and arrest at the G1/S checkpoint, as determined by expression of cell cycle regulatory proteins. These markers are all consistent with cells that have undergone mitotic slippage. Failure of the spindle assembly checkpoint to function properly can lead to mitotic slippage, resulting in the premature exit of mitotic cells into the G1 phase of the cell cycle. Although in G1, these cells have the replicated DNA and centrosomal phenotype of a cell that has entered mitosis and failed to divide. Overall, we demonstrate that PKCι depletion initiates mitotic slippage-induced senescence in glioblastoma cells. To our knowledge, this is the first evidence of markers of mitotic slippage directly in senescent cells by co-staining for senescence-associated ß-galactosidase and immunofluorescence markers in the same cell population. We suggest that markers of mitotic slippage be assessed in future studies of senescence to determine the extent of mitotic slippage in the induction of cellular senescence.


Subject(s)
Cellular Senescence , Glioblastoma/pathology , Isoenzymes/physiology , Mitosis/physiology , Protein Kinase C/physiology , Biomarkers/metabolism , Cell Cycle Checkpoints , Centrosome/ultrastructure , DNA Damage , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Polyploidy , Protein Kinase C/genetics , Protein Kinase C/metabolism , Stress, Physiological
19.
Cancer Cell ; 28(2): 210-24, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26212250

ABSTRACT

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Subject(s)
Neoplasms/virology , Oncolytic Viruses/physiology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Gene Expression/drug effects , Gene Expression Profiling , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/virology , Humans , Mice, Inbred C57BL , Microscopy, Fluorescence , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/virology , Positive Regulatory Domain I-Binding Factor 1 , RNA Interference , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcriptional Activation/drug effects , Vaccinia virus/physiology
20.
Oncotarget ; 5(22): 11541-51, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25426552

ABSTRACT

lethal giant larvae (lgl) was first identified as a tumor suppressor in Drosophila, where its loss repressed the differentiation and promoted the invasion of neuroblasts, the Drosophila equivalent of the neural stem cell. Recently we have shown that a human homolog of Lgl, Lgl1 (LLGL1), is constitutively phosphorylated and inactivated in glioblastoma cells; this occurs as a downstream consequence of PTEN loss, one of the most frequent genetic events in glioblastoma. Here we have investigated the consequences of this loss of functional Lgl1 in glioblastoma in vivo. We used a doxycycline-inducible system to express a non-phosphorylatable, constitutively active version of Lgl1 (Lgl3SA) in either a glioblastoma cell line or primary glioblastoma cells isolated under neural stem cell culture conditions from patients. In both types of cells, expression of Lgl3SA, but not wild type Lgl1, inhibited cell motility in vitro. Induction of Lgl3SA in intracerebral xenografts markedly reduced the in vivo invasion of primary glioblastoma cells. Lgl3SA expression also induced the differentiation of glioblastoma cells in vitro and in vivo along the neuronal lineage. Thus the central features of Lgl function as a tumor suppressor in Drosophila are conserved in human glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Animals , Cell Line, Tumor , Cell Movement , Doxycycline/chemistry , Humans , Immunohistochemistry , Mice , Mice, SCID , Microscopy, Video , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL