1
|
Fischer F, Doll A, Uereyener D, Roenneberg S, Hillig C, Weber L, Hackert V, Meinel M, Farnoud A, Seiringer P, Thomas J, Anand P, Graner L, Schlenker F, Zengerle R, Jonsson P, Jargosch M, Theis FJ, Schmidt-Weber CB, Biedermann T, Howell M, Reich K, Eyerich K, Menden M, Garzorz-Stark N, Lauffer F, Eyerich S. Gene expression based molecular test as diagnostic aid for the differential diagnosis of psoriasis and eczema in formalin fixed and paraffin embedded tissue, microbiopsies and tape strips. J Invest Dermatol 2023:S0022-202X(23)00156-2. [PMID: 36889660 DOI: 10.1016/j.jid.2023.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 03/08/2023]
Abstract
Highly effective targeted therapies are available to treat non-communicable chronic inflammatory skin diseases (ncISD). In contrast, the exact diagnosis of ncISD is complicated by their complex pathogenesis and clinical and histological overlap. Particularly, differential diagnosis of psoriasis and eczema can be challenging in special cases and molecular diagnostic tools need to be developed to support gold standard diagnosis. Aim of this work was to develop a real-time PCR based molecular classifier to distinguish psoriasis from eczema in FFPE-fixed skin samples and to evaluate the use of minimally invasive microbiopsies and tape strips for molecular diagnosis. Here, we present a FFPE-based molecular classifier (MC) that determines the probability for psoriasis with a sensitivity/specificity of 92%/100%, respectively, and an AUC of 0.97 delivering comparable results to our previous published RNAprotect-based MC. The psoriasis probability, as well as levels of NOS2 expression positively correlated with disease hallmarks of psoriasis and negatively with eczema hallmarks. Furthermore, minimally invasive tape strips and microbiopsies were effectively used to differentiate psoriasis from eczema. In summary, the MC offers broad usage in pathology laboratories as well as outpatient settings and can support the differential diagnosis of ncISD on a molecular level FFPE tissue, microbiopsies and tape strips.
Collapse
Affiliation(s)
- Felix Fischer
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Anais Doll
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Deniz Uereyener
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Sophie Roenneberg
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Christina Hillig
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | - Martin Meinel
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Ali Farnoud
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Peter Seiringer
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Jenny Thomas
- ZAUM-Center of Allergy and Environment, Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Philipp Anand
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | | | | | - Roland Zengerle
- Laboratory for MEMS Applications, Department of Microsystems Engineering - IMTEK, University of Freiburg, Freiburg, Germany; Hahn-Schickard, Freiburg, Germany
| | - Pontus Jonsson
- Karolinska Universitetssjukhuset - Hudkliniken Stockholm, Stockholm, Sweden
| | - Manja Jargosch
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany; ZAUM-Center of Allergy and Environment, Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany; Department of Mathematics, Technical University of Munich, Garching, Germany
| | - Carsten B Schmidt-Weber
- ZAUM-Center of Allergy and Environment, Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | | | - Kristian Reich
- Translational Research in Inflammatory Skin Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kilian Eyerich
- Klinik für Dermatologie und Venerologie, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Michael Menden
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Natalie Garzorz-Stark
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Felix Lauffer
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Stefanie Eyerich
- ZAUM-Center of Allergy and Environment, Technical University of Munich and Helmholtz Center Munich, Munich, Germany.
| |
Collapse
|
2
|
Appel LM, Benedum J, Engl M, Platzer S, Schleiffer A, Strobl X, Slade D. SPOC domain proteins in health and disease. Genes Dev 2023; 37:140-170. [PMID: 36927757 PMCID: PMC10111866 DOI: 10.1101/gad.350314.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Since it was first described >20 yr ago, the SPOC domain (Spen paralog and ortholog C-terminal domain) has been identified in many proteins all across eukaryotic species. SPOC-containing proteins regulate gene expression on various levels ranging from transcription to RNA processing, modification, export, and stability, as well as X-chromosome inactivation. Their manifold roles in controlling transcriptional output implicate them in a plethora of developmental processes, and their misregulation is often associated with cancer. Here, we provide an overview of the biophysical properties of the SPOC domain and its interaction with phosphorylated binding partners, the phylogenetic origin of SPOC domain proteins, the diverse functions of mammalian SPOC proteins and their homologs, the mechanisms by which they regulate differentiation and development, and their roles in cancer.
Collapse
Affiliation(s)
- Lisa-Marie Appel
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
| | - Johannes Benedum
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Magdalena Engl
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Sebastian Platzer
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
| | - Alexander Schleiffer
- Research Institute of Molecular Pathology (IMP), 1030 Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Xué Strobl
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Dea Slade
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria;
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
| |
Collapse
|
3
|
El Yaagoubi OM, Oularbi L, Bouyahya A, Samaki H, El Antri S, Aboudkhil S. The role of the ubiquitin-proteasome pathway in skin cancer development: 26S proteasome-activated NF-κB signal transduction. Cancer Biol Ther 2021; 22:479-492. [PMID: 34583610 DOI: 10.1080/15384047.2021.1978785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Ubiquitin-Proteasome System plays a central role in signal transduction associated with stress, in the skin in particular by the control of NF-κB pathways. Under normal conditions, the inhibitory protein IκB is phosphorylated by kinases, then ubiquitinated and ends up at the proteasome to be degraded. The present short review discusses recent progress in the inhibition of NF-κB activation by proteasome inhibitors prevents the degradation of protein IκB, which accumulates in the cytosol, and there by the activation of NF-κB. Moreover, would not only limit the expression of adhesion molecules and cytokines involved in metastatic processes, but also increase the sensitivity of cancer cells to apoptosis. Considering this fact, the activity of NF-κB is regulated by the phosphorylation and proteasome-dependent degradation of its inhibitor Iκb. In this scenario, the use of a proteasome inhibitor might be an effective strategy in the treatment of skin cancer with constitutive activation of NF-κB.
Collapse
Affiliation(s)
- Ouadie Mohamed El Yaagoubi
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36) -Faculty of Sciences and Technology -Mohammedia, Hassan II University, Casablanca, Morocco
| | - Larbi Oularbi
- Laboratory of Materials, Membranes, and Environment, Faculty of Science and Technology-Mohammedia, Hassan II University, Casablanca, Morocco.,Supramolecular Nanomaterials Group (SNG), Mohammed VI Polytechnic University, Benguerir Morocco
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.,Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Hamid Samaki
- National Institute of Social Action (INAS), Tangier, Morocco
| | - Said El Antri
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36) -Faculty of Sciences and Technology -Mohammedia, Hassan II University, Casablanca, Morocco
| | - Souad Aboudkhil
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36) -Faculty of Sciences and Technology -Mohammedia, Hassan II University, Casablanca, Morocco
| |
Collapse
|
4
|
Vesga LC, Silva AMP, Bernal CC, Mendez-Sánchez SC, Romero Bohórquez AR. Tetrahydroquinoline/4,5-dihydroisoxazole hybrids with a remarkable effect over mitochondrial bioenergetic metabolism on melanoma cell line B16F10. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02796-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
5
|
Close DA, Kirkwood JM, Fecek RJ, Storkus WJ, Johnston PA. Unbiased High-Throughput Drug Combination Pilot Screening Identifies Synergistic Drug Combinations Effective against Patient-Derived and Drug-Resistant Melanoma Cell Lines. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:712-729. [PMID: 33208016 PMCID: PMC8128935 DOI: 10.1177/2472555220970917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We describe the development, optimization, and validation of 384-well growth inhibition assays for six patient-derived melanoma cell lines (PDMCLs), three wild type (WT) for BRAF and three with V600E-BRAF mutations. We conducted a pilot drug combination (DC) high-throughput screening (HTS) of 45 pairwise 4×4 DC matrices prepared from 10 drugs in the PDMCL assays: two B-Raf inhibitors (BRAFi), a MEK inhibitor (MEKi), and a methylation agent approved for melanoma; cytotoxic topoisomerase II and DNA methyltransferase chemotherapies; and drugs targeting the base excision DNA repair enzyme APE1 (apurinic/apyrimidinic endonuclease-1/redox effector factor-1), SRC family tyrosine kinases, the heat shock protein 90 (HSP90) molecular chaperone, and histone deacetylases.Pairwise DCs between dasatinib and three drugs approved for melanoma therapy-dabrafenib, vemurafenib, or trametinib-were flagged as synergistic in PDMCLs. Exposure to fixed DC ratios of the SRC inhibitor dasatinib with the BRAFis or MEKis interacted synergistically to increase PDMCL sensitivity to growth inhibition and enhance cytotoxicity independently of PDMCL BRAF status. These DCs synergistically inhibited the growth of mouse melanoma cell lines that either were dabrafenib-sensitive or had acquired resistance to dabrafenib with cross resistance to vemurafenib, trametinib, and dasatinib. Dasatinib DCs with dabrafenib, vemurafenib, or trametinib activated apoptosis and increased cell death in melanoma cells independently of their BRAF status or their drug resistance phenotypes. These preclinical in vitro studies provide a data-driven rationale for the further investigation of DCs between dasatinib and BRAFis or MEKis as candidates for melanoma combination therapies with the potential to improve outcomes and/or prevent or delay the emergence of disease resistance.
Collapse
Affiliation(s)
- David A. Close
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John M. Kirkwood
- Departments of Medicine, Dermatology, Translational Science, and Melanoma and Skin Cancer Program University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ronald J. Fecek
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA 15601, USA
| | - Walter J. Storkus
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Departments of Dermatology, Immunology, Bioengineering and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Paul A. Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
6
|
Leveraging multi-way interactions for systematic prediction of pre-clinical drug combination effects. Nat Commun 2020; 11:6136. [PMID: 33262326 PMCID: PMC7708835 DOI: 10.1038/s41467-020-19950-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
We present comboFM, a machine learning framework for predicting the responses of drug combinations in pre-clinical studies, such as those based on cell lines or patient-derived cells. comboFM models the cell context-specific drug interactions through higher-order tensors, and efficiently learns latent factors of the tensor using powerful factorization machines. The approach enables comboFM to leverage information from previous experiments performed on similar drugs and cells when predicting responses of new combinations in so far untested cells; thereby, it achieves highly accurate predictions despite sparsely populated data tensors. We demonstrate high predictive performance of comboFM in various prediction scenarios using data from cancer cell line pharmacogenomic screens. Subsequent experimental validation of a set of previously untested drug combinations further supports the practical and robust applicability of comboFM. For instance, we confirm a novel synergy between anaplastic lymphoma kinase (ALK) inhibitor crizotinib and proteasome inhibitor bortezomib in lymphoma cells. Overall, our results demonstrate that comboFM provides an effective means for systematic pre-screening of drug combinations to support precision oncology applications. Combinatorial treatments have become a standard of care for various complex diseases including cancers. Here, the authors show that combinatorial responses of two anticancer drugs can be accurately predicted using factorization machines trained on large-scale pharmacogenomic data for guiding precision oncology studies.
Collapse
|
7
|
Resistance of melanoma cells to anticancer treatment: a role of vascular endothelial growth factor. Postepy Dermatol Alergol 2020; 37:11-18. [PMID: 32467677 PMCID: PMC7247075 DOI: 10.5114/ada.2020.93378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Melanoma is one of the most aggressive and resistant to treatment neoplasms. There are still many challenges despite many promising advances in anticancer treatment. Currently, the main problem for all types of treatment is associated with heterogeneity. Due to heterogeneity of cancer cells, "precise" targeting of a medicine against a single phenotype limits the efficacy of treatment and affects resistance to applied therapy. Therefore it is important to understand aetiology and reasons for heterogeneity in order to develop effective and long-lasting treatment. This review summarises roles of vascular endothelial growth factor (VEGF) that may stimulate growth of a melanoma tumour irrespective of its proangiogenic effects, contributing to cancer heterogeneity. VEGF triggers processes associated with extracellular matrix remodelling, cell migration, invasion, angiogenesis, inhibition of immune responses and favours phenotypic plasticity and epithelial-mesenchymal transition. Consequently, it participates in mechanisms of interactions between melanoma cancer cells and microenvironment and it can modify sensitivity to therapeutic factors.
Collapse
|
8
|
Brugnara S, Sicher M, Bonandini EM, Barbareschi M, Girardelli CR, Caffo O. Onset of vitiligo following targeted therapy for BRAF V600E-mutated melanoma: case report. Drugs Context 2019; 8:212582. [PMID: 32158480 PMCID: PMC7048152 DOI: 10.7573/dic.212582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/15/2019] [Accepted: 11/21/2019] [Indexed: 02/08/2023] Open
Abstract
Systemic treatment for metastatic melanoma has advanced dramatically in recent years with an impressive increase in the rate of overall survival. The two main different strategies are targeted therapies (i.e. BRAF and MEK inhibitors) and immunotherapy with monoclonal antibodies against the immune checkpoint proteins programmed death-1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4). Vitiligo often accompanies immunotherapy in melanoma patients and even correlates with tumor regression after checkpoint blockade. At present, a correlation between vitiligo onset and outcome from immunotherapy is acknowledged; however, evidence of a correlation between vitiligo and efficacy of combination-targeted therapy is lacking. We describe our experience in a patient who received dabrafenib and trametinib and developed vitiligo-like depigmentation after treatment cessation.
Collapse
Affiliation(s)
| | | | | | | | | | - Orazio Caffo
- Oncology Unit, S. Chiara Hospital, Trento, Italy
| |
Collapse
|
9
|
Negative regulators of cell death pathways in cancer: perspective on biomarkers and targeted therapies. Apoptosis 2019; 23:93-112. [PMID: 29322476 DOI: 10.1007/s10495-018-1440-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cancer is a primary cause of human fatality and conventional cancer therapies, e.g., chemotherapy, are often associated with adverse side-effects, tumor drug-resistance, and recurrence. Molecularly targeted therapy, composed of small-molecule inhibitors and immunotherapy (e.g., monoclonal antibody and cancer vaccines), is a less harmful alternative being more effective against cancer cells whilst preserving healthy tissues. Drug-resistance, however, caused by negative regulation of cell death signaling pathways, is still a challenge. Circumvention of negative regulators of cell death pathways or development of predictive and response biomarkers is, therefore, quintessential. This review critically discusses the current state of knowledge on targeting negative regulators of cell death signaling pathways including apoptosis, ferroptosis, necroptosis, autophagy, and anoikis and evaluates the recent advances in clinical and preclinical research on biomarkers of negative regulators. It aims to provide a comprehensive platform for designing efficacious polytherapies including novel agents for restoring cell death signaling pathways or targeting alternative resistance pathways to improve the chances for antitumor responses. Overall, it is concluded that nonapoptotic cell death pathways are a potential research arena for drug discovery, development of novel biomarkers and targeted therapies.
Collapse
|
10
|
Ciołczyk-Wierzbicka D, Zarzycka M, Gil D, Laidler P. mTOR inhibitor Everolimus-induced apoptosis in melanoma cells. J Cell Commun Signal 2019; 13:357-368. [PMID: 30848427 PMCID: PMC6732148 DOI: 10.1007/s12079-019-00510-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/22/2019] [Indexed: 12/29/2022] Open
Abstract
Melanoma is the most aggressive, therapy-resistant skin cancer. The mammalian target of rapamycin (mTOR), the serine/threonine kinase which integrates both intracellular and extracellular signals, plays a crucial role in coordinating the balance between the growth and death of cells. The object of this study is a comparison of the influence of mTOR inhibitor everolimus in the concentration range between 20 nM and 10 μM, used individually and in combination with selected downstream protein kinases inhibitors: LY294002 (PI3K), U0126 (ERK1/2), AS-703026 (MEK) and MK-2206 (AKT) on the expression of pro-survival proteins: p-Bcl-2 (S70), p-Bcl-2 (T56), Bcl-2, Bcl-xL, Mcl-1, activity of caspase-3, proliferation and induction of apoptosis in melanoma cells. Current results clearly show that the nanomolar concentration of the mTOR inhibitor everolimus in combination with the inhibitor of MAP kinase (AS-703026) or AKT kinase (MK-2206) is effective in inducing apoptosis and reducing proliferation of melanoma cells. The herein research results confirm the hypothesis on the important role of mTOR signaling in cancer progression, and gives hope that implementation of successful combination of its inhibitors will find recognition and application in cancer treatment in the near future.
Collapse
Affiliation(s)
| | - Marta Zarzycka
- Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034, Kraków, Poland
| | - Dorota Gil
- Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034, Kraków, Poland
| | - Piotr Laidler
- Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034, Kraków, Poland
| |
Collapse
|
11
|
Resnier P, Lepeltier E, Emina AL, Galopin N, Bejaud J, David S, Ballet C, Benvegnu T, Pecorari F, Chourpa I, Benoit JP, Passirani C. Model Affitin and PEG modifications onto siRNA lipid nanocapsules: cell uptake and in vivo biodistribution improvements. RSC Adv 2019; 9:27264-27278. [PMID: 35529231 PMCID: PMC9070605 DOI: 10.1039/c9ra03668g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/06/2019] [Indexed: 01/16/2023] Open
Abstract
Malignant melanoma is an aggressive tumor, associated with the presence of local and/or distant metastases. The development of gene therapy by the use of small interfering RNA (siRNA) represents a promising new treatment. However, the protection of this biomolecule is necessary in order for it to be intravenously administrated, for example via its incorporation into nanomedicines. In parallel to the passive targeting usually obtained by pegylation, various studies have aimed at developing “smart” nanomedicines to efficiently deliver the drug to tumor sites. In this work, siRNA loaded lipid nanocapsules (LNCs) were modified with DSPE-polyethylene glycol (DSPE-PEG), tetraether-PEG (TE-PEG) and/or with an Affitin model, to assay multiple targeting strategies. The uptake of fluorescently labelled LNCs, nanocarrier integrity and siRNA release into human SK-Mel28 melanoma cells were studied by flow cytometry, conventional confocal microscopy and by confocal spectral imaging in a Förster Resonance Energy Transfer (FRET) mode. Surface modified siRNA LNCs were followed after human plasma incubation and after intravenous injection, in order to compare the stealth properties. Finally, the biodistribution of the different siRNA LNCs in healthy and melanoma tumor bearing mice models was assessed by in vivo biofluorescence imaging (BFI), to evaluate the potential tumor targeting ability. The post-insertion of DSPE-PEG induced a strong decrease of the internalization into melanoma cells compared to TE-PEG modification. Both PEG polymer decorations induced a great plasma protection of siRNA but only DSPE-PEG led to stealth properties, even at low concentration (5 mM). The Affitin grafting by thiolation of DSPE-PEG was validated on siRNA LNCs. DSPE-PEG-Affitin LNCs were not detected in this melanoma tumor model but did not show unspecific accumulation in organs. DSPE-PEG and TE-PEG LNCs induced a significant intratumoral accumulation of modified LNCs. Surface modifications of siRNA LNCs were assessed with innovative TE-PEG polymers and an Affitin model, in comparison to classic DSPE-PEG LNCs, in order to evaluate the potential tumor targeting of siRNA after intravenous administration.![]()
Collapse
Affiliation(s)
- Pauline Resnier
- MINT
- UNIV Angers
- INSERM 1066
- CNRS 6021
- Université Bretagne Loire
| | - Elise Lepeltier
- MINT
- UNIV Angers
- INSERM 1066
- CNRS 6021
- Université Bretagne Loire
| | | | | | - Jérôme Bejaud
- MINT
- UNIV Angers
- INSERM 1066
- CNRS 6021
- Université Bretagne Loire
| | - Stephanie David
- EA6295 – Nanomédicaments et Nanosondes
- Université François-Rabelais de Tours
- UFR de Pharmacie
- F-37200 Tours
- France
| | - Caroline Ballet
- Univ Rennes
- Ecole Nationale Supérieure de Chimie de Rennes
- CNRS
- ISCR-UMR 6226
- F-35000 Rennes
| | - Thierry Benvegnu
- Univ Rennes
- Ecole Nationale Supérieure de Chimie de Rennes
- CNRS
- ISCR-UMR 6226
- F-35000 Rennes
| | | | - Igor Chourpa
- EA6295 – Nanomédicaments et Nanosondes
- Université François-Rabelais de Tours
- UFR de Pharmacie
- F-37200 Tours
- France
| | | | | |
Collapse
|
12
|
Sidor-Kaczmarek J, Cichorek M, Spodnik JH, Wójcik S, Moryś J. Proteasome inhibitors against amelanotic melanoma. Cell Biol Toxicol 2017; 33:557-573. [PMID: 28281027 PMCID: PMC5658467 DOI: 10.1007/s10565-017-9390-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/15/2017] [Indexed: 12/11/2022]
Abstract
The incidence of malignant melanoma, the most aggressive skin cancer, is increasing constantly. Despite new targeted therapies, the prognosis for patients with metastatic disease remains poor. Thus, there is a need for new combinational treatments, and antineoplastic agents potentially valuable in this approach are inhibitors of the ubiquitin-proteasome system (UPS). In this work, we analyze the cytotoxicity mechanisms of proteasome inhibitors (MG-132, epoxomicin, and lactacystin) in a specific form of melanoma which does not synthesize melanin-the amelanotic melanoma (Ab cells). We found that the most cytotoxic of the compounds tested was epoxomicin. Caspase-9 activation as well as cytochrome C and AIF release from mitochondria indicated that exposure to epoxomicin induced the mitochondrial pathway of apoptosis. Epoxomicin treatment also resulted in accumulation of Bcl-2 family members-proapoptotic Noxa and antiapoptotic Mcl-1, which were postulated as the targets for bortezomib in melanoma. Inhibition of caspases by BAF revealed that cell death was partially caspase-independent. We observed no cell cycle arrest preceding the apoptosis of Ab cells, even though cdk inhibitors p21Cip1/Waf1 and p27Kip1 were up-regulated. The cell cycle was blocked only after inactivation of caspases by the pan-caspase inhibitor BAF. In summary, this is the first study exploring molecular mechanisms of cell death induced by epoxomicin in melanoma. We found that Ab cells died on the mitochondrial pathway of apoptosis and also partially by the caspase-independent way of death. Apoptosis induction was fast and efficient and was not preceded by cell cycle arrest.
Collapse
Affiliation(s)
| | | | - Jan Henryk Spodnik
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| | - Sławomir Wójcik
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
13
|
Discovery of novel anti-angiogenesis agents. Part 7: Multitarget inhibitors of VEGFR-2, TIE-2 and EphB4. Eur J Med Chem 2017; 141:506-518. [PMID: 29102175 DOI: 10.1016/j.ejmech.2017.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/03/2017] [Accepted: 10/10/2017] [Indexed: 11/20/2022]
Abstract
Herein, we embarked on a structural optimization campaign aiming at the discovery of second generation anti-angiogenesis agents with our previously reported BPS-7 as lead compound. A library of 27 compounds has been afforded based on the highly conserved ATP-binding pocket of VEGFR-2, Tie-2, and EphB4. Several title compounds exhibited simultaneous inhibitory effects against three angiogenic RTKs. These compounds with a 'triplet' inhibition profile have been identified as novel anti-angiogenic and anticancer agents. The representative VDAU11 displayed prominent anti-angiogenic and anticancer potency and could be considered as a candidate for further optimization. These results indicate that N-(pyridin-2-yl)acrylamide could serve as a novel hinge-binding group of triple inhibitors.
Collapse
|
14
|
Menicacci B, Laurenzana A, Chillà A, Margheri F, Peppicelli S, Tanganelli E, Fibbi G, Giovannelli L, Del Rosso M, Mocali A. Chronic Resveratrol Treatment Inhibits MRC5 Fibroblast SASP-Related Protumoral Effects on Melanoma Cells. J Gerontol A Biol Sci Med Sci 2017; 72:1187-1195. [PMID: 28329136 DOI: 10.1093/gerona/glw336] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence is related to organismal aging and is observed after DNA damaging cancer therapies, that induce tumor-suppressive modifications, but it is characterized by a strong increase in secreted factors, termed the "senescence-associated secretory phenotype" (SASP). Particularly, SASP from stroma senescent fibroblasts creates a cancer-favoring microenvironment, providing targets for anti-cancer interventions. In the present article, chronic treatment (5 weeks) with 5 µM resveratrol has been used to modulate senescence-related protumoral features of MRC5 fibroblasts, reducing SASP-related interleukins IL1α, IL1β, IL6, and IL8; transforming-growth-factor-β (TGFβ); matrix metallo-proteinases MMP3 and MMP2; urokinase plasminogen activator (uPA); receptor proteins uPAR, IL6R, insulin growth factor receptor-1 (IGF-1R), TGFβ-R2, and CXCR4. The cellular nuclear-factor-kB (NF-kB) protein level was also reduced, confirming its role in the induction of SASP. Resveratrol pretreated MRC5 fibroblasts were resistant to activation by TGFβ. Resveratrol treatment of senescent MRC5 induced the production of conditioned media (CM) which counteracted the protumoral effect of senescent CM on A375 and A375-M6 melanoma cell proliferation and invasiveness, and reduced the expression of epithelial-to-mesenchymal transition markers related to malignant features. This experimental approach proposes a treatment that targets the senescent stromal cell phenotype to induce an anti-tumor hosting microenvironment, which is suitable for both preventive and therapeutic purposes.
Collapse
Affiliation(s)
- Beatrice Menicacci
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy.,Department of Medical Biotechnologies, University of Siena, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Elisabetta Tanganelli
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Lisa Giovannelli
- Department NeuroFarBa, Section of Pharmacology and Toxicology, University of Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| | - Alessandra Mocali
- Department of Experimental and Clinical Biomedical Science, Section of Experimental Pathology and Oncology, University of Florence, Italy
| |
Collapse
|
15
|
Calero R, Morchon E, Martinez-Argudo I, Serrano R. Synergistic anti-tumor effect of 17AAG with the PI3K/mTOR inhibitor NVP-BEZ235 on human melanoma. Cancer Lett 2017; 406:1-11. [PMID: 28774796 DOI: 10.1016/j.canlet.2017.07.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/07/2017] [Accepted: 07/22/2017] [Indexed: 11/17/2022]
Abstract
Drug resistance by MAPK signaling recovery or activation of alternative signaling pathways, such as PI3K/AKT/mTOR, is an important factor that limits the long-term efficacy of targeted therapies in melanoma patients. In the present study, we investigated the phospho-proteomic profile of RTKs and its correlation with downstream signaling pathways in human melanoma. We found that tyrosine kinase receptors expression correlated with the expression of pivotal downstream components of the RAS/RAF/MAPK and PI3K/AKT/mTOR pathways in melanoma cell lines and tumors. We also found high expression of HSP90 and the PI3K/AKT/mTOR pathway proteins, 4EBP1 and AKT compared with healthy tissue and this correlated with poor overall survival of melanoma patients. The combination of the HSP90 inhibitor 17AAG with the PI3K/mTOR inhibitor NVP-BEZ235 showed a synergistic activity decreasing melanoma cell growth, inducing apoptosis and targeting simultaneously the MAPK and PI3K/AKT/mTOR pathways. These results demonstrate that the combination of HSP90 and PI3K/mTOR inhibitors could be an effective therapeutic strategy that target the main survival pathways in melanoma and must be considered to overcome resistance to BRAF inhibitors in melanoma patients.
Collapse
Affiliation(s)
- R Calero
- Biochemistry Section, Faculty of Biochemistry and Environmental Sciences, University of Castilla-La Mancha, Toledo, Spain
| | - E Morchon
- Albacete University Hospital, Albacete, Spain
| | - I Martinez-Argudo
- Genetics Section, Faculty of Biochemistry and Environmental Sciences, University of Castilla-La Mancha, Toledo, Spain
| | - R Serrano
- Biochemistry Section, Faculty of Biochemistry and Environmental Sciences, University of Castilla-La Mancha, Toledo, Spain.
| |
Collapse
|
16
|
Computed determination of the in vitro optimal chemocombinations of sphaeropsidin A with chemotherapeutic agents to combat melanomas. Cancer Chemother Pharmacol 2017; 79:971-983. [DOI: 10.1007/s00280-017-3293-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/24/2017] [Indexed: 12/19/2022]
|
17
|
Guarneri C, Bevelacqua V, Polesel J, Falzone L, Cannavò PS, Spandidos DA, Malaponte G, Libra M. NF‑κB inhibition is associated with OPN/MMP‑9 downregulation in cutaneous melanoma. Oncol Rep 2017; 37:737-746. [PMID: 28075446 PMCID: PMC5355753 DOI: 10.3892/or.2017.5362] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/20/2016] [Indexed: 01/21/2023] Open
Abstract
The development of cutaneous melanoma is influenced by genetic factors, including BRAF mutations and environmental factors, such as ultraviolet exposure. Its progression has been also associated with the involvement of several tumour microenvironmental molecules. Among these, nuclear factor‑κB (NF‑κB) has been indicated as a key player of osteopontin (OPN) and matrix metalloproteinase‑9 (MMP‑9) activation. However, whether NF‑κB plays a role in the development and progression of melanoma in association with the OPN/MMP‑9 axis according to the BRAFV600E mutation status has not been investigated in detail to date. Thus, in the present study, in order to shed light on this matter, 148 patients with melanoma and 53 healthy donors were recruited for the analysis of OPN, MMP‑9 and NF‑κB. Significantly higher circulating levels of OPN and MMP‑9 were observed in the patients with melanoma when compared to the healthy donors. Similar data were obtained for NF‑κB p65 activity. The OPN levels did not differ significantly between melanomas with or without BRAFV600E mutation. However, as regards NF‑κB and MMP‑9, significant differences were observed between the melanomas with or without BRAFV600E mutation. To determine whether NF‑κB inhibition is associated with a decrease in the levels of OPN and MMP‑9, peripheral blood mononuclear cells from 29 patients with melanoma were treated with the NF‑κB inhibitor, dehydroxymethylepoxyquinomycin (DHMEQ), with or without OPN. As expected, the inhibition of NF‑κB induced a marked decrease in both the OPN and MMP‑9 levels. Furthermore, the decrease in MMP‑9 levels was higher among melanomas harbouring the BRAFV600E mutation. Overall, our data suggest that the activation of MMP‑9 is associated with the BRAFV600E mutation status. Furthermore, such an activation is mediated by NF‑κB, suggesting its role as therapeutic target in patients with melanoma.
Collapse
Affiliation(s)
- Claudio Guarneri
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, I-98125 Messina, Italy
| | - Valentina Bevelacqua
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, CRO Aviano National Cancer Institute, IRCCS, I-33081 Aviano, Italy
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| | - Patrizia S. Cannavò
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, I-98125 Messina, Italy
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71003, Greece
| | - Grazia Malaponte
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| |
Collapse
|
18
|
Resnier P, Galopin N, Sibiril Y, Clavreul A, Cayon J, Briganti A, Legras P, Vessières A, Montier T, Jaouen G, Benoit JP, Passirani C. Efficient ferrocifen anticancer drug and Bcl-2 gene therapy using lipid nanocapsules on human melanoma xenograft in mouse. Pharmacol Res 2017; 126:54-65. [PMID: 28159700 DOI: 10.1016/j.phrs.2017.01.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 12/22/2022]
Abstract
Metastatic melanoma has been described as a highly aggressive cancer with low sensibility to chemotherapeutic agents. New types of drug, such as metal-based drugs (ferrocifens) have emerged and could represent an alternative for melanoma treatment since they show interesting anticancer potential. Furthermore, molecular analysis has evidenced the role of apoptosis in the low sensibility of melanomas and especially of the key regulator, Bcl-2. The objective of this study was to combine two strategies in the same lipid nanocapsules (LNCs): i) gene therapy to modulate anti-apoptotic proteins by the use of Bcl-2 siRNA, and ii) ferrocifens as a new type of anticancer agent. The efficient gene silencing with LNCs was verified by the specific extinction of Bcl-2 in melanoma cells. The cellular toxicity of ferrocifens (ferrociphenol (FcDiOH) or Ansa-FcDiOH) was demonstrated, showing higher efficacy than dacarbazine. Interestingly, the association of siBcl-2 LNCs with Ansa-FcDiOH demonstrated a significant effect on melanoma cell viability. Moreover, the co-encapsulation of siRNA and ferrocifens was successfully performed into LNCs for animal experiments. A reduction of tumor volume and mass was proved after siBcl-2 LNC treatment and Ansa-FcDiOH LNC treatment, individually (around 25%). Finally, the association of both components into the same LNCs increased the reduction of tumor volume to about 50% compared to the control group. In conclusion, LNCs appeared to provide a promising tool for the co-encapsulation of a metal-based drug and siRNA.
Collapse
Affiliation(s)
- Pauline Resnier
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| | - Natacha Galopin
- SCAHU - Faculté de Médecine, Pavillon Ollivier, rue Haute de Reculée, F-49933 Angers, France.
| | - Yann Sibiril
- INSERM U1078 - Equipe 'Transfert de gènes et thérapie génique', Faculté de Médecine, 22 avenue Camille Desmoulins, CS 93837, F-29238 Brest, Cedex 3, France; CHRU de Brest, Service de Génétique Moléculaire et d'histocompatibilité, 5 avenue Maréchal Foch, 29609 Brest, France.
| | - Anne Clavreul
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| | - Jérôme Cayon
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France; PACeM (Plateforme d'Analyse Cellulaire et Moléculaire), SFR ICAT 4208, Université d'Angers, 4 rue Larrey, F-49933 Angers, France.
| | - Alessandro Briganti
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| | - Pierre Legras
- SCAHU - Faculté de Médecine, Pavillon Ollivier, rue Haute de Reculée, F-49933 Angers, France.
| | - Anne Vessières
- CNRS, UMR 8232, ENSCP, 11 rue P. et M. Curie, F-75231 Paris Cedex05, France.
| | - Tristan Montier
- INSERM U1078 - Equipe 'Transfert de gènes et thérapie génique', Faculté de Médecine, 22 avenue Camille Desmoulins, CS 93837, F-29238 Brest, Cedex 3, France; CHRU de Brest, Service de Génétique Moléculaire et d'histocompatibilité, 5 avenue Maréchal Foch, 29609 Brest, France.
| | - Gérard Jaouen
- CNRS, UMR 8232, ENSCP, 11 rue P. et M. Curie, F-75231 Paris Cedex05, France.
| | - Jean-Pierre Benoit
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| | - Catherine Passirani
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| |
Collapse
|
19
|
Kwong A, Sanlorenzo M, Rappersberger K, Vujic I. Update on advanced melanoma treatments: small molecule targeted therapy, immunotherapy, and future combination therapies. Wien Med Wochenschr 2017; 169:314-322. [DOI: 10.1007/s10354-016-0535-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/12/2016] [Indexed: 12/15/2022]
|
20
|
Garzorz-Stark N, Krause L, Lauffer F, Atenhan A, Thomas J, Stark SP, Franz R, Weidinger S, Balato A, Mueller NS, Theis FJ, Ring J, Schmidt-Weber CB, Biedermann T, Eyerich S, Eyerich K. A novel molecular disease classifier for psoriasis and eczema. Exp Dermatol 2016; 25:767-74. [DOI: 10.1111/exd.13077] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Natalie Garzorz-Stark
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
| | - Linda Krause
- Institute of Computational Biology; Helmholtz Center Munich; Neuherberg Germany
| | - Felix Lauffer
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
| | - Anne Atenhan
- ZAUM - Center of Allergy and Environment; Technical University of Munich and Helmholtz Center Munich; Member of the German Center for Lung Research (DZL); Munich Germany
| | - Jenny Thomas
- ZAUM - Center of Allergy and Environment; Technical University of Munich and Helmholtz Center Munich; Member of the German Center for Lung Research (DZL); Munich Germany
| | - Sebastian P. Stark
- ZAUM - Center of Allergy and Environment; Technical University of Munich and Helmholtz Center Munich; Member of the German Center for Lung Research (DZL); Munich Germany
| | - Regina Franz
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
| | - Stephan Weidinger
- Department of Dermatology, Allergology, and Venereology; University Hospital Schleswig-Holstein; Kiel Germany
| | - Anna Balato
- Dipartimento di Scienze biomediche avanzate; Università degli Studi di Napoli Federico II; Naples Italy
| | - Nikola S. Mueller
- Institute of Computational Biology; Helmholtz Center Munich; Neuherberg Germany
| | - Fabian J. Theis
- Institute of Computational Biology; Helmholtz Center Munich; Neuherberg Germany
- Department of Mathematics; Technical University of Munich; Garching Germany
| | - Johannes Ring
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
| | - Carsten B. Schmidt-Weber
- ZAUM - Center of Allergy and Environment; Technical University of Munich and Helmholtz Center Munich; Member of the German Center for Lung Research (DZL); Munich Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
| | - Stefanie Eyerich
- ZAUM - Center of Allergy and Environment; Technical University of Munich and Helmholtz Center Munich; Member of the German Center for Lung Research (DZL); Munich Germany
| | - Kilian Eyerich
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
| |
Collapse
|
21
|
Wei BR, Michael HT, Halsey CHC, Peer CJ, Adhikari A, Dwyer JE, Hoover SB, El Meskini R, Kozlov S, Weaver Ohler Z, Figg WD, Merlino G, Simpson RM. Synergistic targeted inhibition of MEK and dual PI3K/mTOR diminishes viability and inhibits tumor growth of canine melanoma underscoring its utility as a preclinical model for human mucosal melanoma. Pigment Cell Melanoma Res 2016; 29:643-655. [PMID: 27463366 PMCID: PMC5132162 DOI: 10.1111/pcmr.12512] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/17/2016] [Indexed: 12/12/2022]
Abstract
Human mucosal melanoma (MM), an uncommon, aggressive and diverse subtype, shares characteristics with spontaneous MM in dogs. Although BRAF and N-RAS mutations are uncommon in MM in both species, the majority of human and canine MM evaluated exhibited RAS/ERK and/or PI3K/mTOR signaling pathway activation. Canine MM cell lines, with varying ERK and AKT/mTOR activation levels reflective of naturally occurring differences in dogs, were sensitive to the MEK inhibitor GSK1120212 and dual PI3K/mTOR inhibitor NVP-BEZ235. The two-drug combination synergistically decreased cell survival in association with caspase 3/7 activation, as well as altered expression of cell cycle regulatory proteins and Bcl-2 family proteins. In combination, the two drugs targeted their respective signaling pathways, potentiating reduction of pathway mediators p-ERK, p-AKT, p-S6, and 4E-BP1 in vitro, and in association with significantly inhibited solid tumor growth in MM xenografts in mice. These findings provide evidence of synergistic therapeutic efficacy when simultaneously targeting multiple mediators in melanoma with Ras/ERK and PI3K/mTOR pathway activation.
Collapse
Affiliation(s)
- Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.,Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Helen T Michael
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Charles H C Halsey
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Cody J Peer
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Amit Adhikari
- Leidos Biomedical Research, Inc., Frederick, MD, USA.,Frederick National Laboratory for Cancer Research, Center for Advanced Preclinical Research, Frederick, MD, USA
| | - Jennifer E Dwyer
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shelley B Hoover
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Rajaa El Meskini
- Leidos Biomedical Research, Inc., Frederick, MD, USA.,Frederick National Laboratory for Cancer Research, Center for Advanced Preclinical Research, Frederick, MD, USA
| | - Serguei Kozlov
- Leidos Biomedical Research, Inc., Frederick, MD, USA.,Frederick National Laboratory for Cancer Research, Center for Advanced Preclinical Research, Frederick, MD, USA
| | - Zoe Weaver Ohler
- Leidos Biomedical Research, Inc., Frederick, MD, USA.,Frederick National Laboratory for Cancer Research, Center for Advanced Preclinical Research, Frederick, MD, USA
| | - William D Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
22
|
Shi J, Guo B, Zhang Y, Hui Q, Chang P, Tao K. Guanine nucleotide exchange factor H1 can be a new biomarker of melanoma. Biologics 2016; 10:89-98. [PMID: 27462139 PMCID: PMC4939981 DOI: 10.2147/btt.s109643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Guanine nucleotide exchange factor H1 (GEF-H1), which couples microtubule dynamics to RhoA activation, is a microtubule-regulated exchange factor. Studies have shown that GEF-H1 can be involved in various cancer pathways; however, the clinical significance of GEF-H1 expression and functions in melanoma has not been established. In this study, we investigated the relationship between clinical outcomes and GEF-H1 functions in melanoma. A total of 60 cases of different grades of melanoma samples were used to detect the expression of GEF-H1. Results showed that both messenger RNA and protein levels of GEF-H1 were significantly higher in high-grade melanomas. Furthermore, patients with high GEF-H1 expression had a shorter overall survival (22 months) than patients with low level of GEF-H1 expression (33.38 months). We also found that GEF-H1 can promote the proliferation and metastasis of melanoma cells. In summary, these results suggested that GEF-H1 may be a valuable biomarker for assessing the degree and prognosis of melanoma following surgery.
Collapse
Affiliation(s)
- Jie Shi
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, People’s Republic of China
| | - Bingyu Guo
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, People’s Republic of China
| | - Yu Zhang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, People’s Republic of China
| | - Qiang Hui
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, People’s Republic of China
| | - Peng Chang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, People’s Republic of China
| | - Kai Tao
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, People’s Republic of China
| |
Collapse
|
23
|
Guo B, Zhang Y, Hui Q, Wang H, Tao K. Naringin suppresses the metabolism of A375 cells by inhibiting the phosphorylation of c-Src. Tumour Biol 2016; 37:3841-50. [PMID: 26476533 DOI: 10.1007/s13277-015-4235-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/12/2015] [Indexed: 12/27/2022] Open
Abstract
Elevation of glycolysis, increase in lactic acid production, and enhancement of mitochondrial biogenesis are all the changes of energy metabolism of melanoma cells. Melanoma cells' metabolism and energy production networks play an important role in cancer proliferation, survival, motility, invasiveness, metastasis, and angiogenesis. Since the Warburg theory was put forward in the 1930s, more researchers focus on finding new ways for effectively eliminating cancer cells by targeting their energy metabolism. In this study, we found naringin has the inhibitory effects on the glucose metabolism of A375 cells, a melanoma cell line, in a concentration-dependent manner. We also found that naringin could significantly reduce the phosphorylation of c-Src. In summary, we demonstrated that naringin inhibits the malignant phenotype of A375 cells by suppressing c-Src and its downstream signaling pathway. More importantly, we provide the novel mechanism that, as a natural inhibitor of c-Src, naringin could be an effective candidate for the treatment of melanoma.
Collapse
Affiliation(s)
- Bingyu Guo
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, 83#Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yu Zhang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, 83#Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, People's Republic of China
| | - Qiang Hui
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, 83#Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, People's Republic of China
| | - Hongyi Wang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, 83#Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, People's Republic of China
| | - Kai Tao
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, 83#Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, People's Republic of China.
| |
Collapse
|
24
|
The protein kinase IKKepsilon contributes to tumour growth and tumour pain in a melanoma model. Biochem Pharmacol 2016; 103:64-73. [PMID: 26793999 DOI: 10.1016/j.bcp.2015.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/22/2015] [Indexed: 11/24/2022]
Abstract
Inhibitor-kappaB kinase epsilon (IKKε) constitutes a non-canonical I-κB kinase, which amongst others modulates NF-κB activity. IKKε and NF-κB have both been described for their role in cell proliferation and their dysregulation has been associated with tumourigenesis and metastasis in multiple cancer types. Accordingly, overexpression and constitutive activation of NF-κB have also been shown in melanoma, however, the role of IKKε in this cancer type has not been investigated so far. Thus, we determined IKKε expression in malignant melanoma cells and we were able to show a significant overexpression of IKKε in tumour cells in comparison to melanocytes. Inhibition of IKKε either by shRNA or the pharmacological inhibitor amlexanox resulted in reduced cell proliferation associated with a cell cycle block in the G1-phase. Functional analysis indicated that NF-κB, Akt1 and MAPK pathways might be involved in the IKKε-mediated effects. In vivo, we applied a mouse melanoma skin cancer model to assess tumour growth and melanoma-associated pain in IKKε knockout mice as well as C57BL/6 mice after inoculation with IKKε-negative cells. In IKKε knockout mice, tumour growth was not altered as compared to IKKε wild type mice. However, melanoma associated pain was strongly suppressed accompanied by a reduced mRNA expression of a number of pain-relevant genes. In contrast, after inoculation of IKKε-depleted tumour cells, the development of melanoma was almost completely prevented. In conclusion, our data suggest that IKKε in the tumour plays an essential role in tumour initiation and progression while IKKε expression in tumour surrounding tissues contributes to melanoma-associated pain.
Collapse
|
25
|
Association of CDK4 germline and BRAF somatic mutations in a patient with multiple primary melanomas and BRAF inhibitor resistance. Melanoma Res 2015; 25:443-6. [DOI: 10.1097/cmr.0000000000000173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
26
|
|
27
|
Gallagher SJ, Tiffen JC, Hersey P. Histone Modifications, Modifiers and Readers in Melanoma Resistance to Targeted and Immune Therapy. Cancers (Basel) 2015; 7:1959-82. [PMID: 26426052 PMCID: PMC4695870 DOI: 10.3390/cancers7040870] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
The treatment of melanoma has been revolutionized by new therapies targeting MAPK signaling or the immune system. Unfortunately these therapies are hindered by either primary resistance or the development of acquired resistance. Resistance mechanisms involving somatic mutations in genes associated with resistance have been identified in some cases of melanoma, however, the cause of resistance remains largely unexplained in other cases. The importance of epigenetic factors targeting histones and histone modifiers in driving the behavior of melanoma is only starting to be unraveled and provides significant opportunity to combat the problems of therapy resistance. There is also an increasing ability to target these epigenetic changes with new drugs that inhibit these modifications to either prevent or overcome resistance to both MAPK inhibitors and immunotherapy. This review focuses on changes in histones, histone reader proteins and histone positioning, which can mediate resistance to new therapeutics and that can be targeted for future therapies.
Collapse
Affiliation(s)
- Stuart J Gallagher
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Jessamy C Tiffen
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| |
Collapse
|
28
|
Faião-Flores F, Quincoces Suarez JA, Fruet AC, Maria-Engler SS, Pardi PC, Maria DA. Curcumin analog DM-1 in monotherapy or combinatory treatment with dacarbazine as a strategy to inhibit in vivo melanoma progression. PLoS One 2015; 10:e0118702. [PMID: 25742310 PMCID: PMC4350837 DOI: 10.1371/journal.pone.0118702] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/14/2015] [Indexed: 01/22/2023] Open
Abstract
Malignant melanoma is a highly aggressive form of skin cancer with a high mortality rate if not discovered in early stages. Although a limited number of treatment options for melanoma currently exist, patients with a more aggressive form of this cancer frequently decline treatment. DM-1 is a sodium phenolate and curcumin analog with proven anticancer, anti-proliferative and anti-metastatic properties. In this paper, the DM-1 compound showed in vivo antitumor activity alone or in combination with chemotherapeutic DTIC in B16F10 melanoma-bearing mice. Beneficial effects such as melanoma tumor burden reduction with pyknotic nuclei, decreased nuclei/cytoplasmic ratio and nuclear degradation occurred after DM-1 treatment. No toxicological changes were observed in the liver, kidneys, spleen and lungs after DM-1 monotherapy or DTIC combined therapy. DTIC+DM-1 treatment induced the recovery of anemia arising from melanoma and immunomodulation. Both DM-1 treatment alone and in combination with DTIC induced apoptosis with the cleavage of caspase-3, -8 and -9. Furthermore, melanoma tumors treated with DM-1 showed a preferential apoptotic intrinsic pathway by decreasing Bcl-2/Bax ratio. Considering the chemoresistance exhibited by melanoma towards conventional chemotherapy drugs, DM-1 compound in monotherapy or in combination therapy provides a promising improvement in melanoma treatment with a reduction of side effects.
Collapse
Affiliation(s)
- Fernanda Faião-Flores
- Department of Clinical Chemistry & Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- * E-mail: (FFF); (DAM)
| | | | - Andréa Costa Fruet
- Department of Clinical Chemistry & Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Silvya Stuchi Maria-Engler
- Department of Clinical Chemistry & Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paulo Celso Pardi
- Laboratory of Experimental Pathology, Anhanguera University of São Paulo, UNIAN, São Paulo, Brazil
| | - Durvanei Augusto Maria
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São Paulo, Brazil
- * E-mail: (FFF); (DAM)
| |
Collapse
|
29
|
Bhatia P, Friedlander P, Zakaria EA, Kandil E. Impact of BRAF mutation status in the prognosis of cutaneous melanoma: an area of ongoing research. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:24. [PMID: 25738144 PMCID: PMC4322160 DOI: 10.3978/j.issn.2305-5839.2014.12.05] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 11/24/2014] [Indexed: 12/21/2022]
Abstract
This review is intended to provide an updated role of molecular genetics and various targeted therapies that have been developed to treat advanced stages of melanoma. Because of the declining success in melanoma therapy, the curative treatment for advanced stage melanoma has been a challenge for clinicians. Several mutations such as N-RAS, p53, BRAF including mutant-BRAF that lead to activation of kinase pathway, are implicated in the development of malignant melanoma. However, the current literature depicts that the prognostic role of BRAF mutation in disease progression is still controversial. While its higher level in advanced stage disease is associated with decreased overall survival (OS), some studies show that it failed to confer as an independent prognostic predictor of the disease. This has also led researchers to accomplish newer therapeutic strategies that lead to improved disease-response and grant survival benefits. Vemurafenib, a BRAF inhibitor agent, is one of the few available targeted therapies that is FDA approved and provides promising results in metastatic disease. However, its resistance at an early stage is of great concern. Recent implementation of combinational therapies including "targeted therapy", immunotherapy, and biological agents has appealed many researchers to define the adjunctive role of available therapies and their limitations in advanced stage and metastatic melanoma. This commends the need for future multi-institutional studies to confirm the clinical validity of different therapeutic strategies on a large scale population.
Collapse
|
30
|
Porcelli L, Guida G, Quatrale AE, Cocco T, Sidella L, Maida I, Iacobazzi RM, Ferretta A, Stolfa DA, Strippoli S, Guida S, Tommasi S, Guida M, Azzariti A. Aurora kinase B inhibition reduces the proliferation of metastatic melanoma cells and enhances the response to chemotherapy. J Transl Med 2015; 13:26. [PMID: 25623468 PMCID: PMC4314759 DOI: 10.1186/s12967-015-0385-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/08/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The poor response to chemotherapy and the brief response to vemurafenib in metastatic melanoma patients, make the identification of new therapeutic approaches an urgent need. Interestingly the increased expression and activity of the Aurora kinase B during melanoma progression suggests it as a promising therapeutic target. METHODS The efficacy of the Aurora B kinase inhibitor barasertib-HQPA was evaluated in BRAF mutated cells, sensitive and made resistant to vemurafenib after chronic exposure to the drug, and in BRAF wild type cells. The drug effectiveness has been evaluated as cell growth inhibition, cell cycle progression and cell migration. In addition, cellular effectors of drug resistance and response were investigated. RESULTS The characterization of the effectors responsible for the resistance to vemurafenib evidenced the increased expression of MITF or the activation of Erk1/2 and p-38 kinases in the newly established cell lines with a phenotype resistant to vemurafenib. The sensitivity of cells to barasertib-HQPA was irrespective of BRAF mutational status. Barasertib-HQPA induced the mitotic catastrophe, ultimately causing apoptosis and necrosis of cells, inhibited cell migration and strongly affected the glycolytic metabolism of cells inducing the release of lactate. In association i) with vemurafenib the gain in effectiveness was found only in BRAF(V600K) cells while ii) with nab-paclitaxel, the combination was more effective than each drug alone in all cells. CONCLUSIONS These findings suggest barasertib as a new therapeutic agent and as enhancer of chemotherapy in metastatic melanoma treatment.
Collapse
Affiliation(s)
- Letizia Porcelli
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Gabriella Guida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Anna E Quatrale
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Tiziana Cocco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Letizia Sidella
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Immacolata Maida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Rosa M Iacobazzi
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Anna Ferretta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Diana A Stolfa
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Sabino Strippoli
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Stefania Guida
- Unit of Dermatology and Venereology, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Stefania Tommasi
- Molecular Genetics Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Michele Guida
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Amalia Azzariti
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| |
Collapse
|