1
|
Mirzaei Bavil F, Alipour MR, Keyhanmanesh R, Alihemmati A, Ghiyasi R, Mohaddes G. Ghrelin Decreases Angiogenesis, HIF-1α and VEGF Protein Levels in Chronic Hypoxia in Lung Tissue of Male Rats. Adv Pharm Bull 2015; 5:315-20. [PMID: 26504752 DOI: 10.15171/apb.2015.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/14/2014] [Accepted: 10/18/2014] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Hypoxia is a condition of decreased availability of oxygen. When cells are exposed to a low oxygen environment, they impel the hypoxia responses to adapt to new situation. The hypoxia response leads to the activation of various cellular signaling pathways. The aim of this study was to evaluate the effect of ghrelin on angiogenesis, Hypoxia-Inducible-Factor-1α (HIF-1) and Vascular endothelial growth factor (VEGF) levels in normobaric hypoxia situation. METHODS Twenty four animals were divided into 4 groups (n=6): control (C), ghrelin (Gh), hypoxia (H), and hypoxic animals that received ghrelin (H+Gh). Hypoxia (11%) was induced by an Environmental Chamber System GO2 Altitude. Animals in ghrelin groups received a subcutaneous injection of ghrelin (150 μg/kg/day) for 14 days. RESULTS Our results showed that hypoxia significantly (p<0.05) increased angiogenesis without any significant changes on HIF-1 and VEGF levels, whereas ghrelin significantly (p<0.05) decreased angiogenesis, expression of HIF-1 and VEGF in this condition. Ghrelin administration did not show any significant changes in normal conditions. CONCLUSION Ghrelin had no effect on angiogenesis, expression of HIF-1 and VEGF in normal oxygen conditions but it reduced angiogenesis process in lung tissue with reducing the level of HIF and VEGF in hypoxic condition. Therefore, effect of ghrelin on angiogenesis could be related to blood oxygen level.
Collapse
Affiliation(s)
- Fariba Mirzaei Bavil
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alipour
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rafigheh Ghiyasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gisou Mohaddes
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Che Y, Zhang DY, Ye F, Wu B, Jiang B. Combination of flavopiridol and embelin effectively inhibit cell growth in hepatocellular carcinoma depending on regulatory relationship between CDK6 and XIAP. Med Chem Res 2011. [DOI: 10.1007/s00044-011-9867-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
3
|
Nelson DM, Joseph B, Hillion J, Segal J, Karp JE, Resar LMS. Flavopiridol induces BCL-2 expression and represses oncogenic transcription factors in leukemic blasts from adults with refractory acute myeloid leukemia. Leuk Lymphoma 2011; 52:1999-2006. [PMID: 21728742 PMCID: PMC3214625 DOI: 10.3109/10428194.2011.591012] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Flavopiridol is a cyclin-dependent kinase inhibitor that induces cell cycle arrest, apoptosis, and clinical responses in selected patients with acute myeloid leukemia (AML). A better understanding of the molecular pathways targeted by flavopiridol is needed to design optimal combinatorial therapy. Here, we report that in vivo administration of flavopiridol induced expression of the BCL-2 anti-apoptotic gene in leukemic blasts from adult patients with refractory AML. Moreover, flavopiridol repressed the expression of genes encoding oncogenic transcription factors (HMGA1, STAT3, E2F1) and the major subunit of RNA Polymerase II. Our results provide mechanistic insight into the cellular pathways targeted by flavopiridol. Although further studies are needed, our findings also suggest that blocking anti-apoptotic pathways could enhance cytotoxicity with flavopiridol.
Collapse
Affiliation(s)
- Dwella M. Nelson
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Biju Joseph
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Joelle Hillion
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Jodi Segal
- Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Judith E. Karp
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Linda M. S. Resar
- Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Division of General Internal Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Division of Hematologic Malignancy; The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
4
|
Sun Y, Li YX, Wu HJ, Wu SH, Wang YA, Luo DZ, Liao DJ. Effects of an Indolocarbazole-Derived CDK4 Inhibitor on Breast Cancer Cells. J Cancer 2011; 2:36-51. [PMID: 21234300 PMCID: PMC3020360 DOI: 10.7150/jca.2.36] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Accepted: 01/06/2011] [Indexed: 01/13/2023] Open
Abstract
Introduction: Cyclin D1 (D1) binds to cyclin-dependent kinases (CDK) 4 or 6 to form a holoenzyme that phosphorylates the Rb protein to promote cell cycle progression from G1 to S phase. Therefore, targeting CDK4/6 may be a good strategy for chemotherapy of cancer. We performed a proof-of-principle study to determine the effect of Naphtho [2, 1-α] pyrrolo [3, 4-c] carbazole-5, 7 (6H, 12H)-dione (NPCD), a novel CDK4 inhibitor, on breast cancer cell lines. Methods: NPCD was synthesized and purified to over 99% purity verified by HPLC. MCF7, MB231, MCF15, T47D and GI101Ap human breast cancer cells were analyzed for the efficacy of NPCD with MTT and clonogenic assays, with FACS and staining for ethidium bromide and acridine orange for cell death and cell cycle profile. Western blot, reverse transcription and PCR were used for studies of gene expression, and co-immunoprecipitation for protein-complex formation. Results: MTT assay showed that NPCD caused growth arrest and apoptosis of MCF7, MDA-MB231, T47D, MCF15 and GI101Ap cells with an IC50 ranging between 3 to 8 µM given as a single dose. The growth arrest persisted for many days after cessation of the treatment, as shown in a clonogenic assay. NPCD could induce or reduce the D1 and CDK4 protein levels, depending on the cell line, but this effect was not correlated with its efficacy. Phosphorylation of D1 at Thr286 was decreased but it unexpectedly did not correlate with the change in D1 level in the cell lines studied. Phosphorylation of the Rb protein was decreased as expected whereas the p27kip1 protein level was decreased unexpectedly. Protein levels of p21cip1, CDK2 and cyclin E were also decreased in some, but not all, of the cell lines, whereas the mRNA levels of D1, CDK4, cyclin E, CDK2, p27kip1 and p21cip1 were increased in different cell lines. Conclusions: NPCD can cause long-lasting growth arrest and cell death of breast cancer cell lines at an IC50 of 3-8 µM. Decreased phosphorylation of Rb by D1-CDK4/6 and decreased p27kip1 protein level may be part of the underlying mechanism.
Collapse
Affiliation(s)
- Yuan Sun
- 1. Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | | | | | | | | | | | |
Collapse
|
5
|
Galatin PS, Advani RH, Fisher GA, Francisco B, Julian T, Losa R, Sierra MI, Sikic BI. Phase I trial of oblimersen (Genasense®) and gemcitabine in refractory and advanced malignancies. Invest New Drugs 2010; 29:971-7. [PMID: 20349264 DOI: 10.1007/s10637-010-9416-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 03/02/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Overexpression of Bcl-2 is associated with worse prognosis for a number of cancer types. The present study was designed to determine the maximum tolerated dose (MTD) of oblimersen (antisense Bcl-2) and gemcitabine when administered to patients with refractory malignancies. MATERIALS AND METHODS Sixteen patients with advanced solid tumors refractory to standard therapies were treated with escalating doses of oblimersen continuous, 120-h intravenous infusion given every 14 days, with a fixed-dose-rate intravenous infusion of gemcitabine administered on day 5 of each cycle. Serial plasma samples were collected to calculate the pharmacokinetics of oblimersen and gemcitabine, and also to measure the effect of oblimersen on Bcl-2 expression. RESULTS 7 women and 9 men, median age 55 years (range 35-74 years), received a 5-day infusion of oblimersen at dose levels of 5 mg/kg/day (n = 4) or 7 mg/kg/day (n = 12). On the 5th day of the infusion, gemcitabine was given at 10 mg/m(2)/h for a total dose of 1,000 mg/m(2) (n = 7; cohorts I and II), 1,200 mg/m(2) (n = 3; cohort III), or 1,500 mg/m(2) (n = 6; cohort IV). Edema was the dose-limiting toxicity (DLT), necessitating expansion of cohort IV. No subsequent DLTs were noted. Thus, the maximum planned doses were well tolerated, and a formal MTD was not determined. Most hematologic toxicities were grade 1 or 2. There was low-grade fatigue, nausea/vomiting, and myalgias/arthralgias. Oblimersen C(ss) and AUC increased in relation to the dose escalation, but gemcitabine triphosphate levels did not correlate well with dose. There were no objective responses, though 5 patients had stable disease. A >75% reduction in Bcl-2 expression in peripheral blood mononuclear leucocytes was seen more frequently in patients who achieved stable disease than in progressing patients. CONCLUSIONS The maximal planned dose levels of oblimersen and gemcitabine in combination were well tolerated. Only one DLT (edema) occurred. There was a correlation between Bcl-2 reduction and stable disease. The recommended doses of the drugs for future studies are 7 mg/kg/day of oblimersen on days 1-5, and gemcitabine 1,500 mg/m(2) on day 5, every two weeks.
Collapse
Affiliation(s)
- Peter S Galatin
- Department of Medicine, Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Abstract
The standard therapeutic approaches for acute myeloid leukemia (AML) continue to be based on anthracyclines and cytarabine. However, the prognosis for AML remains poor, especially for patients with high-risk disease. During the past decade, promising novel agents that target DNA replication and repair, as well as cell cycling and apoptosis, have been developed and are being actively investigated in AML. Among these agents is flavopiridol, which interferes with key steps of the cell cycle and effectively promotes cell death, and voreloxin, an intercalating agent that also targets topoisomerase II. Also under clinical study in AML are oligonucleotide antisense constructs, which suppress the translation of proteins essential for leukemic blast survival and proliferation, and agents that target antiapoptotic cascades. In summary, it is hoped that novel therapies such as these will augment and/or supplant our current cytarabine- and anthracycline-based approaches, overcome active drug-resistance pathways, and eventually improve outcomes for patients with AML.
Collapse
|
7
|
Fathi AT, Grant S, Karp JE. Exploiting cellular pathways to develop new treatment strategies for AML. Cancer Treat Rev 2010; 36:142-50. [PMID: 20056334 DOI: 10.1016/j.ctrv.2009.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 12/01/2009] [Accepted: 12/05/2009] [Indexed: 12/14/2022]
Abstract
The standard approaches to the treatment of acute myeloid leukemia (AML) have been predominantly based on cytarabine and anthracyclines. Yet, the outcomes associated with AML continue to be poor, especially for those patients who are older or carry higher-risk disease. In recent years, extensive research has led to the development and study of novel agents which target AML by diverse and varied mechanisms. Among these are targeted therapeutics such as kinase inhibitors and oligonucleotide constructs. These aim to suppress the production or activity of proteins, such as FLT3 and BCL2, among others, and thus disrupt related signaling cascades essential for leukemogenesis and proliferation. In addition, other agents like flavopiridol appear to target the myeloid blast by various mechanisms including suppression of cyclin-dependent kinases and interference with nucleotide synthesis. Another class of novel therapies includes inhibitors of histone deacetylase, which cause growth arrest and apoptosis through histone acetylation and resultant conformational changes. Clinical trials are now studying these and other agents alone and in combination with traditional cytotoxic therapies, with some encouraging results. In this review, we aim to provide a summary of the preclinical and clinical investigations of selected promising agents currently under study.
Collapse
Affiliation(s)
- Amir T Fathi
- Division of Hematologic Malignancies, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, 1650 Orleans Street, Baltimore, MD 21231, USA.
| | | | | |
Collapse
|
8
|
Rom J, von Minckwitz G, Marmé F, Ataseven B, Kozian D, Sievert M, Schlehe B, Schuetz F, Scharf A, Kaufmann M, Sohn C, Schneeweiss A. Phase I study of apoptosis gene modulation with oblimersen within preoperative chemotherapy in patients with primary breast cancer. Ann Oncol 2009; 20:1829-35. [PMID: 19605509 DOI: 10.1093/annonc/mdp208] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Expression of the Bcl-2 protein confers resistance to chemotherapy-mediated apoptotic signals in patients with breast cancer. We investigated effects of Bcl-2 down-regulation by the Bcl-2 antisense oligodeoxynucleotide oblimersen in breast tumor biopsies. Oblimersen targets Bcl-2 messenger RNA (mRNA), down-regulates Bcl-2 protein translation and enhances antitumor effects of subtherapeutic chemotherapy doses. Within a phase I trial, we administered escalating doses of oblimersen (3, 5 or 7 mg/kg/day) as continuous infusion on days 1-7 in combination with standard-dose docetaxel (Taxotere), Adriamycin and cyclophosphamide (TAC) on day 5 as preoperative chemotherapy in 28 patients with T2-4 tumors. Effects of oblimersen were evaluated in tumor biopsies and peripheral blood mononuclear cells (PBMCs) 4 days after start of oblimersen and before TAC treatment by quantitative microfluidic real-time PCR. Read-outs consisted in measurement of Bcl-2 mRNA modulations and of 18 putative predictive markers. Two of 13 patients showed a diminution of Bcl-2 transcripts after 4 days of treatment with oblimersen 5 mg/kg/day. PBMCs could not be evaluated as a surrogate tissue because no qualified RNA could be isolated. Nevertheless, we demonstrated feasibility to process clinical samples and to obtain good quality RNA from tumor biopsies and indicated the potential of oblimersen to lower Bcl-2 mRNA in breast cancer.
Collapse
Affiliation(s)
- J Rom
- Department of Gynecology and Obstetrics, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Affiliation(s)
- Ali Hachem
- University of Maryland Greenebaum Cancer Center, 9-011 BRB, 655 West Baltimore St, Baltimore MD 21201, USA
| | | |
Collapse
|
10
|
Caligaris-Cappio F, Cignetti A, Granziero L, Ghia P. Chronic lymphocytic leukaemia: a model for investigating potential new targets for the therapy of indolent lymphomas. Best Pract Res Clin Haematol 2002; 15:563-75. [PMID: 12468406 DOI: 10.1053/beha.2002.0212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We address two key issues whose investigation may help to define new prognostic parameters and new potential targets for therapeutic intervention. First, which are the conceptual implications of the cellular origin of indolent lymphomas? Second, how may deciphering the biology of chronic lymphocytic leukaemia (CLL) lead to the development of new modalities of treatment? The latter issue is articulated in the following three key questions. (1) Which are the molecular pathways through which the microenvironment exerts its influence on the malignant clone? (2) What are the relationships between proliferation and defective apoptosis? (3)Is there any evidence of a role for antigenic stimulation?
Collapse
MESH Headings
- Apoptosis
- Cell Transformation, Neoplastic/pathology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphoma, Non-Hodgkin/etiology
- Lymphoma, Non-Hodgkin/pathology
- Models, Biological
Collapse
Affiliation(s)
- Federico Caligaris-Cappio
- Department of Oncological Sciences, University of Torino, Division of Clinical Immunology and Haematology, Ospedale Mauriziano Umberto I, Torino, Italy
| | | | | | | |
Collapse
|