Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Sci Rep ; 9(1): 16832, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31728045

ABSTRACT

Therapeutically targeting receptor tyrosine kinases has proven to be paramount to overcoming chemotherapy resistance in several cancer indications, improving patient outcomes. Insulin-Like Growth Factor Receptor 1 (IGF-1R) and Epidermal Growth Factor Receptor 3 (ErbB3) have been implicated as two such drivers of resistance, however their simultaneous role in ovarian cancer chemotherapy resistance remains poorly elucidated. The aim of this work is to determine the effects of dual IGF-1R/ErbB3 inhibition on ovarian cancer cell signaling, growth, and in vivo efficacy. Assessment of in vitro chemotherapy response across a panel of ovarian cancer cell lines revealed that increased IGF-1R cell surface expression correlates with decreased sensitivity to chemotherapy, and that growth induced by IGF-1R and ErbB3 ligands is blocked by the tetravalent bispecific antibody targeting IGF-1R and ErbB3, istiratumab. In vitro chemotherapy treatment increased ovarian cancer cell line capacity to activate prosurvival PI3K signaling in response to ligand, which could be prevented with istiratumab treatment. Furthermore, in vivo efficacy of standard of care chemotherapies using a xenograft model of ovarian cancer was potentiated with istiratumab. Our results suggest a role for IGF-1R and ErbB3 in driving chemotherapy resistance of ovarian cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Ovarian Neoplasms/drug therapy , Receptor, ErbB-3/metabolism , Receptor, IGF Type 1/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacology , Receptor, ErbB-3/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 24(12): 2873-2885, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29549161

ABSTRACT

Purpose: Insulin-like growth factor receptor 1 (IGF-1R) is critically involved in pancreatic cancer pathophysiology, promoting cancer cell survival and therapeutic resistance. Assessment of IGF-1R inhibitors in combination with standard-of-care chemotherapy, however, failed to demonstrate significant clinical benefit. The aim of this work is to unravel mechanisms of resistance to IGF-1R inhibition in pancreatic cancer and develop novel strategies to improve the activity of standard-of-care therapies.Experimental Design: Growth factor screening in pancreatic cancer cell lines was performed to identify activators of prosurvival PI3K/AKT signaling. The prevalence of activating growth factors and their receptors was assessed in pancreatic cancer patient samples. Effects of a bispecific IGF-1R and ErbB3 targeting antibody on receptor expression, signaling, cancer cell viability and apoptosis, spheroid growth, and in vivo chemotherapy activity in pancreatic cancer xenograft models were determined.Results: Growth factor screening in pancreatic cancer cells revealed insulin-like growth factor 1 (IGF-1) and heregulin (HRG) as the most potent AKT activators. Both growth factors reduced pancreatic cancer cell sensitivity to gemcitabine or paclitaxel in spheroid growth assays. Istiratumab (MM-141), a novel bispecific antibody that blocks IGF-1R and ErbB3, restored the activity of paclitaxel and gemcitabine in the presence of IGF-1 and HRG in vitro Dual IGF-1R/ErbB3 blocking enhanced chemosensitivity through inhibition of AKT phosphorylation and promotion of IGF-1R and ErbB3 degradation. Addition of istiratumab to gemcitabine and nab-paclitaxel improved chemotherapy activity in vivoConclusions: Our findings suggest a critical role for the HRG/ErbB3 axis and support the clinical exploration of dual IGF-1R/ErbB3 blocking in pancreatic cancer. Clin Cancer Res; 24(12); 2873-85. ©2018 AACR.


Subject(s)
Albumins/pharmacology , Deoxycytidine/analogs & derivatives , Paclitaxel/pharmacology , Pancreatic Neoplasms/metabolism , Receptor, ErbB-3/antagonists & inhibitors , Receptors, Somatomedin/antagonists & inhibitors , Animals , Caspases/metabolism , Cell Line, Tumor , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Receptor, ErbB-3/metabolism , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
3.
Mol Cancer Ther ; 14(11): 2642-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26310543

ABSTRACT

Heregulin-driven ERBB3 signaling has been implicated as a mechanism of resistance to cytotoxic and antiendocrine therapies in preclinical breast cancer models. In this study, we evaluated the effects of seribantumab (MM-121), a heregulin-blocking anti-ERBB3 monoclonal antibody, alone and in combination with the aromatase inhibitor letrozole, on cell signaling and tumor growth in a preclinical model of postmenopausal estrogen receptor-positive (ER(+)) breast cancer. In vitro, heregulin treatment induced estrogen receptor phosphorylation in MCF-7Ca cells, and long-term letrozole-treated (LTLT-Ca) cells had increased expression and activation levels of EGFR, HER2, and ERBB3. Treatment with seribantumab, but not letrozole, inhibited basal and heregulin-mediated ERBB receptor phosphorylation and downstream effector activation in letrozole-sensitive (MCF-7Ca) and -refractory (LTLT-Ca) cells. Notably, in MCF-7Ca-derived xenograft tumors, cotreatment with seribantumab and letrozole had increased antitumor activity compared with letrozole alone, which was accompanied by downregulated PI3K/MTOR signaling both prior to and after the development of resistance to letrozole. Moreover, the addition of an MTOR inhibitor to this treatment regimen did not improve antitumor activity and was not well tolerated. Our results demonstrate that heregulin-driven ERBB3 signaling mediates resistance to letrozole in a preclinical model of ER(+) breast cancer, suggesting that heregulin-expressing ER(+) breast cancer patients may benefit from the addition of seribantumab to antiendocrine therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Nitriles/pharmacology , Receptor, ErbB-3/antagonists & inhibitors , Triazoles/pharmacology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunoblotting , Letrozole , Mice, Inbred BALB C , Mice, Nude , Neuregulin-1/pharmacology , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
4.
PLoS One ; 6(11): e28077, 2011.
Article in English | MEDLINE | ID: mdl-22140510

ABSTRACT

BACKGROUND: Recent evidence links aberrant activation of Hedgehog (Hh) signaling with the pathogenesis of several cancers including medulloblastoma, basal cell, small cell lung, pancreatic, prostate and ovarian. This investigation was designed to determine if inhibition of this pathway could inhibit serous ovarian cancer growth. METHODOLOGY: We utilized an in vivo pre-clinical model of serous ovarian cancer to characterize the anti-tumor activity of Hh pathway inhibitors cyclopamine and a clinically applicable derivative, IPI-926. Primary human serous ovarian tumor tissue was used to generate tumor xenografts in mice that were subsequently treated with cyclopamine or IPI-926. PRINCIPAL FINDINGS: Both compounds demonstrated significant anti-tumor activity as single agents. When IPI-926 was used in combination with paclitaxel and carboplatinum (T/C), no synergistic effect was observed, though sustained treatment with IPI-926 after cessation of T/C continued to suppress tumor growth. Hh pathway activity was analyzed by RT-PCR to assess changes in Gli1 transcript levels. A single dose of IPI-926 inhibited mouse stromal Gli1 transcript levels at 24 hours with unchanged human intra-tumor Gli1 levels. Chronic IPI-926 therapy for 21 days, however, inhibited Hh signaling in both mouse stromal and human tumor cells. Expression data from the micro-dissected stroma in human serous ovarian tumors confirmed the presence of Gli1 transcript and a significant association between elevated Gli1 transcript levels and worsened survival. CONCLUSIONS/SIGNIFICANCE: IPI-926 treatment inhibits serous tumor growth suggesting the Hh signaling pathway contributes to the pathogenesis of ovarian cancer and may hold promise as a novel therapeutic target, especially in the maintenance setting.


Subject(s)
Hedgehog Proteins/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays , Animals , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/genetics , Humans , Maintenance Chemotherapy , Mice , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Veratrum Alkaloids/pharmacology , Veratrum Alkaloids/therapeutic use , Zinc Finger Protein GLI1
5.
PLoS One ; 6(6): e20715, 2011.
Article in English | MEDLINE | ID: mdl-21695255

ABSTRACT

Despite the fact that epithelial ovarian cancers are the leading cause of death from gynecological cancer, very little is known about the pathophysiology of the disease. Mutations in the WNT and PI3K pathways are frequently observed in the human ovarian endometrioid adenocarcinomas (OEAs). However, the role of WNT/ß-catenin and PTEN/AKT signaling in the etiology and/or progression of this disease is currently unclear. In this report we show that mice with a gain-of-function mutation in ß-catenin that leads to dysregulated nuclear accumulation of ß-catenin expression in the ovarian surface epithelium (OSE) cells develop indolent, undifferentiated tumors with both mesenchymal and epithelial characteristics. Combining dysregulated ß-catenin with homozygous deletion of PTEN in the OSE resulted in development of significantly more aggressive tumors, which was correlated with inhibition of p53 expression and cellular senescence. Induced expression of both mTOR kinase, a master regulator of proliferation, and phosphorylation of its downstream target, S6Kinase was also observed in both the indolent and aggressive mouse tumors, as well as in human OEA with nuclear ß-catenin accumulation. Ectopic allotransplants of the mouse ovarian tumor cells with a gain-of-function mutation in ß-catenin and PTEN deletion developed into tumors with OEA histology, the growth of which were significantly inhibited by oral rapamycin treatment. These studies demonstrate that rapamycin might be an effective therapeutic for human ovarian endometrioid patients with dysregulated Wnt/ß-catenin and Pten/PI3K signaling.


Subject(s)
Carcinoma, Endometrioid/enzymology , Ovarian Neoplasms/enzymology , PTEN Phosphohydrolase/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Carcinoma, Endometrioid/pathology , Cell Death/drug effects , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Female , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Sirolimus/pharmacology , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism
6.
Front Biosci (Landmark Ed) ; 16(1): 368-92, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196176

ABSTRACT

Ovarian cancer represents the most lethal gynecologic malignancy, primarily due to a lack of early detection, which results in most patients being diagnosed at an advanced stage of disease. Though the ovarian surface epithelium is thought to provide the primary site of tumorigenesis, the exact etiology of the various tumor types associated with this disease remain undefined. Recent evidence suggests that ovarian tumors, like other solid tumors, contain distinct populations of cells that are responsible for tumor initiation, maintenance and growth. These specialized cells, termed cancer stem cells, display some of the hallmarks of normal stem cells and are thought to evade current chemotherapeutic strategies, resulting in an increased risk of recurrence. Here we review evidence for the existence of cancer stem cells in ovarian malignancies and their contribution to the pathology of this disease, critically evaluate the methods used for ovarian cancer stem cell definition and isolation, and discuss their clinical relevance.


Subject(s)
Neoplastic Stem Cells/pathology , Ovarian Neoplasms/etiology , AC133 Antigen , Animals , Antigens, CD/physiology , Cell Transformation, Neoplastic/pathology , Female , Glycoproteins/physiology , Humans , Hyaluronan Receptors/physiology , Mice , Myeloid Differentiation Factor 88/physiology , Neoplasm Recurrence, Local/physiopathology , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/pathology , Ovary/pathology , Peptides/physiology , Proto-Oncogene Proteins c-kit/physiology , Signal Transduction/drug effects , Stem Cells/pathology , Transplantation, Heterologous
7.
Reprod Biol Endocrinol ; 8: 147, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21122138

ABSTRACT

BACKGROUND: Recent data provide significant evidence to support the hypothesis that there are sub-populations of cells within solid tumors that have an increased tumor initiating potential relative to the total tumor population. CD133, a cell surface marker expressed on primitive cells of neural, hematopoietic, endothelial and epithelial lineages has been identified as a marker for tumor initiating cells in solid tumors of the brain, colon, pancreas, ovary and endometrium. Our objectives were to assess the relative level of CD133 expressing cells in primary human endometrial tumors, confirm their tumorigenic potential, and determine whether CD133 expression was epigenetically modified. METHODS: We assessed CD133 expression in primary human endometrial tumors by flow cytometry and analyzed the relative tumorigenicity of CD133+ and CD133- cells in an in vivo NOD/SCID mouse model. We assessed potential changes in CD133 expression over the course of serial transplantation by immunofluorescence and flow cytometry. We further examined CD133 promoter methylation and expression in normal endometrium and malignant tumors. RESULTS: As determined by flow cytometric analysis, the percentage of CD133+ cells in primary human endometrial cancer samples ranged from 5.7% to 27.4%. In addition, we confirmed the tumor initiating potential of CD133+ and CD133- cell fractions in NOD/SCID mice. Interestingly, the percentage of CD133+ cells in human endometrial tumor xenografts, as evidenced by immunofluorescence, increased with serial transplantation although this trend was not consistently detected by flow cytometry. We also determined that the relative levels of CD133 increased in endometrial cancer cell lines following treatment with 5-aza-2'-deoxycytidine suggesting a role for methylation in the regulation of CD133. To support this finding, we demonstrated that regions of the CD133 promoter were hypomethylated in malignant endometrial tissue relative to benign control endometrial tissue. Lastly, we determined that methylation of the CD133 promoter decreases over serial transplantation of an endometrial tumor xenograft. CONCLUSIONS: These findings support the hypotheses that CD133 expression in endometrial cancer may be epigenetically regulated and that cell fractions enriched for CD133+ cells may well contribute to endometrial cancer tumorigenicity, pathology and recurrence.


Subject(s)
Antigens, CD/genetics , Cell Transformation, Neoplastic/pathology , Endometrial Neoplasms/pathology , Epigenomics , Glycoproteins/genetics , Neoplastic Stem Cells/pathology , Peptides/genetics , AC133 Antigen , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Decitabine , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/immunology
8.
Aviat Space Environ Med ; 81(6): 537-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20540443

ABSTRACT

INTRODUCTION: CO2 accumulation may limit crew survival in a disabled submarine. Reversible sedation using diazepam and flumazenil was proposed to reduce CO2 production. METHODS: Two groups of three resting subjects were studied during a 48-h placebo phase with diazepam and flumazenil placebos, followed by a 48-h drug phase with oral diazepam to induce sedation and intranasal flumazenil to reverse it. CO2 exchange was measured every 2.5 h; twice a day, cognitive testing and meals were preceded by placebo or flumazenil. Return to sedated state was produced with either placebo or diazepam. In the drug phase, initial diazepam doses (10 to 40 mg) were followed by maintenance doses to achieve sedation corresponding to Alertness Scores of 3 or 4. RESULTS: In the drug phase, subjects received a total of 360-495 mg of diazepam (with doses of 5-40 mg), average alertness score was 3.75, and mean Vco2 was 14% less than in the placebo phase (0.212 vs. 0.248 L x min(-)). Subjects were 21-36% less active when sedated with diazepam. The mean flumazenil dose to restore full alertness was 0.36 mg, with subjects being conversant and oriented within 5 min, performing cognitive tasks at 86-97% of their baseline. Subjects could follow instructions and ambulate independently, though unsteadily 6 h after final flumazenil dose; at 72 h they exhibited normal cognitive and physical functions. DISCUSSION: Reversible sedation to lower crew metabolism in a disabled submarine may be effective, safe, and practical.


Subject(s)
Carbon Dioxide/metabolism , Diazepam/therapeutic use , Flumazenil/therapeutic use , Oxygen Consumption , Submarine Medicine , Acceleration , Adult , Analysis of Variance , Antidotes/administration & dosage , Antidotes/therapeutic use , Carbon Dioxide/physiology , Cognition/drug effects , Diazepam/administration & dosage , Flumazenil/administration & dosage , GABA Modulators/administration & dosage , GABA Modulators/therapeutic use , Health Status Indicators , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/therapeutic use , Male , Respiration/drug effects , Time Factors , Young Adult
9.
Stem Cells ; 27(12): 2875-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19816957

ABSTRACT

Evidence is accumulating that solid tumors contain a rare phenotypically distinct population of cells, termed cancer stem cells (CSC), which give rise to and maintain the bulk of the tumor. These CSC are thought to be resistant to current chemotherapeutic strategies due to their intrinsic stem-like properties and thus may provide the principal driving force behind recurrent tumor growth. Given the high frequency of recurrent metastasis associated with human ovarian cancer, we sought to determine whether primary human ovarian tumors contain populations of cells with enhanced tumor-initiating capacity, a characteristic of CSC. Using an in vivo serial transplantation model, we show that primary uncultured human ovarian tumors can be reliably propagated in NOD/SCID mice, generating heterogeneous tumors that maintain the histological integrity of the parental tumor. The observed frequency of tumor engraftment suggests only certain subpopulations of ovarian tumor cells have the capacity to recapitulate tumor growth. Further profiling of human ovarian tumors for expression of candidate CSC surface markers indicated consistent expression of CD133. To determine whether CD133 expression could define a tumor-initiating cell population in primary human ovarian tumors, fluorescence-activated cell sorting (FACS) methods were employed. Injection of sorted CD133(+) and CD133(-) cell populations into NOD/SCID mice established that tumor-derived CD133(+) cells have an increased tumorigenic capacity and are capable of recapitulating the original heterogeneous tumor. Our data indicate that CD133 expression defines a NOD/SCID tumor initiating subpopulation of cells in human ovarian cancer that may be an important target for new chemotherapeutic strategies aimed at eliminating ovarian cancer.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peptides/metabolism , AC133 Antigen , Animals , Biomarkers, Tumor/metabolism , Cell Count , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Xenograft Model Antitumor Assays
10.
Aviat Space Environ Med ; 75(5): 397-404, 2004 May.
Article in English | MEDLINE | ID: mdl-15152891

ABSTRACT

INTRODUCTION: Psychophysical measures of wet-suit hood sound attenuation are needed to provide the diving community with guidance on protection from underwater sound. METHODS: Underwater hearing thresholds were obtained from 15 male and 5 female recreational divers with and without a 3-mm thick wet-suit hood. Dives were conducted at a depth of 1 m in a large quiet anechoic pool. Thresholds were determined using a two-interval forced-choice procedure with a 0.71 probability of positive response at convergence. A 1-s pure tone was presented with a 20-ms rise and fall time at 100, 200, 250, 300, 400, and 500 Hz. RESULTS: Without a wet-suit hood, mean thresholds decreased from 99 dB re 1 microPa at 100 Hz to 85 dB at 500 Hz. Thresholds were statistically similar at 100 to 300 Hz with and without the wet-suit hood, but were significantly increased at 400 and 500 Hz with the hood (p < 0.001). CONCLUSIONS: In conclusion, at shallow depths, a 3-mm neoprene wet-suit hood attenuates underwater sound by approximately 10 dB for frequencies between 400 Hz and 500 Hz. At frequencies below 400 Hz, a 3-mm neoprene wet-suit hood offers no sound protection.


Subject(s)
Auditory Threshold , Diving , Head Protective Devices , Adult , Female , Hearing Tests , Humans , Male , Military Science , Neoprene
SELECTION OF CITATIONS
SEARCH DETAIL
...