1
|
Urgard E, Reigo A, Reinmaa E, Rebane A, Metspalu A. Human basonuclin 2 up-regulates a cascade set of interferon-stimulated genes with anti-cancerous properties in a lung cancer model. Cancer Cell Int 2017; 17:18. [PMID: 28184177 PMCID: PMC5294813 DOI: 10.1186/s12935-017-0394-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 02/01/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Human basonuclin 2 (BNC2) acts as a tumor suppressor in multiple cancers in an as yet unidentified manner. The role and expression of the BNC2 gene in lung cancer has not yet been investigated. METHODS BNC2 expression was studied in the A549 and BEAS-2B cell lines, as well as in lung cancer tissue. Illumina array analysis and a viability assay were used to study the effects of transient transfection of BNC2 in A549 cells. Ingenuity pathway analysis and g:Profiler were applied to identify affected pathways and networks. RT-qPCR was used to validate the array results. RESULTS We showed the reduced mRNA expression of BNC2 in non-small cell lung cancer tissue and lung cancer cell line A549 compared to non-cancerous lung tissue and BEAS-2B cells, respectively. Further array analysis demonstrated that the transfection of BNC2 into A549 cells resulted in the increased expression of 139 genes and the down-regulation of 13 genes. Pathway analysis revealed that half of the up-regulated genes were from the interferon/signal transducer and activator of transcription signaling pathways. The differential expression of selected sets of genes, including interferon-stimulated and tumor suppressor genes of the XAF1 and OAS families, was confirmed by RT-qPCR. In addition, we showed that the over-expression of BNC2 inhibited the proliferation of A549 cells. CONCLUSION Our data suggest that human BNC2 is an activator of a subset of IFN-regulated genes and might thereby act as a tumor suppressor.
Collapse
Affiliation(s)
- Egon Urgard
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia.,Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Anu Reigo
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Eva Reinmaa
- Department of Immunoanalysis, United Laboratories, Tartu University Hospital, Tartu, Estonia
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia.,Estonian Genome Center, University of Tartu, Tartu, Estonia
| |
Collapse
|
2
|
Sabbatino F, Wang Y, Scognamiglio G, Favoino E, Feldman SA, Villani V, Flaherty KT, Nota S, Giannarelli D, Simeone E, Anniciello AM, Palmieri G, Pepe S, Botti G, Ascierto PA, Ferrone CR, Ferrone S. Antitumor Activity of BRAF Inhibitor and IFNα Combination in BRAF-Mutant Melanoma. J Natl Cancer Inst 2016; 108:djv435. [PMID: 26851802 PMCID: PMC4948304 DOI: 10.1093/jnci/djv435] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 12/21/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND BRAF(V600E)-mediated MAPK pathway activation is associated in melanoma cells with IFNAR1 downregulation. IFNAR1 regulates melanoma cell sensitivity to IFNα, a cytokine used for the adjuvant treatment of melanoma. These findings and the limited therapeutic efficacy of BRAF-I prompted us to examine whether the efficacy of IFNα therapy of BRAF(V600E) melanoma can be increased by its combination with BRAF-I. METHODS BRAF/NRAS genotype, ERK activation, IFNAR1, and HLA class I expression were tested in 60 primary melanoma tumors from treatment-naive patients. The effect of BRAF-I on IFNAR1 expression was assessed in three melanoma cell lines and in four biopsies of BRAF(V600E) metastases. The antiproliferative, pro-apoptotic and immunomodulatory activity of BRAF-I and IFNα combination was tested in vitro and in vivo utilizing three melanoma cell lines, HLA class I-MA peptide complex-specific T-cells and immunodeficient mice (5 per group for survival and 10 per group for tumor growth inhibition). All statistical tests were two-sided. Differences were considered statistically significant when the P value was less than .05. RESULTS The IFNAR1 level was statistically significantly (P < .001) lower in BRAF(V600E) primary melanoma tumors than in BRAF wild-type tumors. IFNAR1 downregulation was reversed by BRAF-I treatment in the three melanoma cell lines (P ≤ .02) and in three out of four metastases. The IFNAR1 level in the melanoma tumors analyzed was increased as early as 10 to 14 days following the beginning of the treatment. These changes were associated with: 1) an increased susceptibility in vitro of melanoma cells to the antiproliferative (P ≤ .04), pro-apoptotic (P ≤ .009) and immunomodulatory activity, including upregulation of HLA class I antigen APM component (P ≤ .04) and MA expression as well as recognition by cognate T-cells (P < .001), of BRAF-I and IFNα combination and 2) an increased survival (P < .001) and inhibition of tumor growth of melanoma cells (P < .001) in vivo by BRAF-I and IFNα combination. CONCLUSIONS The described results provide a strong rationale for the clinical trials implemented in BRAF(V600E) melanoma patients with BRAF-I and IFNα combination.
Collapse
|
3
|
Synthesis of new indole-based bisphosphonates and evaluation of their chelating ability in PE/CA-PJ15 cells. Eur J Med Chem 2015; 102:403-12. [PMID: 26301557 DOI: 10.1016/j.ejmech.2015.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/22/2015] [Accepted: 08/08/2015] [Indexed: 12/19/2022]
Abstract
Bisphosphonates are the most important class of antiresorptive agents used against osteoclast-mediated bone loss, and, more recently, in oncology. These compounds have high affinity for calcium ions (Ca(2+)) and therefore target bone mineral, where they appear to be internalized selectively by bone-resorbing osteoclasts and inhibit osteoclast function. They are extensively used in healthcare, however they are affected by severe side effects; pharmacological properties of bisphosphonates depend on their molecular structure, which is frequently the cause of poor intestinal adsorption and low distribution. In this work we synthesized six novel bisphosphonate compounds having a variably substituted indole moiety to evaluate their extra- and intracellular calcium chelating ability in PE/CA-PJ15 cells. Preliminary in silico and in vitro ADME studies were also performed and the results suggested that the indole moiety plays an important role in cell permeability and metabolism properties.
Collapse
|
4
|
Mortazavi F, Lu J, Phan R, Lewis M, Trinidad K, Aljilani A, Pezeshkpour G, Tamanoi F. Significance of KRAS/PAK1/Crk pathway in non-small cell lung cancer oncogenesis. BMC Cancer 2015; 15:381. [PMID: 25956913 PMCID: PMC4477307 DOI: 10.1186/s12885-015-1360-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/23/2015] [Indexed: 12/11/2022] Open
Abstract
Background Key effector(s) of mutated KRAS in lung cancer progression and metastasis are unknown. Here we investigated the role of PAK1/Crk axis in transduction of the oncogenic KRAS signal in non-small cell lung cancer (NSCLC). Methods We used NSCLC clinical specimens to examine the correlation among KRAS mutations (codon 12, 13 and 61); PAK1/Crk axis activation [p-PAK1(Thr423), p-Crk(Ser41)]; and adhesion molecules expression by immunohistochemistry. For assessing the role of proto-oncogene c-Crk as a KRAS effector, we inhibited KRAS in NSCLC cells by a combination of farnesyltransferase inhibitor (FTI) and geranylgeranyltransferase inhibitor (GGTI) and measured p-Crk-II(Ser41) by western blotting. Finally, we disrupted the signaling network downstream of KRAS by blocking KRAS/PAK1/Crk axis with PAK1 inhibitors (i.e., IPA-3, FRAX597 or FRAX1036) along with partial inhibition of all other KRAS effectors by prenylation inhibitors (FTI + GGTI) and examined the motility, morphology and proliferation of the NSCLC cells. Results Immunohistochemical analysis demonstrated an inverse correlation between PAK1/Crk phosphorylation and E-cadherin/p120-catenin expression. Furthermore, KRAS mutant tumors expressed higher p-PAK1(Thr423) compared to KRAS wild type. KRAS prenylation inhibition by (FTI + GGTI) completely dephosphorylated proto-oncogene c-Crk on Serine 41 while Crk phosphorylation did not change by individual prenylation inhibitors or diluent. Combination of PAK1 inhibition and partial inhibition of all other KRAS effectors by (FTI + GGTI) dramatically altered morphology, motility and proliferation of H157 and A549 cells. Conclusions Our data provide evidence that proto-oncogene c-Crk is operative downstream of KRAS in NSCLC. Previously we demonstrated that Crk receives oncogenic signals from PAK1. These data in conjunction with the work of others that have specified the role of PAK1 in transduction of KRAS signal bring forward the importance of KRAS/PAK1/Crk axis as a prominent pathway in the oncogenesis of KRAS mutant lung cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1360-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fariborz Mortazavi
- Division of Hematology/Oncology, West Los Angeles VA, Los Angeles, CA, USA. .,Department of Medicine, University of California, Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Jie Lu
- Department of Microbiology Immunology & Molecular Genetics, University of California
- , Los Angeles, CA, USA.
| | - Ryan Phan
- Department of Pathology, West Los Angeles VA, Los Angeles, CA, USA.
| | - Michael Lewis
- Department of Pathology, West Los Angeles VA, Los Angeles, CA, USA.
| | - Kenny Trinidad
- Division of Hematology/Oncology, West Los Angeles VA, Los Angeles, CA, USA.
| | - Amir Aljilani
- Division of Hematology/Oncology, West Los Angeles VA, Los Angeles, CA, USA.
| | | | - Fuyuhiko Tamanoi
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA. .,Department of Microbiology Immunology & Molecular Genetics, University of California
- , Los Angeles, CA, USA.
| |
Collapse
|
5
|
He X, Aglio T, Deschamps JR, Rai R, Xue F. Synthesis of 1,2-dihydro-2-oxo-4-quinolinyl phosphates from 2-acyl-benzoic acids. Tetrahedron Lett 2015; 56:1441-1444. [PMID: 25937677 DOI: 10.1016/j.tetlet.2015.01.188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We report a facile synthesis of 1,2-dihydro-2-oxo-4-quinolinyl phosphates (1a-l) starting from 2-acyl-benzoic acids (2a-l) in the presence of phosphoryl azides via a one-pot cascade reaction involving a Curtius rearrangement, an intramolecular nucleophilic addition of the enol carbon to the isocyanate intermediate, and an addition-elimination of the enol oxygen to the phosphoryl azide. During the reaction three new bonds are formed under mild conditions to yield 1,2-dihydro-2-oxo-4-quinolinyl phosphates in modest yields.
Collapse
Affiliation(s)
- Xinhua He
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201
| | - Tharcilla Aglio
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201
| | - Jeffrey R Deschamps
- Naval Research Laboratory, Code 6930, 4555 Overlook Ave., Washington, DC 20375
| | - Rachita Rai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201
| |
Collapse
|
6
|
Booy S, Hofland L, van Eijck C. Potentials of interferon therapy in the treatment of pancreatic cancer. J Interferon Cytokine Res 2014; 35:327-39. [PMID: 25551196 DOI: 10.1089/jir.2014.0157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with limited treatment options. To improve survival for patients with pancreatic cancer, research has focused on other treatment modalities like adding biological modulators such as type-I interferons (IFNs). Type I IFNs (ie, IFN-α/IFN-β) have antiproliferative, antiviral, and immunoregulatory activities. Furthermore, they are able to induce apoptosis, exert cell cycle blocking, and sensitize tumor cells for chemo- and radiotherapy. A few years ago in vitro, in vivo, and several clinical trials have been described regarding adjuvant IFN-α therapy in the treatment of pancreatic cancer. Some studies reported a remarkable increase in the 2- and 5-year survival. Unfortunately, the only randomized clinical trial did not show a significant increase in overall survival, although the increased median survival implicated that some patients in the experimental group benefited from the adjuvant IFN-α therapy. Furthermore, encouraging in vitro and in vivo data points to a possible role for adjuvant IFN therapy. However, up till now, the use of IFNs in the treatment of pancreatic cancer remains controversial. This review, therefore, aims to describe, based on the available data, whether there is a distinct role for IFN therapy in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Stephanie Booy
- 1 Department of Surgery, Erasmus Medical Centre , Rotterdam, The Netherlands
| | | | | |
Collapse
|
7
|
Dicitore A, Caraglia M, Gaudenzi G, Manfredi G, Amato B, Mari D, Persani L, Arra C, Vitale G. Type I interferon-mediated pathway interacts with peroxisome proliferator activated receptor-γ (PPAR-γ): at the cross-road of pancreatic cancer cell proliferation. Biochim Biophys Acta Rev Cancer 2013; 1845:42-52. [PMID: 24295567 DOI: 10.1016/j.bbcan.2013.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/14/2013] [Accepted: 11/22/2013] [Indexed: 12/12/2022]
Abstract
Pancreatic adenocarcinoma remains an unresolved therapeutic challenge because of its intrinsically refractoriness to both chemo- and radiotherapy due to the complexity of signaling and the activation of survival pathways in cancer cells. Recent studies have demonstrated that the combination of some drugs, targeting most of aberrant pathways crucial for the survival of pancreatic cancer cells may be a valid antitumor strategy for this cancer. Type I interferons (IFNs) may have a role in the pathogenesis and progression of pancreatic adenocarcinoma, but the limit of their clinical use is due to the activation of tumor resistance mechanisms, including JAK-2/STAT-3 pathway. Moreover, aberrant constitutive activation of STAT-3 proteins has been frequently detected in pancreatic adenocarcinoma. The selective targeting of these cell survival cascades could be a promising strategy in order to enhance the antitumor effects of type I IFNs. The activation of peroxisome proliferator-activated receptor γ (PPAR-γ), on the other hand, has a suppressive activity on STAT-3. In fact, PPAR-γ agonists negatively modulate STAT-3 through direct and/or indirect mechanisms in several normal and cancer models. This review provides an overview on the current knowledge about the molecular mechanisms and antitumor activity of these two promising classes of drugs for pancreatic cancer therapy. Finally, the synergistic antiproliferative activity of combined IFN-β and troglitazone treatment on pancreatic cancer cell lines, evaluated in vitro, and the consequent potential clinical applications will be discussed.
Collapse
Affiliation(s)
- Alessandra Dicitore
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Germano Gaudenzi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Gloria Manfredi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Bruno Amato
- Department of Clinical Medicine and Surgery, University "Federico II" of Naples, Italy
| | - Daniela Mari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Geriatric Unit IRCCS Ca' Grande Foundation Maggiore Policlinico Hospital, Milan, Italy
| | - Luca Persani
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Claudio Arra
- Animal Facility, National Cancer Institute of Naples Fondazione "G. Pascale", Naples, Italy
| | - Giovanni Vitale
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| |
Collapse
|
8
|
Perna AF, Sepe I, Lanza D, Capasso R, Zappavigna S, Capasso G, Caraglia M, Ingrosso D. Hydrogen sulfide reduces cell adhesion and relevant inflammatory triggering by preventing ADAM17-dependent TNF-α activation. J Cell Biochem 2013; 114:1536-48. [DOI: 10.1002/jcb.24495] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/18/2012] [Indexed: 12/31/2022]
|
9
|
Franco R, Nicoletti G, Lombardi A, Di Domenico M, Botti G, Zito Marino F, Caraglia M. Current treatment of cutaneous squamous cancer and molecular strategies for its sensitization to new target-based drugs. Expert Opin Biol Ther 2012; 13:51-66. [PMID: 22998482 DOI: 10.1517/14712598.2012.725720] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Cutaneous squamous cell carcinoma (cSCC) is considered one of the most common skin malignancy with a relatively high risk of metastasis occurrence. AREAS COVERED We discuss the pathogenetic mechanisms of cSCC and the main therapeutic strategies available for the treatment of cSCC. EXPERT OPINION Chemotherapy and biological therapy with Interferon α (IFN-α) and cis retinoic acid are active but give limited results. Recently, strategies based on the use of molecularly target-based agents (MTA) have been used with promising results. Based on the available findings, we hypothesize that SCC cells can develop survival and resistance mechanisms to MTAs. The detection of these mechanisms could be useful in designing strategies able to overcome the latter and to potentiate the anticancer activity of MTAs. We describe the example of the EGF-dependent survival pathway elicited by IFN-α and the different strategies to abrogate this survival pathway. Other strategies to potentiate the antitumor activity of cytotoxic agents such as docetaxel or cisplatin are also discussed. Illuminating examples are the inhibition of multichaperone activity or the inactivation of the proteasome. In conclusion, a new dawn based upon the rationale use of MTAs is rising up in the treatment of advanced cSCC.
Collapse
Affiliation(s)
- Renato Franco
- Second University of Naples, Department of Biochemistry and Biophysics, Naples Italy
| | | | | | | | | | | | | |
Collapse
|
10
|
Baldi A, Piccolo MT, Boccellino MR, Donizetti A, Cardillo I, La Porta R, Quagliuolo L, Spugnini EP, Cordero F, Citro G, Menegozzo M, Calogero RA, Crispi S. Apoptosis induced by piroxicam plus cisplatin combined treatment is triggered by p21 in mesothelioma. PLoS One 2011; 6:e23569. [PMID: 21858171 PMCID: PMC3157425 DOI: 10.1371/journal.pone.0023569] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 07/20/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Malignant mesothelioma (MM) is a rare, highly aggressive tumor, associated to asbestos exposure. To date no chemotherapy regimen for MM has proven to be definitively curative, and new therapies for MM treatment need to be developed. We have previously shown in vivo that piroxicam/cisplatin combined treatment in MM, specifically acts on cell cycle regulation triggering apoptosis, with survival increase. METHODOLOGY/PRINCIPAL FINDINGS We analyzed, at molecular level, the apoptotic increase caused by piroxicam/cisplatin treatment in MM cell lines. By means of genome wide analyses, we analyzed transcriptional gene deregulation both after the single piroxicam or cisplatin and the combined treatment. Here we show that apoptotic increase following combined treatment is mediated by p21, since apoptotic increase in piroxicam/cisplatin combined treatment is abolished upon p21 silencing. CONCLUSIONS/SIGNIFICANCE Piroxicam/cisplatin combined treatment determines an apoptosis increase in MM cells, which is dependent on the p21 expression. The results provided suggest that piroxicam/cisplatin combination might be tested in clinical settings in tumor specimens that express p21.
Collapse
Affiliation(s)
- Alfonso Baldi
- Department of Biochemistry, Section of Pathology, Second University of Naples, Naples, Italy
| | - Maria Teresa Piccolo
- Gene Expression & Human Molecular Genetics Laboratory, Institute of Genetics and Biophysics, CNR, Naples, Italy
| | | | - Aldo Donizetti
- Gene Expression & Human Molecular Genetics Laboratory, Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Irene Cardillo
- S.A.F.U. Department, Regina Elena Cancer Institute, Rome, Italy
| | - Raffaele La Porta
- Department of Biochemistry, Section of Pathology, Second University of Naples, Naples, Italy
| | - Lucio Quagliuolo
- Department of Biochemistry, Section of Pathology, Second University of Naples, Naples, Italy
| | | | - Francesca Cordero
- Bioinformatics and Genomics Unit, Department of Clinical and Biological Science, University of Turin, Turin, Italy
| | - Gennaro Citro
- S.A.F.U. Department, Regina Elena Cancer Institute, Rome, Italy
| | - Massimo Menegozzo
- Campania Regional Operating Center (COR) of the National Mesothelioma Registry (ReNaM) - Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Raffaele A. Calogero
- Bioinformatics and Genomics Unit, Department of Clinical and Biological Science, University of Turin, Turin, Italy
| | - Stefania Crispi
- Gene Expression & Human Molecular Genetics Laboratory, Institute of Genetics and Biophysics, CNR, Naples, Italy
| |
Collapse
|
11
|
Epling-Burnette PK, Loughran TP. Suppression of farnesyltransferase activity in acute myeloid leukemia and myelodysplastic syndrome: current understanding and recommended use of tipifarnib. Expert Opin Investig Drugs 2010; 19:689-98. [PMID: 20402600 DOI: 10.1517/13543781003801076] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) incidence in the United States increases with age. Given the progressive ageing of the general population, incidence of these diseases is likely to continue to rise in the future. There is an acute need for therapeutic developments because of the poor prognosis of these diseases. Since the knowledge of molecular genetics in AML and MDS has expanded recently, targeted therapeutics should offer an exciting new frontier for advancement. Of all the targeted inhibitors developed, tipifarnib represents one of the few compounds with some activity as a single agent. AREAS COVERED IN THIS REVIEW Described in this review are the molecular targets of tipifarnib, safety and tolerability of the drug, chemistry, and clinical efficacy in AML. WHAT THE READER WILL GAIN The reader will gain a thorough understanding of tipifarnib as it relates to the current and future use of the drug in AML. TAKE HOME MESSAGE The future of tipifarnib, along with other molecularly-targeted drugs, lies in achieving a better understanding of leukemia biology and harnessing the activity of this agent using predictive biomarkers for improved patient selection.
Collapse
|
12
|
Combined therapies for cancer: a review of EGFR-targeted monotherapy and combination treatment with other drugs. J Cancer Res Clin Oncol 2009; 135:1137-48. [DOI: 10.1007/s00432-009-0622-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 06/02/2009] [Indexed: 12/21/2022]
|
13
|
Naviglio S, Caraglia M, Abbruzzese A, Chiosi E, Di Gesto D, Marra M, Romano M, Sorrentino A, Sorvillo L, Spina A, Illiano G. Protein kinase A as a biological target in cancer therapy. Expert Opin Ther Targets 2009; 13:83-92. [PMID: 19063708 DOI: 10.1517/14728220802602349] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND cAMP is a second messenger that plays a role in intracellular signal transduction of various stimuli. a major function of cAMP in eukaryotes is activation of cAMP-dependent protein kinase (PKA). PKA is the best understood member of the serine-threonine protein kinase superfamily, and is involved in the control of a variety of cellular processes. since it has been implicated in the initiation and progression of many tumors, PKA has been suggested as a novel molecular target for cancer therapy. OBJECTIVE/METHODS Here, after describing some features of cAMP/PKA signaling that are relevant to cancer biology, we review targeting of PKA in cancer therapy, also discussing PKA as a biomarker for cancer detection and monitoring of therapy. RESULTS/CONCLUSIONS PKA is an increasingly relevant biological target in the therapy and management of cancer.
Collapse
Affiliation(s)
- Silvio Naviglio
- Second University of Naples, Medical School, Department of Biochemistry and Biophysics, Via L. De Crecchio 7, 80138 Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bovine serum amine oxidase and spm potentiate docetaxel and interferon-α effects in inducing apoptosis on human cancer cells through the generation of oxidative stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:2269-78. [DOI: 10.1016/j.bbamcr.2008.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 08/25/2008] [Accepted: 09/05/2008] [Indexed: 11/16/2022]
|
15
|
Minville J, Poulin J, Dufresne C, Sturino CF. A general synthesis of quinolinones and benzothiazine 1,1-dioxides via ring closing metathesis. Tetrahedron Lett 2008. [DOI: 10.1016/j.tetlet.2008.03.144] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Abstract
Over the past 5 years, the Raf kinase family has emerged as a promising target for protein-directed cancer therapy development. The goal of this review is to first provide a concise summary of the data validating Raf proteins as high-interest therapeutic targets. The authors then outline the mode of action of Raf kinases, emphasizing how Raf activities and protein interactions suggest specific approaches to inhibiting Raf. The authors then summarize the set of drugs, antisense reagents and antibodies available or in development for therapeutically targeting Raf or Raf-related proteins, as well as existing strategies combining these and other therapeutic agents. Finally, the authors discuss recent results from systems biology analyses that have the potential to increasingly guide the intelligent selection of combination therapies involving Raf-targeting agents and other therapeutics.
Collapse
|