21 results on '"Inderjit Mehmi"'
Search Results
2. Longitudinal SARS-CoV-2 mRNA Vaccine-Induced Humoral Immune Responses in Patients with Cancer
- Author
-
Kimia Sobhani, Jennifer E. Van Eyk, James L. Stewart, Warren G. Tourtellotte, Justin Darrah, Joslyn Foley, Wendy Cozen, Karen L. Reckamp, Susan Cheng, Alain C. Mita, So Yung Choi, Carissa A. Huynh, Noah Merin, Sandy Joung, Tucker Lemos, Robert Vescio, Ronald Paquette, Dermot P.B. McGovern, Edwin C. Frias, John Prostko, Omid Hamid, Joseph E. Ebinger, Emebet Mengesha, Reva Basho, Greg Botwin, Jun Gong, Nathalie Nguyen, Jonathan Braun, Inderjit Mehmi, Jane C. Figueiredo, Akil Merchant, Gil Y. Melmed, and Laurel J Finster
- Subjects
Adult ,Male ,Cancer Research ,Time Factors ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Antibodies, Viral ,Article ,Immune system ,Neoplasms ,Surveys and Questionnaires ,Humans ,Medicine ,Longitudinal Studies ,Prospective Studies ,Prospective cohort study ,BNT162 Vaccine ,Aged ,Messenger RNA ,biology ,Immunization Programs ,SARS-CoV-2 ,business.industry ,Vaccination ,COVID-19 ,Cancer ,Middle Aged ,medicine.disease ,Immunity, Humoral ,Oncology ,Immunoglobulin G ,Immunology ,biology.protein ,Female ,Antibody ,business ,2019-nCoV Vaccine mRNA-1273 - Abstract
Longitudinal studies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine-induced immune responses in patients with cancer are needed to optimize clinical care. In a prospective cohort study of 366 (291 vaccinated) patients, we measured antibody levels [anti-spike (IgG-(S-RBD) and anti-nucleocapsid immunoglobulin] at three time points. Antibody level trajectories and frequency of breakthrough infections were evaluated by tumor type and timing of treatment relative to vaccination. IgG-(S-RBD) at peak response (median = 42 days after dose 2) was higher (P = 0.002) and remained higher after 4 to 6 months (P = 0.003) in patients receiving mRNA-1273 compared with BNT162b2. Patients with solid tumors attained higher peak levels (P = 0.001) and sustained levels after 4 to 6 months (P < 0.001) compared with those with hematologic malignancies. B-cell targeted treatment reduced peak (P = 0.001) and sustained antibody responses (P = 0.003). Solid tumor patients receiving immune checkpoint inhibitors before vaccination had lower sustained antibody levels than those who received treatment after vaccination (P = 0.043). Two (0.69%) vaccinated and one (1.9%) unvaccinated patient had severe COVID-19 illness during follow-up. Our study shows variation in sustained antibody responses across cancer populations receiving various therapeutic modalities, with important implications for vaccine booster timing and patient selection. Significance: Long-term studies of immunogenicity of SARS-CoV-2 vaccines in patients with cancer are needed to inform evidence-based guidelines for booster vaccinations and to tailor sequence and timing of vaccinations to elicit improved humoral responses.
- Published
- 2021
- Full Text
- View/download PDF
3. Efficacy of anti-PD-1 and ipilimumab alone or in combination in acral melanoma
- Author
-
Prachi Bhave, Tasnia Ahmed, Serigne N Lo, Alexander Shoushtari, Anne Zaremba, Judith M Versluis, Joanna Mangana, Michael Weichenthal, Lu Si, Thierry Lesimple, Caroline Robert, Claudia Trojanello, Alexandre Wicky, Richard Heywood, Lena Tran, Kathleen Batty, Florentia Dimitriou, Anna Stansfeld, Clara Allayous, Julia K Schwarze, Meghan J Mooradian, Oliver Klein, Inderjit Mehmi, Rachel Roberts-Thomson, Andrea Maurichi, Hui-Ling Yeoh, Adnan Khattak, Lisa Zimmer, Christian U Blank, Egle Ramelyte, Katharina C Kähler, Severine Roy, Paolo A Ascierto, Olivier Michielin, Paul C Lorigan, Douglas B Johnson, Ruth Plummer, Celeste Lebbe, Bart Neyns, Ryan Sullivan, Omid Hamid, Mario Santinami, Grant A McArthur, Andrew M Haydon, Georgina V Long, Alexander M Menzies, Matteo S Carlino, Clinical sciences, Laboratory of Molecular and Medical Oncology, Internal Medicine, Faculty of Medicine and Pharmacy, and Medical Oncology
- Subjects
Pharmacology ,Cancer Research ,Skin Neoplasms ,Manchester Cancer Research Centre ,ResearchInstitutes_Networks_Beacons/mcrc ,Immunology ,Programmed Cell Death 1 Receptor ,Medizin ,Ipilimumab ,oncology ,Molecular Medicine ,Immunology and Allergy ,Humans ,CTLA-4 Antigen ,Immunotherapy ,immunotherapy ,Prospective Studies ,Melanoma ,Retrospective Studies - Abstract
BackgroundAcral melanoma is a rare melanoma subtype with poor prognosis. Importantly, these patients were not identified as a specific subgroup in the landmark melanoma trials involving ipilimumab and the anti-programmed cell death protein-1 (PD-1) agents nivolumab and pembrolizumab. There is therefore an absence of prospective clinical trial evidence regarding the efficacy of checkpoint inhibitors (CPIs) in this population. Acral melanoma has lower tumor mutation burden (TMB) than other cutaneous sites, and primary site is associated with differences in TMB. However the impact of this on the effectiveness of immune CPIs is unknown. We examined the efficacy of CPIs in acral melanoma, including by primary site.MethodsPatients with unresectable stage III/IV acral melanoma treated with CPI (anti-PD-1 and/or ipilimumab) were studied. Multivariable logistic and Cox regression analyses were conducted. Primary outcome was objective response rate (ORR); secondary outcomes were progression-free survival (PFS) and overall survival (OS).ResultsIn total, 325 patients were included: 234 (72%) plantar, 69 (21%) subungual and 22 (7%) palmar primary sites. First CPI included: 184 (57%) anti-PD-1, 59 (18%) anti-PD-1/ipilimumab combination and 82 (25%) ipilimumab. ORR was significantly higher with initial anti-PD-1/ipilimumab compared with anti-PD-1 (43% vs 26%, HR 2.14, p=0.0004) and significantly lower with ipilimumab (15% vs 26%, HR 0.49, p=0.0016). Landmark PFS at 1 year was highest for anti-PD-1/ipilimumab at 34% (95% CI 24% to 49%), compared with 26% (95% CI 20% to 33%) with anti-PD-1 and 10% (95% CI 5% to 19%) with ipilimumab. Despite a trend for increased PFS, anti-PD-1/ipilimumab combination did not significantly improve PFS (HR 0.85, p=0.35) or OS over anti-PD-1 (HR 1.30, p=0.16), potentially due to subsequent therapies and high rates of acquired resistance. No outcome differences were found between primary sites.ConclusionWhile the ORR to anti-PD-1/ipilimumab was significantly higher than anti-PD-1 and PFS numerically higher, in this retrospective cohort this benefit did not translate to improved OS. Future trials should specifically include patients with acral melanoma, to help determine the optimal management of this important melanoma subtype.
- Published
- 2022
- Full Text
- View/download PDF
4. Eflapegrastim, a Long-Acting Granulocyte-Colony Stimulating Factor for the Management of Chemotherapy-Induced Neutropenia: Results of a Phase III Trial
- Author
-
Inderjit Mehmi, Gajanan Bhat, Julio Antonio Peguero, Lee S. Schwartzberg, Zane Yang, Patrick Wayne Cobb, Shanta Chawla, Jayaram S. Bharadwaj, Osama Hlalah, Steven J. Hasal, Richy Agajanian, and Alvaro Restrepo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neutropenia ,Filgrastim ,Eflapegrastim ,Cyclophosphamide ,Neutrophils ,medicine.medical_treatment ,Antineoplastic Agents ,Breast Neoplasms ,Chemotherapy‐induced neutropenia ,Polyethylene Glycols ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Granulocyte Colony-Stimulating Factor ,medicine ,Clinical endpoint ,Humans ,Chemotherapy ,business.industry ,medicine.disease ,Chemotherapy regimen ,Recombinant Proteins ,Pegfilgrastim ,Granulocyte colony-stimulating factor ,030104 developmental biology ,Docetaxel ,Symptom Management and Supportive Care ,030220 oncology & carcinogenesis ,Female ,business ,medicine.drug - Abstract
Background Eflapegrastim, a novel, long‐acting recombinant human granulocyte‐colony stimulating factor (rhG‐CSF), consists of a rhG‐CSF analog conjugated to a human IgG4 Fc fragment via a short polyethylene glycol linker. Preclinical and phase I and II pharmacodynamic and pharmacokinetic data showed increased potency for neutrophil counts for eflapegrastim versus pegfilgrastim. This open‐label phase III trial compared the efficacy and safety of eflapegrastim with pegfilgrastim for reducing the risk of chemotherapy‐induced neutropenia. Materials and Methods Patients with early‐stage breast cancer were randomized 1:1 to fixed‐dose eflapegrastim 13.2 mg (3.6 mg G‐CSF) or standard pegfilgrastim (6 mg G‐CSF) following standard docetaxel plus cyclophosphamide chemotherapy for 4 cycles. The primary objective was to demonstrate the noninferiority of eflapegrastim compared with pegfilgrastim in mean duration of severe neutropenia (DSN; grade 4) in cycle 1. Results Eligible patients were randomized 1:1 to study arms (eflapegrastim, n = 196; pegfilgrastim, n = 210). The incidence of cycle 1 severe neutropenia was 16% (n = 31) for eflapegrastim versus 24% (n = 51) for pegfilgrastim, reducing the relative risk by 35% (p = .034). The difference in mean cycle 1 DSN (−0.148 day) met the primary endpoint of noninferiority (p < .0001) and also showed statistical superiority for eflapegrastim (p = .013). Noninferiority was maintained for the duration of treatment (all cycles, p < .0001), and secondary efficacy endpoints and safety results were also comparable for study arms. Conclusion These results demonstrate noninferiority and comparable safety for eflapegrastim at a lower G‐CSF dose versus pegfilgrastim. The potential for increased potency of eflapegrastim to deliver improved clinical benefit warrants further clinical study in patients at higher risk for CIN. Implications for Practice Chemotherapy‐induced neutropenia (CIN) remains a significant clinical dilemma for oncology patients who are striving to complete their prescribed chemotherapy regimen. In a randomized, phase III trial comparing eflapegrastim to pegfilgrastim in the prevention of CIN, the efficacy of eflapegrastim was noninferior to pegfilgrastim and had comparable safety. Nevertheless, the risk of CIN remains a great concern for patients undergoing chemotherapy, as the condition frequently results in chemotherapy delays, dose reductions, and treatment discontinuations., Myelosuppression, particularly neutropenia, has presented a major challenge in cancer treatment since the introduction of cytotoxic chemotherapy. This article reports the results of a phase III trial that compared the efficacy and safety of eflapegrastim with pegfilgrastim for reducing the risk of chemotherapy‐induced neutropenia.
- Published
- 2020
- Full Text
- View/download PDF
5. Fatty Acid Synthase Is a Key Enabler for Endocrine Resistance in Heregulin-Overexpressing Luminal B-Like Breast Cancer
- Author
-
Sara Verdura, Ella Atlas, Ingrid Espinoza, Travis Vander Steen, Javier A. Menendez, Inderjit Mehmi, Luciano Vellon, Adriana Papadimitropoulou, Elisabet Cuyàs, and Ruth Lupu
- Subjects
Receptor, ErbB-2 ,Medicaments antineoplàstics ,purl.org/becyt/ford/1 [https] ,lcsh:Chemistry ,Mice ,Phosphatidylinositol 3-Kinases ,Transactivation ,Antineoplastic agents ,skin and connective tissue diseases ,lcsh:QH301-705.5 ,Spectroscopy ,biology ,tamoxifen ,fulvestrant ,Chemistry ,General Medicine ,purl.org/becyt/ford/3.1 [https] ,Computer Science Applications ,Fatty Acid Synthase, Type I ,Fatty acid synthase ,MCF-7 Cells ,Neuregulin ,Female ,purl.org/becyt/ford/3 [https] ,Signal transduction ,Mama -- Càncer -- Aspectes endocrins ,medicine.drug ,Antineoplastic Agents, Hormonal ,MAP Kinase Signaling System ,Mice, Nude ,Breast Neoplasms ,Mama -- Càncer -- Tractament ,Article ,Catalysis ,Inorganic Chemistry ,endocrine resistance ,Tamoxifèn ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,TAMOXIFEN ,Autocrine signalling ,LUMINAL ,purl.org/becyt/ford/1.6 [https] ,Molecular Biology ,Protein kinase B ,luminal ,Breast -- Cancer -- Treatment ,Fulvestrant ,Organic Chemistry ,Proteins ,ENDOCRINE RESISTANCE ,lcsh:Biology (General) ,lcsh:QD1-999 ,Drug Resistance, Neoplasm ,FULVESTRANT ,Breast -- Cancer -- Endocrine aspects ,biology.protein ,Cancer research ,Proto-Oncogene Proteins c-akt ,Tamoxifen - Abstract
HER2 transactivation by the HER3 ligand heregulin (HRG) promotes an endocrine-resistant phenotype in the estrogen receptor-positive (ER+) luminal-B subtype of breast cancer. The underlying biological mechanisms that link them are, however, incompletely understood. Here, we evaluated the putative role of the lipogenic enzyme fatty acid synthase (FASN) as a major cause of HRG-driven endocrine resistance in ER+/HER2-negative breast cancer cells. MCF-7 cells engineered to stably overexpress HRG (MCF-7/HRG), an in vitro model of tamoxifen/fulvestrant-resistant luminal B-like breast cancer, showed a pronounced up-regulation of FASN gene/FASN protein expression. Autocrine HRG up-regulated FASN expression via HER2 transactivation and downstream activation of PI-3K/AKT and MAPK-ERK1/2 signaling pathways. The HRG-driven FASN-overexpressing phenotype was fully prevented in MCF-7 cells expressing a structural deletion mutant of HRG that is sequestered in a cellular compartment and lacks the ability to promote endocrine-resistance in an autocrine manner. Pharmacological inhibition of FASN activity blocked the estradiol-independent and tamoxifen/fulvestrant-refractory ability of MCF-7/HRG cells to anchorage-independently grow in soft-agar. In vivo treatment with a FASN inhibitor restored the anti-tumor activity of tamoxifen and fulvestrant against fast-growing, hormone-resistant MCF-7/HRG xenograft tumors in mice. Overall, these findings implicate FASN as a key enabler for endocrine resistance in HRG+/HER2-breast cancer and highlight the therapeutic potential of FASN inhibitors for the treatment of endocrine therapy-resistant luminal-B breast cancer. Fil: Menendez, Javier A.. Institut D'investigació Biomèdica de Girona Dr. Josep Trueta; España. Institut Català d'Oncologia ; España Fil: Mehmi, Inderjit. The Angeles Clinic And Research Institute; Estados Unidos Fil: Papadimitropoulou, Adriana. Academy of Athens; Grecia Fil: Steen, Travis Vander. Mayo Clinic; Estados Unidos Fil: Cuyàs, Elisabet. Institut D'investigació Biomèdica de Girona Dr. Josep Trueta; España. Institut Català d'Oncologia ; España Fil: Verdura, Sara. Institut Català d'Oncologia ; España. Institut D'investigació Biomèdica de Girona Dr. Josep Trueta; España Fil: Espinoza, Ingrid. University of Mississippi; Estados Unidos Fil: Vellón, Luciano. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Atlas, Ella. University of Ottawa; Canadá Fil: Lupu, Ruth. Mayo Clinic Cancer Center; Estados Unidos. Mayo Clinic; Estados Unidos. Academy of Athens; Grecia
- Published
- 2020
6. Metastatic Sarcomatoid Carcinoma of the Small Intestine: a Case Report of Rare Tumor with Literature Review
- Author
-
Mohamed Alsharedi, Yousef Khelfa, Inderjit Mehmi, Ali Raufi, Toni Pacioles, Amanda K. Arrington, and Yehuda Lebowicz
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Pathology ,Intestinal Neoplasm ,medicine.medical_treatment ,MEDLINE ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Intestinal Neoplasms ,Intestine, Small ,medicine ,Humans ,Neoplasm Metastasis ,Sarcomatoid carcinoma ,business.industry ,Gastroenterology ,Middle Aged ,medicine.disease ,Small intestine ,Radiation therapy ,Rare tumor ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,business - Published
- 2015
- Full Text
- View/download PDF
7. Endogenous aryl hydrocarbon receptor promotes basal and inducible expression of tumor necrosis factor target genes in MCF-7 cancer cells
- Author
-
Justin K. Tomblin, James Denvir, Gary Z. Morris, Nalini Santanam, Donald A. Primerano, Jackie Fletcher, Jun Fan, Inderjit Mehmi, Travis B. Salisbury, Estil Hurn, and Goran Boskovic
- Subjects
Small interfering RNA ,Polychlorinated Dibenzodioxins ,Biology ,Biochemistry ,Article ,Gene expression ,Basic Helix-Loop-Helix Transcription Factors ,Humans ,RNA, Small Interfering ,Transcription factor ,Pharmacology ,Regulation of gene expression ,Gene knockdown ,Superoxide Dismutase ,Tumor Necrosis Factor-alpha ,respiratory system ,Aryl hydrocarbon receptor ,Molecular biology ,Enzymes ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,Gene Knockdown Techniques ,Inactivation, Metabolic ,Cancer cell ,MCF-7 Cells ,Cancer research ,biology.protein ,Tumor necrosis factor alpha - Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that upon activation by the toxicant 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) stimulates gene expression and toxicity. AHR is also important for normal mouse physiology and may play a role in cancer progression in the absence of environmental toxicants. The objective of this report was to identify AHR-dependent genes (ADGs) whose expression is regulated by AHR in the absence of toxicants. RNA-Seq analysis revealed that AHR regulated the expression of over 600 genes at an FDR
- Published
- 2014
- Full Text
- View/download PDF
8. Inhibition of tumor-associated fatty acid synthase activity antagonizes estradiol- and tamoxifen-induced agonist transactivation of estrogen receptor (ER) in human endometrial adenocarcinoma cells
- Author
-
Ruth Lupu, Vishal Verma, Javier A. Menendez, Inderjit Mehmi, Bharvi P. Oza, and Ella Atlas
- Subjects
Transcriptional Activation ,Agonist ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Estrogen receptor ,Adenocarcinoma ,Transactivation ,chemistry.chemical_compound ,Cell Line, Tumor ,Internal medicine ,Genetics ,medicine ,Humans ,Molecular Biology ,Estradiol ,biology ,Endometrial cancer ,medicine.disease ,Endometrial Neoplasms ,Cerulenin ,Tamoxifen ,Fatty acid synthase ,Endocrinology ,Receptors, Estrogen ,chemistry ,biology.protein ,Female ,Fatty Acid Synthases ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Overexpression of the lipogenic enzyme fatty acid synthase (FAS) is a common molecular feature in subsets of sex-steroid-related tumors including endometrium and breast carcinomas that are associated with poor prognosis. Pharmacological inhibition of tumor-associated FAS hyperactivity is under investigation as a chemotherapeutic target. We examined the effects of the mycotoxin cerulenin (a covalent FAS inactivator), and the novel small compound C75 (a slow-binding FAS inhibitor) on estradiol (E2)- and tamoxifen (TAM)-stimulated ER-driven molecular responses in Ishikawa cells, an in vitro model of well-differentiated human endometrial carcinoma. We evaluated the effects of FAS inhibition on E2- and TAM-induced estrogen receptor (ER) transcriptional activity by using transient cotransfection assays with an estrogen-response element reporter construct (ERE-Luciferase). Antiestrogenic effects of cerulenin and C75 were observed by dose-dependent inhibition of E2-stimulated ERE-dependent transcription, whereas FAS inhibitors did not significantly increase the levels of ERE transcriptional activity in the absence of E2. Moreover, pharmacological blockade of FAS activity completely abolished TAM-stimulated ERE activity. To address the reliability of transient transfection assays, the effects of FAS inhibitors on E2-inducible gene products were evaluated. FAS blockade induced a dose-dependent decrease in E2-inducible alkaline phosphatase activity. E2-stimulated accumulation of progesterone receptor (PR) and HER-2/neu oncogene was abolished in the presence of FAS blockers. FAS inhibition also resulted in a marked downregulation of E2-stimulated ERalpha expression, and noticeably impaired E2-induced ERalpha nuclear accumulation. A dose-dependent decrease in cell proliferation and cell viability was observed after FAS blockade. A Cell Death ELISA, detecting DNA fragmentation, demonstrated that FAS inhibitors stimulated apoptosis of Ishikawa cells. The analysis of critical E2- and TAM-related cell cycle proteins revealed an increase of both the expression and the nuclear accumulation of cyclin-dependent kinase inhibitors p21WAF1/CIP1 and p27Kip1 following FAS inhibition. To rule out non-FAS cerulenin- and C75-related effects, we finally monitored ER signaling after silencing of FAS gene expression using the highly sequence-specific mechanism of RNA interference (RNAi). The concentrations of E2 and TAM inducing half-maximal ERE activity (EC50) dramatically increased (100 times) in FAS RNAi-transfected Ishikawa cells. Moreover, depletion of FAS by RNAi also caused loss of ERalpha expression, downregulation of PR, and accumulation of p21WAF1/CIP1 and p27Kip1 in E2-stimulated Ishikawa cells. If chemically stable FAS inhibitors or cell-selective vector systems able to deliver RNAi targeting FAS gene demonstrate systemic anticancer effects in vivo, our results render FAS as a novel target for the prevention and treatment of endometrial carcinoma.
- Published
- 2004
- Full Text
- View/download PDF
9. The angiogenic factor CYR61 in breast cancer: molecular pathology and therapeutic perspectives
- Author
-
Javier A. Menendez, Ruth Lupu, Inderjit Mehmi, and David W. Griggs
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasms, Hormone-Dependent ,Angiogenesis ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Breast Neoplasms ,Adenocarcinoma ,medicine.disease_cause ,Immediate-Early Proteins ,Endocrinology ,Breast cancer ,Cell Movement ,Internal medicine ,Cell Adhesion ,medicine ,Humans ,Neoplasm Invasiveness ,Molecular pathology ,business.industry ,Growth factor ,medicine.disease ,Gene Expression Regulation, Neoplastic ,CYR61 ,Cancer cell ,Disease Progression ,Intercellular Signaling Peptides and Proteins ,Angiogenesis Inducing Agents ,Female ,Signal transduction ,Carcinogenesis ,business ,Cell Division ,Cysteine-Rich Protein 61 - Abstract
CYR61 (CNN1), a member of the cysteine rich 61/connective tissue growth factor/nephroblastoma overexpressed (CYR61/CTFG/NOV) family of growth regulators (CNN), is a pro-angiogenic factor that mediates diverse roles in development, cell proliferation, and tumorigenesis. We have recently shown that CYR61 is overexpressed in invasive and metastatic human breast cancer cells. Accordingly, elevated levels of CYR61 in breast cancer are associated with more advanced disease. Unfortunately, the exact mechanisms by which CYR61 promotes an aggressive breast cancer phenotype are still largely unknown. This review examines the functional role of CYR61 in breast cancer disease, presenting evidence that CYR61 signaling may play a major role in estrogen- as well as growth factor-dependent breast cancer progression. We also emphasize the functional significance of the molecular connection of CYR61 and its integrin receptor alpha(v)beta(3) enhancing breast cancer aggressiveness. Moreover, we describe experimental evidence that establishes a novel role for CYR61 determining the protection of human breast cancer cells against chemotherapy-induced apoptosis through its interactions with the integrin receptor alpha(v)beta(3). All these findings delineate a new noteworthy function of a CYR61/alpha(v)beta(3) autocrine-paracrine signaling pathway within both angiogenesis and breast cancer progression, which would allow a dual anti-angiogenic and anti-tumor benefit with a single drug.
- Published
- 2003
- Full Text
- View/download PDF
10. A deletion mutant of heregulin increases the sensitivity of breast cancer cells to chemotherapy without promoting tumorigenicity
- Author
-
Kozystof Bojanowski, Ella Atlas, Ruth Lupu, and Inderjit Mehmi
- Subjects
Cancer Research ,medicine.medical_specialty ,Neuregulin-1 ,Breast Neoplasms ,In Vitro Techniques ,Biology ,Transfection ,ErbB Receptors ,Internal medicine ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,ERBB3 ,skin and connective tissue diseases ,Receptor ,Molecular Biology ,Sequence Deletion ,Activator (genetics) ,Culture Media ,Endocrinology ,Doxorubicin ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,Neuregulin ,Female ,Signal transduction - Abstract
Heregulin (HRG) is an activator of the erbB2-, erbB3- and erbB4-(erbB-2/3/4) signaling pathway. Transfection of full-length HRG cDNA into the estrogen (E2)-dependent cell line MCF-7 promoted an invasive E2-independent phenotype, as well as persistent activation of the erbB-2/3/4 receptors. Moreover, HRG expression in MCF-7 cells renders the cells sensitive to the topoisomerase II inhibitor doxorubicin (Doxo). In an attempt to dissociate the tumorigenic effect of HRG from the sensitizing effect to chemotherapy, we constructed a structural deletion mutant of HRG. Transfection of the deletion mutant of HRG described in this study (HRG/M) into MCF-7 cells resulted in the dissociation of the tumor-promoting activity of HRG from the sensitization to Doxo, that is, although the cells did not become more aggressive or E2-independent they became more sensitive to Doxo. HRG/M was unable to autophosphorylate the erbB receptors and did not affect the level of MAPK phosphorylation. Furthermore, the intracellular localization of the protein was different from that of the full-length protein. Our data show that the HRG/M sequences are sufficient to sensitize MCF-7 cells to Doxo, and provide evidence that this sensitization is independent of erbB2 activation.
- Published
- 2003
- Full Text
- View/download PDF
11. Trastuzumab in combination with heregulin-activated Her-2 (erbB-2) triggers a receptor-enhanced chemosensitivity effect in the absence of Her-2 overexpression
- Author
-
Javier A. Menendez, Inderjit Mehmi, and Ruth Lupu
- Subjects
Transcriptional Activation ,Cancer Research ,Vincristine ,Paclitaxel ,Receptor, ErbB-2 ,medicine.medical_treatment ,Neuregulin-1 ,Apoptosis ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Transactivation ,chemistry.chemical_compound ,ErbB ,Trastuzumab ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Phosphorylation ,skin and connective tissue diseases ,neoplasms ,Cisplatin ,Chemotherapy ,business.industry ,Antibodies, Monoclonal ,Drug Synergism ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,Neuregulin ,Female ,Fluorouracil ,business ,medicine.drug ,Signal Transduction - Abstract
Purpose The decision for treating breast cancer patients with trastuzumab is based on HER-2 amplification and/or overexpression. Methods Using MCF-7 cells (Her-2 ±) engineered to overexpress heregulin (MCF-7/HRG), a ligand for the Her-2/3/4 network, we investigated whether HRG-induced transactivation of Her-2 affected breast cancer cell sensitivity to chemotherapy and whether trastuzumab trigger receptor-enhanced chemosensitivity (REC) when combined with chemotherapy without Her-2 overexpression. Results MCF-7/HRG cells were more than 10-fold resistant to the alkylating agent cisplatin (CDDP), while trastuzumab coexposure completely reversed HRG-promoted CDDP resistance. A synergistic interaction between trastuzumab in combination with CDDP (paclitaxel or vincristine) was obtained in MCF-7/HRG cells. Trastuzumab prevented activation of the antiapoptotic and proliferative cascades and inhibited HRG-induced Her-2/3 phosphorylation. CDDP efficacy was enhanced by trastuzumab in cells expressing endogenously high levels of HRG. Conversely, trastuzumab coexposure was ineffective in enhancing chemotherapy efficacy in cells that did not secrete HRG, such as MCF-7 cells overexpressing a structural mutated HRG isoform. Therefore, trastuzumab-induced REC, in the absence of Her-2 overexpression, occurs through the kinase activity of Her-2/3. Interestingly, HRG expression in tumor biopsies from invasive breast carcinomas (n = 189) revealed that, whereas the minority (12%) of Her-2 positive tumors (n = 60; 32%) demonstrated Her-2 phosphorylation, the majority (67%) of HRG-overexpressing and Her-2 tumors (n = 57; 30%) were in active Her-2 status. Conclusion We demonstrate that assessment of HRG expression and Her-2 activation define a particular breast cancer patient population for which trastuzumab plus CDDP or taxol are extremely efficient without Her-2 overexpression.
- Published
- 2006
12. Heregulin-triggered Her-2/neu signaling enhances nuclear accumulation of p21WAF1/CIP1 and protects breast cancer cells from cisplatin-induced genotoxic damage
- Author
-
Inderjit Mehmi, Ruth Lupu, and Javier A. Menendez
- Subjects
MAPK/ERK pathway ,Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,DNA Repair ,Cell Survival ,Receptor, ErbB-2 ,medicine.medical_treatment ,Neuregulin-1 ,Antineoplastic Agents ,Breast Neoplasms ,Cell Cycle Proteins ,Protein degradation ,Biology ,Transactivation ,medicine ,Tumor Cells, Cultured ,Humans ,neoplasms ,Protein kinase B ,Cellular localization ,Cell Proliferation ,Growth factor ,Cell cycle ,Gene Expression Regulation, Neoplastic ,Oncology ,Drug Resistance, Neoplasm ,Cancer research ,Female ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Cisplatin ,DNA Damage ,Signal Transduction - Abstract
Elevated levels of p21 WAF1/CIP1 , an important mediator of DNA repair, have been observed in various aggressive tumors as well as linked to chemoresistance. We examined whether heregulin (HRG), a member of the EGF-like growth factor family closely related to breast cancer tumurigencsis and metastasis, modulates p21 WAF1/CIP1 expression and cellular localization. We used a model system that consisted of MCF-7 cells and MCF-7 cells engineered to overexpress the full-length cDNA of the human HRG gene (MCF-7/HRG). MCF-7/HRG cells demonstrate constitutive hyperactivation of Her-2/neu receptor as well as activation of down-stream PI-3'K/AKT and MAPK signaling cascades. Immunoblotting analyses showed that MCF-7/HRG cells significantly up-regulate p21 WAF1/CIP1 expression relative to control MCF-7/pBABE cells, while a strong nuclear accumulation of p21 WAF1/CIP1 in MCF-7/HRG cells was revealed by immunofluorescence microscopy studies. Protein degradation analyses demonstrated that the half-life of p21 WAF1/CIP1 protein was increased from -35 min in control MCF-7/pBABE cells to ≥3 h in MCF-7/HRG cells. Pharmacological inactivation of the PI-3'K/AKT and MAPK completely prevented HRG-induced accumulation of p21 WAF1/CIP1 . A structural deletion mutant of HRG (HRG-M4) lacking the N-terminus sequence and the cytoplasmic-transmembrane region of HRG was generated to investigate whether secretion of HRG and transactivation of Her-2/neu actively contributed to HRG-regulated p21 WAF1/CIP1 expression and cellular localization. MCF-7 cells engineered to overexpress HRG-M4 did not demonstrate either activation of Her-2/neu, PI-3'K/AKT, or MAPK. Remarkably, HRG-M4 overexpression completely abolished the ability of HRG to promote nuclear accumulation of p21 WAF1/CIP1 and concomitantly enhanced the apoptotic effects of cisplatin towards breast cancer cells. This novel interplay between HRG and p21 WAF1/CIP1 strongly suggests that one mechanism of HRG-rcgulmcd breast cancer cell proliferation, survival, and/or sensitivity to genotoxic damage is to stabilize and promote a nuclear accumulation of p21 WAF1/CIP1
- Published
- 2005
13. A novel CYR61-triggered 'CYR61-alphavbeta3 integrin loop' regulates breast cancer cell survival and chemosensitivity through activation of ERK1/ERK2 MAPK signaling pathway
- Author
-
Poh K. Teng, Ruth Lupu, David W. Griggs, Inderjit Mehmi, Javier A. Menendez, and Luciano Vellon
- Subjects
MAPK/ERK pathway ,Cancer Research ,Integrins ,Paclitaxel ,Cell Survival ,MAP Kinase Signaling System ,Apoptosis ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Immediate-Early Proteins ,Breast cancer ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Receptors, Vitronectin ,Autocrine signalling ,Molecular Biology ,Protein kinase B ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Cell Cycle ,Cell cycle ,medicine.disease ,Metastatic breast cancer ,Gene Expression Regulation, Neoplastic ,CYR61 ,Cancer research ,Intercellular Signaling Peptides and Proteins ,Angiogenesis Inducing Agents ,Female ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Cysteine-Rich Protein 61 - Abstract
The angiogenic inducer CYR61 is differentially overexpressed in breast cancer cells exhibiting high levels of Heregulin (HRG), a growth factor closely associated with a metastatic breast cancer phenotype. Here, we examined whether CYR61, independently of HRG, actively regulates breast cancer cell survival and chemosensitivity, and the pathways involved. Forced expression of CYR61 in HRG-negative MCF-7 cells notably upregulated the expression of its own integrin receptor alphavbeta3 (200 times). Small peptidomimetic alphavbeta3 integrin antagonists dramatically decreased cell viability of CYR61-overexpressing MCF-7 cells, whereas control MCF-7/V remained insensitive. Mechanistically, functional blockade of alphavbeta3 specifically abolished CYR6-induced hyperactivation of ERK1/ERK2 MAPK, whereas the activation status of AKT did not decrease. Moreover, CYR61 overexpression rendered MCF-7 cells significantly resistant (10-fold) to Taxol-induced cytotoxicity. Remarkably, alphavbeta3 inhibition converted the CYR61-induced Taxol-resistant phenotype into a hypersensitive one. Thus, the augmentation of Taxol-induced apoptotic cell death in the presence of alphavbeta3 antagonists demonstrated a strong synergism as verified by the terminal transferase-mediated dUTP nick-end labeling (TUNEL) assay and by flow cytometric analysis for DNA content. Indeed, functional blockade of alphavbeta3, similarly to the pharmacological MAPK inhibitor U0126, synergistically increased both the proportion of CYR61-overexpressing breast cancer cells in the G2 phase of the cell cycle and the appearance of sub-G1 hypodiploid (apoptotic) cells caused by Taxol. Strikingly, CYR61 overexpression impaired the accumulation of wild-type p53 following Taxol exposure, while inhibition of alphavbeta3 or ERK1/ERK2 MAPK signalings completely restored Taxol-induced upregulation of p53. Moreover, antisense downregulation of CYR61 expression abolished the anchorage-independent growth of breast cancer cells engineered to overexpress HRG, and significantly increased their sensitivity to Taxol. Our data provide evidence that CYR61 is sufficient to promote breast cancer cell proliferation, cell survival, and Taxol resistance through a alphavbeta3-activated ERK1/ERK2 MAPK signaling. The identification of a 'CYR61-alphavbeta3 autocrine loop' in the epithelial compartment of breast carcinoma strongly suggests that targeting alphavbeta3 may simultaneously prevent breast cancer angiogenesis, growth, and chemoresistance.
- Published
- 2004
14. Pharmacological inhibition of fatty acid synthase (FAS): a novel therapeutic approach for breast cancer chemoprevention through its ability to suppress Her-2/neu (erbB-2) oncogene-induced malignant transformation
- Author
-
Vishal Verma, Javier A. Menendez, Poh K. Teng, Inderjit Mehmi, and Ruth Lupu
- Subjects
Cancer Research ,Antifungal Agents ,Receptor, ErbB-2 ,Apoptosis ,Breast Neoplasms ,Enzyme-Linked Immunosorbent Assay ,Biology ,Protein Serine-Threonine Kinases ,Chemoprevention ,Fas ligand ,Receptors, Tumor Necrosis Factor ,Malignant transformation ,chemistry.chemical_compound ,Mice ,Phosphatidylinositol 3-Kinases ,4-Butyrolactone ,ErbB ,Proto-Oncogene Proteins ,Cell Adhesion ,In Situ Nick-End Labeling ,Animals ,Humans ,fas Receptor ,Enzyme Inhibitors ,Molecular Biology ,Protein kinase B ,Cells, Cultured ,Oncogene ,Kinase ,Fatty Acids ,Fibroblasts ,Molecular biology ,Cerulenin ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,chemistry ,NIH 3T3 Cells ,Fatty Acid Synthases ,Mitogen-Activated Protein Kinases ,Proto-Oncogene Proteins c-akt - Abstract
We designed our experiments to evaluate whether fatty acid synthase (FAS), a lipogenic enzyme linked to tumor virulence in population studies of human cancer, is necessary for the malignant transformation induced by Her-2/neu (erbB-2) oncogene, which is overexpressed not only in invasive breast cancer but also in premalignant atypical duct proliferations and in ductal carcinoma in situ of the breast. To avoid the genetic complexities associated with established breast cancer cell lines, we employed NIH-3T3 mouse fibroblasts engineered to overexpress human Her-2/neu coding sequence. NIH-3T3/Her-2 cells demonstrated a significant upregulation of FAS protein expression, which was dependent on the upstream activation of mitogen-activated protein kinase and phosphatidylinositol 3′-kinase/AKT pathways. Remarkably, pharmacological FAS blockade using the mycotoxin cerulenin or the novel small compound C75 completely suppressed the state of Her-2/neu-induced malignant transformation by inhibiting the ability of NIH-3T3/Her-2 cells to grow under either anchorage-independent (i.e., to form colonies in soft agar) or low-serum monolayer conditions. Moreover, NIH-3T3/Her-2 fibroblasts were up to three times more sensitive to chemical FAS inhibitors relative to untransformed controls as determined by MTT-based cell viability assays. In addition, pharmacological FAS blockade preferentially induced apoptotic cell death of NIH-3T3/Her-2 fibroblasts, as determined by an ELISA for histone-associated DNA fragments and by the terminal deoxynucleotidyltransferase (TdT)-mediated nick end labeling assay (TUNEL). Interestingly, the degree of Her-2/neu oncogene expression in a panel of breast cancer cell lines was predictive of sensitivity to chemical FAS inhibitors-induced cytotoxicity, while low-FAS expressing and chemical FAS inhibitors-resistant MDA-MB-231 breast cancer cells became hypersensitive to FAS blockade when they were engineered to overexpress Her-2/neu. Our observations strongly suggest that inhibition of FAS activity may provide a new molecular avenue for chemotherapeutic prevention and/or treatment of Her-2/neu-related breast carcinomas. © 2004 Wiley-Liss, Inc.
- Published
- 2004
15. Inhibition of fatty acid synthase (FAS) suppresses HER2/neu (erbB-2) oncogene overexpression in cancer cells
- Author
-
Bharvi P. Oza, Santiago Ropero, Ruth Lupu, Ramon Colomer, Inderjit Mehmi, Javier A. Menendez, and Luciano Vellon
- Subjects
Small interfering RNA ,Antifungal Agents ,Cell Survival ,Receptor, ErbB-2 ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,Biology ,Antibodies, Monoclonal, Humanized ,Fas ligand ,HER2/neu ,Cell Line ,chemistry.chemical_compound ,4-Butyrolactone ,ErbB ,RNA interference ,Biomarkers, Tumor ,Gene silencing ,Humans ,RNA, Small Interfering ,skin and connective tissue diseases ,Cell Size ,Ovarian Neoplasms ,Multidisciplinary ,Antibodies, Monoclonal ,Genes, erbB-2 ,Trastuzumab ,Biological Sciences ,Fas receptor ,Cerulenin ,Gene Expression Regulation, Neoplastic ,chemistry ,Cancer research ,biology.protein ,Female ,Fatty Acid Synthases ,Signal Transduction ,Transcription Factors - Abstract
Fatty acid synthase (FAS) activity is a potential therapeutic target to treat cancer and obesity. Here, we have identified a molecular link between FAS and HER2 ( erb B-2) oncogene, a marker for poor prognosis that is overexpressed in 30% of breast and ovarian cancers. Pharmacological FAS inhibitors cerulenin and C75 were found to suppress p185 HER2 oncoprotein expression and tyrosine-kinase activity in breast and ovarian HER2 overexpressors. Similarly, p185 HER2 expression was dramatically down-regulated when FAS gene expression was silenced by using the highly sequence-specific mechanism of RNA interference (RNAi). Pharmacological and RNAi-mediated silencing of FAS specifically down-regulated HER2 mRNA and, concomitantly, caused a prominent up-regulation of PEA3, a transcriptional repressor of HER2 . A cytoplasmic redistribution of p185 HER2 was associated with marked morphological changes of FAS RNAi-transfected cells, whereas chemical inhibitors of FAS promoted a striking nuclear accumulation of p185 HER2 . The simultaneous targeting of FAS and HER2 by chemical FAS inhibitors and the humanized antibody directed against p185 HER2 trastuzumab, respectively, was synergistically cytotoxic toward HER2 overexpressors. Similarly, concurrent RNAi-mediated silencing of FAS and HER2 genes synergistically stimulated apoptotic cell death in HER2 overexpressors. p185 HER2 was synergistically down-regulated after simultaneous inhibition of FAS and HER2 by either pharmacological inhibitors or small interfering RNA. These findings provide evidence of an active role of FAS in cancer evolution by specifically regulating oncogenic proteins closely related to malignant transformation, strongly suggesting that HER2 oncogene may act as the key molecular sensor of energy imbalance after the perturbation of tumor-associated FAS hyperactivity in cancer cells.
- Published
- 2004
16. Overexpression and hyperactivity of breast cancer-associated fatty acid synthase (oncogenic antigen-519) is insensitive to normal arachidonic fatty acid-induced suppression in lipogenic tissues but it is selectively inhibited by tumoricidal alpha-linolenic and gamma-linolenic fatty acids: a novel mechanism by which dietary fat can alter mammary tumorigenesis
- Author
-
Javier A, Menendez, Santiago, Ropero, Inderjit, Mehmi, Ella, Atlas, Ramon, Colomer, and Ruth, Lupu
- Subjects
Dose-Response Relationship, Drug ,Blotting, Western ,Down-Regulation ,Fluorescent Antibody Technique ,alpha-Linolenic Acid ,Breast Neoplasms ,Arachidonic Acids ,Dietary Fats ,Cerulenin ,Adipocytes ,Tumor Cells, Cultured ,Humans ,Female ,Enzyme Inhibitors ,Fatty Acid Synthases ,Mitogen-Activated Protein Kinases ,gamma-Linolenic Acid ,Cell Division - Abstract
Activity and expression of fatty acid synthase (FAS), a critical enzyme in the de novo biosynthesis of fatty acids in mammals, is exquisitely sensitive to nutritional regulation of lipogenesis in liver or adipose tissue. Surprisingly, a number of studies have demonstrated hyperactivity and overexpression of FAS (oncogenic antigen-519) in a biologically aggressive subset of human breast carcinomas, suggesting that FAS-dependent neoplastic lipogenesis is unresponsive to nutritional regulation. We have assessed the role of omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) on the enzymatic activity and protein expression of tumor-associated FAS in SK-Br3 human breast cancer cells, an experimental paradigm of FAS-overexpressing tumor cells in which FAS enzyme constitutes up to 28%, by weight, of the cytosolic proteins. Of the omega-3 PUFAs tested, alpha-linolenic acid (ALA) dramatically reduced FAS activity in a dose-dependent manner (up to 61%). omega-3 PUFA docosahexaenoic acid (DHA) demonstrated less marked but still significant inhibitory effects on FAS activity (up to 37%), whereas eicosapentaenoic acid (EPA) was not effective. Of the omega-6 fatty acids tested, gamma-linolenic acid (GLA) was the most effective dose-dependent inhibitor of FAS activity, with a greater than 75% FAS activity reduction. Remarkably, omega-6 PUFAs linoleic acid (LA) and arachidonic acid (ARA), suppressors of both hepatic and adipocytic FAS-dependent lipogenesis, had no significant inhibitory effects on the activity of tumor-associated FAS in SK-Br3 breast cancer cells. Western blotting studies showed that down-regulation of FAS protein expression tightly correlated with previously observed inhibition of FAS activity, suggesting that ALA-, DHA-, and GLA-induced changes in FAS activity resulted from effects at the protein level. We investigated whether the FAS inhibitory effect of GLA and omega-3 PUFAs correlated with a cytotoxic effect related to a peroxidative mechanism. Measurement of cell viability by MTT assay indicated a significant cellular toxicity after ALA and GLA exposures. Furthermore, we observed a significant correlation between the ability of PUFAs to repress FAS and cause cell toxicity. In the presence of anti-oxidants (vitamin E), ALA and GLA dramatically lost their ability to inhibit FAS activity. Interestingly, a combination of ALA and GLA was FAS inhibitory in an additive manner, and this FAS repression was only partially reversible by vitamin E. In examining the molecular mechanisms underlying resistance of breast cancer-associated FAS to normal dietary fatty acid-induced suppression, a dramatic decrease of FAS accumulation was found after exposure of SK-Br3 cells to mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase (MAPK ERK1/2) inhibitor U0126, phosphatidylinositol-3'-kinase (PI-3'K) blocker LY294002, and/or anti-HER-2/neu antibody trastuzumab. Interestingly, a long-term exposure to pharmacological inhibitors of FAS activity cerulenin [(2S,3R) 2,3-epoxy-4-oxo-7E,10E-dodecadienamide] or C75 also resulted in a significant reduction of FAS accumulation. These data indicate that: a) GLA- and omega-3 PUFA-induced repression of tumor-associated FAS may result, at least in part, from a non-specific cytotoxic effect due to peroxidative mechanisms; b) alternatively, GLA and omega-3 PUFAs have a suppressive effect on FAS expression and activity that can result in the accumulation of toxic fluxes of the FAS substrate malonyl-CoA; c) GLA- and/or omega-3 PUFA-induced repression of tumor-associated FAS may represent a novel mechanism of PUFA-induced cytotoxicity clinically useful against breast carcinomas carrying overexpression of FAS enzyme; d) fundamental differences in the ability of FAS gene to respond to normal fatty acid's regulatory actions in lipogenic tissues may account for the observed extremely high levels of FAS in breast carcinoma; and e) FAS overexpression in SK-Br3 breast cancer cells is driven by increases in HER-2/neu signaling, acting in major part through a constitutive downstream art through a constitutive downstream activation of the MAPK ERK1/2 and PI-3'K/AKT transduction cascades.
- Published
- 2004
17. Novel signaling molecules implicated in tumor-associated fatty acid synthase-dependent breast cancer cell proliferation and survival: Role of exogenous dietary fatty acids, p53-p21WAF1/CIP1, ERK1/2 MAPK, p27KIP1, BRCA1, and NF-kappaB
- Author
-
Javier A, Menendez, Inderjit, Mehmi, Ella, Atlas, Ramon, Colomer, and Ruth, Lupu
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Time Factors ,Cell Survival ,Immunoblotting ,Active Transport, Cell Nucleus ,Breast Neoplasms ,Cell Cycle Proteins ,DNA Fragmentation ,Inhibitory Concentration 50 ,Cell Line, Tumor ,Cyclins ,In Situ Nick-End Labeling ,Humans ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Dose-Response Relationship, Drug ,BRCA1 Protein ,Tumor Suppressor Proteins ,Fatty Acids ,NF-kappa B ,Cerulenin ,Microscopy, Fluorescence ,Fatty Acid Synthases ,Mitogen-Activated Protein Kinases ,Tumor Suppressor Protein p53 ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction - Abstract
A biologically aggressive subset of human breast cancers has been demonstrated to overexpress fatty acid synthase (FAS), the key enzyme of endogenous FA biosynthesis. This breast cancer-specific activation of FAS-dependent lipogenesis, an anabolic-energy-storage pathway of minor importance in normal cells, would render breast cancer cells more vulnerable to anti-metabolite interventions with FAS as therapeutic target. Not surprisingly, pharmacological inhibitors of FAS have been reported to produce both cytostatic and cytotoxic effects in human breast cancer cells, as well as to suppress DNA replication. However, the signal transduction pathway(s) that link FAS hyperactivity and breast cancer cell growth has been unresolved. Here, we have attempted to provide a systematic approach to assess the role of FAS signaling on the survival and proliferation of human breast cancer cells. First, we assessed the level of FAS protein in a panel of human breast cancer cell lines (MCF-7, MDA-MB-231, MDA-MB-453, MDA-MB-435, ZR-75B, T47-D, BT-474, and SK-Br3). FAS expression was graded from ++++ (overexpression) in SK-Br3 cells to + (very low expression) in MDA-MB-231 cells. No correlation was noted between FAS overexpression and estrogen receptor (ER) or progesterone receptor (PR) status, whereas a positive correlation was found between high levels of FAS expression and the amplification and/or overexpression of HER-2/neu oncogene. Because metabolic adaptation of breast cancer cells to the ambient fatty acid concentration may be relevant to the goal of utilizing FAS inhibition as a chemotherapeutic target, we evaluated the effect of exogenous dietary fatty acids on the cytotoxicity resulting from the inhibition of FAS activity. Pharmacological inhibition of FAS activity by the natural antibiotic cerulenin [(2S,3R)-2,3-epoxy-4-oxo-7E,10E-dodecadienamide] resulted in a dose-dependent cytotoxicity which positively paralleled the endogenous level of FAS. Supraphysiological levels of exogenous oleic acid (OA), a omega-9 monounsaturated fatty acid synthesized from a primary-end product of FAS palmitate, significantly diminished cell toxicity caused by cerulenin. Indeed, OA exposure significantly reduced FAS activity and expression by 55% in FAS-overexpressing SK-Br3 cells. omega-3 (alpha-linolenic acid, eicosapentaenoic acid and docosahexaenoic acid) and omega-6 (linoleic acid and arachidonic acid) polyunsaturated fatty acids (PUFAs), however, were unable to rescue breast cancer cells from cerulenin-induced cytotoxicity. Pharmacological blockade of FAS activity in FAS-overexpressing SK-Br3 cells resulted in apoptosis as determined by an enzyme-linked immunosorbent assay for histone-associated DNA fragments, and confirmed by TUNEL DNA-end labeling experiments. We further characterized signaling molecules that participate in the cellular events that follow inhibition of FAS activity and precede apoptosis in breast cancer cells. In SK-Br3 cells, cerulenin-induced inhibition of FAS activity resulted in down-regulation of p53, and up-regulation of cyclin-dependent kinase inhibitor (CDKi) p21WAF1/CIP1. Treatment with cerulenin or a novel small-molecule inhibitor of FAS C75 resulted in a dramatic accumulation of CDKi p27KIP1, which was accompanied by a noteworthy translocation of p27KIP1 from cytosol to cell nuclei. Strikingly, FAS inhibition also caused a significant activation of the Raf-mitogen-activated protein kinase (MEK) extracellular signal-regulated kinase (ERK1/2) cell survival pathway. Interestingly, we demonstrated that inhibition of FAS activity increased the nuclear-to-cytoplasmic ratio of BRCA1, a breast cancer tumor suppressor protein, as well as it induced a nuclear translocalization of the anti-apoptotic nuclear transcription factor-kappaB (NF-kappaB). In conclusion, here we demonstrate that: a) breast cancer cells retain dependence on endogenous fatty acid synthesis and sensitivity to FAS inhibition in the presence of supraphysiological levels of dietary fatty acids, supporting the notion that FAS inhibition may be useful in treFAS inhibition may be useful in treating breast cancer in vivo; b) endogenous fatty acid synthesis is functional in breast cancer cells and is vital since its pharmacological inhibition is cytotoxic by promoting apoptosis, and c) specific blockade of FAS activity induces the accumulation, activation, and/or cellular relocalization of multiple and diverse pro- and anti-apoptotic signaling pathways, suggesting that p53-p21WAF1/CIP1, ERK1/2 MAPK, p27KIP1, BRCA1, and NF-kappaB play a novel role in the breast cancer cell response to a metabolic stress after perturbation of FAS-dependent de novo fatty acid biosynthesis.
- Published
- 2004
18. Black cohosh, a menopausal remedy, does not have estrogenic activity and does not promote breast cancer cell growth
- Author
-
Kronenberg F, Nuntanakorn P, Ruth Lupu, Atlas E, Tsai Ms, Hellen A. Oketch-Rabah, Inderjit Mehmi, Pisha E, and Edward J. Kennelly
- Subjects
Oncology ,Cimicifuga ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Mammary gland ,Black cohosh ,Breast Neoplasms ,Ribonucleases ,Actaea racemosa ,Breast cancer ,Cell Line, Tumor ,Internal medicine ,Cell Adhesion ,medicine ,Humans ,RNA, Messenger ,biology ,Plant Extracts ,business.industry ,Cancer ,Estrogens ,Alkaline Phosphatase ,biology.organism_classification ,medicine.disease ,Menopause ,medicine.anatomical_structure ,Endocrinology ,Receptors, Estrogen ,Estrogen ,Adenocarcinoma ,business ,Cell Division - Abstract
Black cohosh is an increasingly popular alternative to estrogen replacement therapy for the relief of menopausal symptoms, primarily hot flushes. However, an important consideration for long-term therapy is potential toxicity and carcinogenicity. Therefore, we undertook a study to assess the estrogenic activity of black cohosh to examine its safety for those with, or at high risk of developing, breast cancer. Several assays were utilized as listed: RNAse protection assays, which ascertain the regulation of the expression of E2-responsive genes; estrogen-responsive-element (ERE)-luciferase, which determines modulation of the ER function by transactivation of the ERE; the Ishikawa cell system, which has an E2-regulated endogenous alkaline phosphatase; and colony formation of ER-expressing breast cancer cells, which indicates possible progression of early stage breast cancer into a more aggressive state. Black cohosh extracts did not demonstrate estrogenic activity in any of these assay systems. This is an encouraging step in the assessment of the safety of black cohosh for treatment of menopausal hot flushes.
- Published
- 2003
- Full Text
- View/download PDF
19. Blockage of heregulin expression inhibits tumorigenicity and metastasis of breast cancer
- Author
-
Careen K. Tang, Lisa A. Shamon-Taylor, Miaw Sheue Tsai, Inderjit Mehmi, and Ruth Lupu
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Neuregulin-1 ,Breast Neoplasms ,Biology ,Metastasis ,Breast cancer ,ErbB ,Internal medicine ,Genetics ,medicine ,Gene silencing ,Humans ,Neoplasm Metastasis ,skin and connective tissue diseases ,Molecular Biology ,Cell growth ,Growth factor ,Genes, erbB-2 ,medicine.disease ,Matrix Metalloproteinases ,Endocrinology ,Cell Transformation, Neoplastic ,Cancer research ,Neuregulin ,Female ,Breast disease ,Mitogen-Activated Protein Kinases ,Signal Transduction - Abstract
The growth factor heregulin (HRG), expressed in about 30% of breast cancer tumors, activates the erbB-2 receptor via induction of heterodimeric complexes of erbB-2 with erbB-3 or erbB-4. HRG induces tumorigenicity and metastasis of breast cancer cells. Our investigation into whether HRG is a factor likely to promote tumor formation independently of erbB-2 overexpression concludes that blockage of HRG expression suppresses the aggressive phenotype of MDA-MB-231 breast cancer cells by inhibiting cell proliferation, preventing anchorage-independent growth, and suppressing the invasive potential of the cells in vitro. More importantly, we observed a marked reduction in tumor formation, tumor size, and a lack of metastasis in vivo. These studies were achieved by blocking HRG expression in MDA-MB-231 cells using an HRG antisense cDNA. In the search for the mechanism by which blockage of HRG reverts this aggressive phenotype, we discovered that the cells in which HRG is blocked exhibit a marked decrease in erbB activation and a significant reduction in MMP-9 activity, demonstrating a direct causal role in HRG induction of tumorigenicity. Our study is the first report and serves as a proof of the concept that HRG is a key promoter of breast cancer tumorigenicity and metastasis independently of erbB-2 overexpression and should be deemed a potential target in developing therapies for breast cancer.
- Published
- 2003
20. Heregulin is sufficient for the promotion of tumorigenicity and metastasis of breast cancer cells in vivo
- Author
-
Ella, Atlas, Marina, Cardillo, Inderjit, Mehmi, Hengameh, Zahedkargaran, Careen, Tang, and Ruth, Lupu
- Subjects
Vascular Endothelial Growth Factor A ,Lymphokines ,Carcinogenicity Tests ,MAP Kinase Signaling System ,Vascular Endothelial Growth Factors ,Neuregulin-1 ,Ovariectomy ,Mice, Nude ,Breast Neoplasms ,Endothelial Growth Factors ,Gene Expression Regulation, Neoplastic ,Mice ,Cell Transformation, Neoplastic ,Mammary Glands, Animal ,Matrix Metalloproteinase 9 ,Lymphatic Metastasis ,Axilla ,Tumor Cells, Cultured ,ras Proteins ,Animals ,Humans ,Intercellular Signaling Peptides and Proteins ,Female - Abstract
Resistance of breast carcinomas to hormonal therapy is a clinical obstacle for the treatment of breast cancer. The molecular mechanisms and the factors involved in the progression of tumors from an estrogen (E2)-dependent to an E2-independent phenotype are not entirely understood. Heregulin (HRG) is a pleiotropic growth factor that binds to the erbB family of receptors, which are correlated with breast cancer progression and an aggressive phenotype in the breast carcinomas overexpressing the receptors. Previous studies in transgenic mice have shown that HRG is sufficient to induce mammary gland transformation and proliferation in the presence of hormonal stimulation. However, these studies did not address the important issue of the E2 independence that is part of the progression of breast cancer. In this study, we investigated the role of HRG in E2 independence. We were able to determine that HRG up-regulation was sufficient for the development of mammary tumors in the absence of E2 stimulation, a situation that mimics the progression of the human disease. We demonstrated that in ovariectomized nude mice, HRG induced E2 independence and antiestrogen resistance and promoted metastasis and preneoplastic transformation of the adjacent mouse mammary tissue. We show that one of the mechanisms by which HRG achieves the aggressive phenotype may be mediated via an increase in activated mitogen-activated protein kinase, an increase in a matrix-degrading enzyme, MMP-9, and the overexpression of vascular endothelial growth factors. The up-regulation of these genes occurred in the absence of any additional stimulation, in an autocrine manner. Our data provide new insights into the mechanisms of breast cancer progression in vivo, and reinforce the important role that HRG plays in this process.
- Published
- 2003
21. Expression and regulation of Cyr61 in human breast cancer cell lines
- Author
-
Ruth Lupu, Inderjit Mehmi, Miaw Sheue Tsai, Patricia Li, and Daphne F. Bogart
- Subjects
Selective Estrogen Receptor Modulators ,Transcriptional Activation ,Cancer Research ,Neoplasms, Hormone-Dependent ,Active Transport, Cell Nucleus ,Drug Resistance ,Estrogen receptor ,Breast Neoplasms ,Tretinoin ,Biology ,Adenocarcinoma ,Transfection ,Immediate-Early Proteins ,Breast cancer ,Calcitriol ,Estrogen Receptor Modulators ,medicine ,Genetics ,Tumor Cells, Cultured ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,RNA, Neoplasm ,skin and connective tissue diseases ,Growth Substances ,Molecular Biology ,Fulvestrant ,Genes, Immediate-Early ,Regulation of gene expression ,Estradiol ,Estrogens ,medicine.disease ,Neoplasm Proteins ,Protein Structure, Tertiary ,Gene Expression Regulation, Neoplastic ,Tamoxifen ,Phenotype ,Receptors, Estrogen ,CYR61 ,Cancer cell ,Cancer research ,Intercellular Signaling Peptides and Proteins ,Tetradecanoylphorbol Acetate ,Female ,medicine.drug ,Cysteine-Rich Protein 61 - Abstract
We have shown that Cyr61, an angiogenic regulator, is overexpressed in invasive and metastatic human breast cancer cells and tumor biopsies. We have further demonstrated that Cyr61 promotes acquisition of estrogen-independence and anti-estrogen resistance in vivo in breast cancer cells. Moreover, we have demonstrated that Cyr61 induces tumor formation and tumor vascularization in vivo, events mediated through the activation of the MAPK and the Akt signaling pathways. Here we investigate how Cyr61 expression is regulated in both estrogen receptor (ER)-positive and ER-negative breast cancer cells. We demonstrate that Cyr61 mRNA and protein expression is inducible by estrogen and anti-estrogens in ER-positive breast cancer cells. We show that a labile protein as well as a negative regulator might be involved in Cyr61 expression in estrogen-dependent breast cancer cells. Other important regulators of Cyr61 expression in breast cancer cells that we found are the phorbol ester TPA, vitamin D, and retinoic acid. TPA causes positive regulation of Cyr61 expression in ER-positive MCF-7 cells. Vitamin D induces a transient stimulatory effect on Cyr61 gene expression. Lastly, retinoic acid has a negative effect on Cyr61 expression and downregulates its expression in MCF-7 cells. Interestingly, most of these effects are not seen in aggressive breast cancer cells that do not express ER and express high levels of Cyr61, such as the MDA-MB-231 cells. Our results are in agreement with our knowledge that Cyr61 promotes tumor growth, and that tumor-promoting agents have a positive impact on cells that express low levels of Cyr61, such as the ER-positive breast cancer cells; however, these agents have no significant effect on cells that express high levels of Cyr61. Our findings suggest an association between increased Cyr61 expression and an aggressive phenotype of breast cancer cells.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.