1
|
Hasan AU, Serada S, Sato S, Obara M, Hirata S, Nagase Y, Kondo Y, Taira E. KDM4B Histone Demethylase Inhibition Attenuates Tumorigenicity of Malignant Melanoma Cells by Overriding the p53-Mediated Tumor Suppressor Pathway. J Cell Biochem 2024:e30643. [PMID: 39358852 DOI: 10.1002/jcb.30643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024]
Abstract
Despite significant advances in the treatment of cutaneous melanoma (hereafter melanoma), the prognosis remains less favorable due to therapeutic resistance, which is presumably linked to epigenetic dysregulation. We hypothesized that the histone lysine demethylase KDM4B could play a pivotal role in controlling therapy-resistant melanoma. To validate our hypothesis, we retrieved RNA sequencing data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) program and observed upregulation of KDM4B in both primary and metastatic melanoma, which was associated with poor survival. To explore its role, we used murine B16, human SK-MEL-5, and G-361 melanoma cells as in vitro models of melanoma. We found that KDM4B inhibition using NCGC00244536 increased global levels of H3K9me3 and downregulated the expressions of cell cycle progression-related genes Cdk1, Cdk4, Ccnb1, and Ccnd1. Moreover, genetic ablation of KDM4B or its chemical inhibition using NCGC00244536 reduced p53 production by upregulating MDM2, which enhances the proteolytic degradation of p53. Interestingly, despite the reduction of p53, these interventions augmented apoptosis and senescence-induced cell death by activating pathways downstream of p53, as evidenced by reduced levels of pro-survival Bcl-2 and Bcl-xL proteins and increased production of pro-apoptotic cleaved caspase-3, caspase-7, Bax, and the senescence inducer Cdkn1a. Compared to the FDA-approved anti-melanoma agent dacarbazine, NCGC00244536 exhibited more pronounced cytotoxic and antiproliferative effects in melanoma cells. Importantly, NCGC00244536 demonstrated minimal cytotoxicity to low Kdm4b-expressing mouse embryonic fibroblasts. In conclusion, our findings suggest that KDM4B inhibition can override the antitumor effect of p53, and potentially serve as a therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- Arif Ul Hasan
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Satoshi Serada
- Department of Molecular Pathophysiology, Institute for Biomedical Sciences, Iwate Medical University, Iwate, Japan
| | - Sachiko Sato
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Mami Obara
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Sho Hirata
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yukako Nagase
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yukiko Kondo
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Eiichi Taira
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| |
Collapse
|
2
|
Ingelshed K, Melssen MM, Kannan P, Chandramohan A, Partridge AW, Jiang L, Wermeling F, Lane DP, Nestor M, Spiegelberg D. MDM2/MDMX inhibition by Sulanemadlin synergizes with anti-Programmed Death 1 immunotherapy in wild-type p53 tumors. iScience 2024; 27:109862. [PMID: 38784022 PMCID: PMC11112618 DOI: 10.1016/j.isci.2024.109862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/29/2023] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
Immunotherapy has revolutionized cancer treatment but its efficacy depends on a robust immune response in the tumor. Silencing of the tumor suppressor p53 is common in tumors and can affect the recruitment and activation of different immune cells, leading to immune evasion and poor therapy response. We found that the p53 activating stapled peptide MDM2/MDMX inhibitor Sulanemadlin (ALRN-6924) inhibited p53 wild-type cancer cell growth in vitro and in vivo. In mice carrying p53 wild-type CT26.WT tumors, monotherapy with the PD-1 inhibitor DX400 or Sulanemadlin delayed tumor doubling time by 50% and 37%, respectively, while combination therapy decreased tumor doubling time by 93% leading to an increased median survival time. Sulanemadlin treatment led to increased immunogenicity and combination treatment with PD-1 inhibition resulted in an increased tumor infiltration of lymphocytes. This combination treatment strategy could potentially turn partial responders into responders of immunotherapy, expanding the patient target group for PD-1-targeting immunotherapy.
Collapse
Affiliation(s)
- Katrine Ingelshed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Marit M. Melssen
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Pavitra Kannan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | | | - Long Jiang
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, 17177 Stockholm, Sweden
- Center for Molecular Medicine, 17176 Stockholm, Sweden
| | - Fredrik Wermeling
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, 17177 Stockholm, Sweden
- Center for Molecular Medicine, 17176 Stockholm, Sweden
| | - David P. Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
- Department of Surgical Sciences, Uppsala University, 75185 Uppsala, Sweden
| |
Collapse
|
3
|
Sanchez‐Pupo RE, Finch GA, Johnston DE, Craig H, Abdo R, Barr K, Kerfoot S, Dagnino L, Penuela S. Global pannexin 1 deletion increases tumor-infiltrating lymphocytes in the BRAF/Pten mouse melanoma model. Mol Oncol 2024; 18:969-987. [PMID: 38327091 PMCID: PMC10994229 DOI: 10.1002/1878-0261.13596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/20/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
Immunotherapies for malignant melanoma seek to boost the anti-tumoral response of CD8+ T cells, but have a limited patient response rate, in part due to limited tumoral immune cell infiltration. Genetic or pharmacological inhibition of the pannexin 1 (PANX1) channel-forming protein is known to decrease melanoma cell tumorigenic properties in vitro and ex vivo. Here, we crossed Panx1 knockout (Panx1-/-) mice with the inducible melanoma model BrafCA, PtenloxP, Tyr::CreERT2 (BPC). We found that deleting the Panx1 gene in mice does not reduce BRAF(V600E)/Pten-driven primary tumor formation or improve survival. However, tumors in BPC-Panx1-/- mice exhibited a significant increase in the infiltration of CD8+ T lymphocytes, with no changes in the expression of early T-cell activation marker CD69, lymphocyte activation gene 3 protein (LAG-3) checkpoint receptor, or programmed cell death ligand-1 (PD-L1) in tumors when compared to the BPC-Panx1+/+ genotype. Our results suggest that, although Panx1 deletion does not overturn the aggressive BRAF/Pten-driven melanoma progression in vivo, it does increase the infiltration of effector immune T-cell populations in the tumor microenvironment. We propose that PANX1-targeted therapy could be explored as a strategy to increase tumor-infiltrating lymphocytes to boost anti-tumor immunity.
Collapse
Affiliation(s)
| | - Garth A. Finch
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | | | - Heather Craig
- Department of Microbiology and ImmunologyWestern UniversityLondonCanada
| | - Rober Abdo
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | - Kevin Barr
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
| | - Steven Kerfoot
- Department of Microbiology and ImmunologyWestern UniversityLondonCanada
| | - Lina Dagnino
- Department of Physiology and PharmacologyWestern UniversityLondonCanada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and DentistryWestern UniversityLondonCanada
| | - Silvia Penuela
- Department of Anatomy and Cell BiologyWestern UniversityLondonCanada
- Division of Experimental Oncology, Department of Oncology, Schulich School of Medicine and DentistryWestern UniversityLondonCanada
| |
Collapse
|
4
|
Tzenaki N, Xenou L, Goulielmaki E, Tsapara A, Voudouri I, Antoniou A, Valianatos G, Tzardi M, De Bree E, Berdiaki A, Makrigiannakis A, Papakonstanti EA. A combined opposite targeting of p110δ PI3K and RhoA abrogates skin cancer. Commun Biol 2024; 7:26. [PMID: 38182748 PMCID: PMC10770346 DOI: 10.1038/s42003-023-05639-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024] Open
Abstract
Malignant melanoma is the most aggressive and deadly skin cancer with an increasing incidence worldwide whereas SCC is the second most common non-melanoma human skin cancer with limited treatment options. Here we show that the development and metastasis of melanoma and SCC cancers can be blocked by a combined opposite targeting of RhoA and p110δ PI3K. We found that a targeted induction of RhoA activity into tumours by deletion of p190RhoGAP-a potent inhibitor of RhoA GTPase-in tumour cells together with adoptive macrophages transfer from δD910A/D910A mice in mice bearing tumours with active RhoA abrogated growth progression of melanoma and SCC tumours. Τhe efficacy of this combined treatment is the same in tumours lacking activating mutations in BRAF and in tumours harbouring the most frequent BRAF(V600E) mutation. Furthermore, the efficiency of this combined treatment is associated with decreased ATX expression in tumour cells and tumour stroma bypassing a positive feedback expression of ATX induced by direct ATX pharmacological inactivation. Together, our findings highlight the importance of targeting cancer cells and macrophages for skin cancer therapy, emerge a reverse link between ATX and RhoA and illustrate the benefit of p110δ PI3K inhibition as a combinatorial regimen for the treatment of skin cancers.
Collapse
Affiliation(s)
- Niki Tzenaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Lydia Xenou
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Evangelia Goulielmaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Anna Tsapara
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Irene Voudouri
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Angelika Antoniou
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - George Valianatos
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Maria Tzardi
- Department of Pathology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Eelco De Bree
- Department of Surgical Oncology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Aikaterini Berdiaki
- Department of Obstetrics and Gynaecology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | - Antonios Makrigiannakis
- Department of Obstetrics and Gynaecology, School of Medicine, University of Crete, University Hospital, Heraklion, Greece
| | | |
Collapse
|
5
|
Probst HC, Stoitzner P, Amon L, Backer RA, Brand A, Chen J, Clausen BE, Dieckmann S, Dudziak D, Heger L, Hodapp K, Hornsteiner F, Hovav AH, Jacobi L, Ji X, Kamenjarin N, Lahl K, Lahmar I, Lakus J, Lehmann CHK, Ortner D, Picard M, Roberti MP, Rossnagel L, Saba Y, Schalla C, Schlitzer A, Schraml BU, Schütze K, Seichter A, Seré K, Seretis A, Sopper S, Strandt H, Sykora MM, Theobald H, Tripp CH, Zitvogel L. Guidelines for DC preparation and flow cytometry analysis of mouse nonlymphoid tissues. Eur J Immunol 2023; 53:e2249819. [PMID: 36512638 DOI: 10.1002/eji.202249819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various nonlymphoid tissues. DC are sentinels of the immune system present in almost every mammalian organ. Since they represent a rare cell population, DC need to be extracted from organs with protocols that are specifically developed for each tissue. This article provides detailed protocols for the preparation of single-cell suspensions from various mouse nonlymphoid tissues, including skin, intestine, lung, kidney, mammary glands, oral mucosa and transplantable tumors. Furthermore, our guidelines include comprehensive protocols for multiplex flow cytometry analysis of DC subsets and feature top tricks for their proper discrimination from other myeloid cells. With this collection, we provide guidelines for in-depth analysis of DC subsets that will advance our understanding of their respective roles in healthy and diseased tissues. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all coauthors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Ronald A Backer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jianzhou Chen
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Björn E Clausen
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sophie Dieckmann
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
- Friedrich-Alexander University (FAU), Erlangen-Nürnberg, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Katrin Hodapp
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Florian Hornsteiner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Avi-Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Lukas Jacobi
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Xingqi Ji
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 82152, Planegg-Martinsried, Germany
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Nadine Kamenjarin
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katharina Lahl
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, 2800, Denmark
- Immunology Section, Lund University, Lund, 221 84, Sweden
| | - Imran Lahmar
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Jelena Lakus
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
| | - Daniela Ortner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marion Picard
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lukas Rossnagel
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Yasmin Saba
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Carmen Schalla
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Barbara U Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 82152, Planegg-Martinsried, Germany
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Kristian Schütze
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Athanasios Seretis
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Sieghart Sopper
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Center, Innsbruck, Austria
| | - Helen Strandt
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina M Sykora
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Center, Innsbruck, Austria
| | - Hannah Theobald
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Christoph H Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| |
Collapse
|
6
|
Capoferri D, Chiodelli P, Corli M, Belleri M, Scalvini E, Mignani L, Guerra J, Grillo E, De Giorgis V, Manfredi M, Presta M. The Pro-Oncogenic Sphingolipid-Metabolizing Enzyme β-Galactosylceramidase Modulates the Proteomic Landscape in BRAF(V600E)-Mutated Human Melanoma Cells. Int J Mol Sci 2023; 24:10555. [PMID: 37445731 DOI: 10.3390/ijms241310555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/13/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
β-Galactosylceramidase (GALC) is a lysosomal enzyme involved in sphingolipid metabolism by removing β-galactosyl moieties from β-galactosylceramide and β-galactosylsphingosine. Previous observations have shown that GALC may exert pro-oncogenic functions in melanoma and Galc silencing, leading to decreased oncogenic activity in murine B16 melanoma cells. The tumor-driving BRAF(V600E) mutation is present in approximately 50% of human melanomas and represents a major therapeutic target. However, such mutation is missing in melanoma B16 cells. Thus, to assess the impact of GALC in human melanoma in a more relevant BRAF-mutated background, we investigated the effect of GALC overexpression on the proteomic landscape of A2058 and A375 human melanoma cells harboring the BRAF(V600E) mutation. The results obtained by liquid chromatography-tandem mass spectrometry (LC-MS/MS) demonstrate that significant differences exist in the protein landscape expressed under identical cell culture conditions by A2058 and A375 human melanoma cells, both harboring the same BRAF(V600E)-activating mutation. GALC overexpression resulted in a stronger impact on the proteomic profile of A375 cells when compared to A2058 cells (261 upregulated and 184 downregulated proteins versus 36 and 14 proteins for the two cell types, respectively). Among them, 25 proteins appeared to be upregulated in both A2058-upGALC and A375-upGALC cells, whereas two proteins were significantly downregulated in both GALC-overexpressing cell types. These proteins appear to be involved in melanoma biology, tumor invasion and metastatic dissemination, tumor immune escape, mitochondrial antioxidant activity, endoplasmic reticulum stress responses, autophagy, and/or apoptosis. Notably, analysis of the expression of the corresponding genes in human skin cutaneous melanoma samples (TCGA, Firehose Legacy) using the cBioPortal for Cancer Genomics platform demonstrated a positive correlation between GALC expression and the expression levels of 14 out of the 27 genes investigated, thus supporting the proteomic findings. Overall, these data indicate for the first time that the expression of the lysosomal sphingolipid-metabolizing enzyme GALC may exert a pro-oncogenic impact on the proteomic landscape in BRAF-mutated human melanoma.
Collapse
Affiliation(s)
- Davide Capoferri
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Paola Chiodelli
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Marzia Corli
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mirella Belleri
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Elisa Scalvini
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Luca Mignani
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Jessica Guerra
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Elisabetta Grillo
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Allergic and Autoimmune Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Allergic and Autoimmune Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Marco Presta
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Consorzio Interuniversitario Biotecnologie (CIB), Unit of Brescia, 25123 Brescia, Italy
| |
Collapse
|
7
|
Pinho JO, Matias M, Godinho-Santos A, Amaral JD, Mendes E, Jesus Perry M, Paula Francisco A, Rodrigues CMP, Manuela Gaspar M. A step forward on the in vitro and in vivo assessment of a novel nanomedicine against melanoma. Int J Pharm 2023; 640:123011. [PMID: 37146952 DOI: 10.1016/j.ijpharm.2023.123011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023]
Abstract
Melanoma is the most aggressive form of skin cancer, with increasing incidence and mortality rates. To overcome current treatment limitations, a hybrid molecule (HM) combining a triazene and a ʟ-tyrosine analogue, was recently synthesized, incorporated in long blood circulating liposomes (LIP HM) and validated in an immunocompetent melanoma model. The present work constitutes a step forward in the therapeutic assessment of HM formulations. Here, human melanoma cells, A375 and MNT-1, were used and dacarbazine (DTIC), a triazene drug clinically available as first-line treatment for melanoma, constituted the positive control. In cell cycle analysis, A375 cells, after 24-h incubation with HM (60 μM) and DTIC (70 μM), resulted in a 1.2 fold increase (related to control) in the percentage of cells in G0/G1 phase. The therapeutic activity was evaluated in a human murine melanoma model (subcutaneously injected with A375 cells) to most closely resemble the human pathology. Animals treated with LIP HM exhibited the highest antimelanoma effect resulting in a 6-, 5- and 4-fold reduction on tumor volume compared to negative control, Free HM and DTIC groups, respectively. No toxic side effects were detected. Overall, these results constitute another step forward in the validation of the antimelanoma activity of LIP HM, using a murine model that more accurately simulates the pathology that occurs in human patients.
Collapse
Affiliation(s)
- Jacinta O Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Mariana Matias
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Ana Godinho-Santos
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Joana D Amaral
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Eduarda Mendes
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Jesus Perry
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Ana Paula Francisco
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Cecília M P Rodrigues
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - M Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
8
|
Alam MM, Gower T, Jiang M, Oppenheim JJ, Yang D. A Therapeutic Vaccine in Combination with Cyclic GMP-AMP Cures More Differentiated Melanomas in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1428-1436. [PMID: 36947147 PMCID: PMC10121855 DOI: 10.4049/jimmunol.2200371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 02/22/2023] [Indexed: 03/23/2023]
Abstract
We have identified a combinational immunotherapy termed TheraVac vaccine (TheraVac) that can cure multiple large established mouse tumors, but it failed to cure melanoma in mice. TheraVac consists of an immunostimulating arm containing an agonist (HMGN1 [N1]) for TLR4 and an agonist (R848) for TLR7/8 that synergize to activate tumor-infiltrating dendritic cells (DCs) and promote Th1 immune responses. The second arm uses an immune checkpoint blockade, anti-PDL-1, to diminish tumor-associated immunosuppression. In this study, we investigated supplementation of TheraVac by a stimulator of IFN genes (STING) agonist, cyclic GMP-AMP (cGAMP), because together they synergize in activating DCs and produced more immunostimulating IL-12p70 and TNF-α cytokines. The synergistic activation and maturation of DCs is dependent on the activation of tank binding kinase-1 (TBK1). Treatment of three different melanin-producing mouse melanomas (B16F1, M3, and M4) with intratumoral delivery of cGAMP and TheraVac eradicated 60-80% of these melanomas. Immunoprofiling of M3 tumor treated with TheraVac plus cGAMP showed an increase in CD8+ CTLs and macrophages in the tumor. There was also a marked increase of CD4, CD8 effector and memory T cells and generation of functional tumor-specific CTLs in tumor-draining lymph nodes. The resultant tumor-free mice were selectively resistant to subsequent challenge with the same tumors, indicating long-term tumor-specific protective immunity. Overall, our findings have important implications for clinical trials with a combination of these immunotherapeutics to cure melanin-producing human melanomas, without the need for exogenous tumor Ags and no clear toxic effects in mice.
Collapse
Affiliation(s)
- Md Masud Alam
- Cellular Immunology Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Timothy Gower
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Mengmeng Jiang
- Cellular Immunology Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Joost J Oppenheim
- Cellular Immunology Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - De Yang
- Cellular Immunology Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
9
|
Costas-Insua C, Seijo-Vila M, Blázquez C, Blasco-Benito S, Rodríguez-Baena FJ, Marsicano G, Pérez-Gómez E, Sánchez C, Sánchez-Laorden B, Guzmán M. Neuronal Cannabinoid CB 1 Receptors Suppress the Growth of Melanoma Brain Metastases by Inhibiting Glutamatergic Signalling. Cancers (Basel) 2023; 15:cancers15092439. [PMID: 37173906 PMCID: PMC10177062 DOI: 10.3390/cancers15092439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Melanoma is one of the deadliest forms of cancer. Most melanoma deaths are caused by distant metastases in several organs, especially the brain, the so-called melanoma brain metastases (MBMs). However, the precise mechanisms that sustain the growth of MBMs remain elusive. Recently, the excitatory neurotransmitter glutamate has been proposed as a brain-specific, pro-tumorigenic signal for various types of cancers, but how neuronal glutamate shuttling onto metastases is regulated remains unknown. Here, we show that the cannabinoid CB1 receptor (CB1R), a master regulator of glutamate output from nerve terminals, controls MBM proliferation. First, in silico transcriptomic analysis of cancer-genome atlases indicated an aberrant expression of glutamate receptors in human metastatic melanoma samples. Second, in vitro experiments conducted on three different melanoma cell lines showed that the selective blockade of glutamatergic NMDA receptors, but not AMPA or metabotropic receptors, reduces cell proliferation. Third, in vivo grafting of melanoma cells in the brain of mice selectively devoid of CB1Rs in glutamatergic neurons increased tumour cell proliferation in concert with NMDA receptor activation, whereas melanoma cell growth in other tissue locations was not affected. Taken together, our findings demonstrate an unprecedented regulatory role of neuronal CB1Rs in the MBM tumour microenvironment.
Collapse
Affiliation(s)
- Carlos Costas-Insua
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Marta Seijo-Vila
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Cristina Blázquez
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Sandra Blasco-Benito
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Francisco Javier Rodríguez-Baena
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH), 03550 San Juan de Alicante, Spain
| | - Giovanni Marsicano
- Physiopathologie de la Plasticité Neuronale, NeuroCentre Magendie, U1215 Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Neurocampus, University of Bordeaux, 33077 Bordeaux, France
| | - Eduardo Pérez-Gómez
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Cristina Sánchez
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Berta Sánchez-Laorden
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH), 03550 San Juan de Alicante, Spain
| | - Manuel Guzmán
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University of Madrid, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| |
Collapse
|
10
|
Ingelshed K, Spiegelberg D, Kannan P, Påvénius L, Hacheney J, Jiang L, Eisinger S, Lianoudaki D, Lama D, Castillo F, Bosdotter C, Kretzschmar WW, Al-Radi O, Fritz N, Villablanca EJ, Karlsson MCI, Wermeling F, Nestor M, Lane DP, Sedimbi SK. The MDM2 Inhibitor Navtemadlin Arrests Mouse Melanoma Growth In Vivo and Potentiates Radiotherapy. CANCER RESEARCH COMMUNICATIONS 2022; 2:1075-1088. [PMID: 36922937 PMCID: PMC10010373 DOI: 10.1158/2767-9764.crc-22-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/21/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022]
Abstract
The tumor suppressor protein p53 is mutated in close to 50% of human tumors and is dysregulated in many others, for instance by silencing or loss of p14ARF. Under steady-state conditions, the two E3 ligases MDM2/MDM4 interact with and inhibit the transcriptional activity of p53. Inhibition of p53-MDM2/4 interaction to reactivate p53 in tumors with wild-type (WT) p53 has therefore been considered a therapeutic strategy. Moreover, studies indicate that p53 reactivation may synergize with radiation and increase tumor immunogenicity. In vivo studies of most MDM2 inhibitors have utilized immunodeficient xenograft mouse models, preventing detailed studies of action of these molecules on the immune response. The mouse melanoma cell line B16-F10 carries functional, WT p53 but does not express the MDM2 regulator p19ARF. In this study, we tested a p53-MDM2 protein-protein interaction inhibitor, the small molecule Navtemadlin, which is currently being tested in phase II clinical trials. Using mass spectrometry-based proteomics and imaging flow cytometry, we identified specific protein expression patterns following Navtemadlin treatment of B16-F10 melanoma cells compared with their p53 CRISPR-inactivated control cells. In vitro, Navtemadlin induced a significant, p53-dependent, growth arrest but little apoptosis in B16-F10 cells. When combined with radiotherapy, Navtemadlin showed synergistic effects and increased apoptosis. In vivo, Navtemadlin treatment significantly reduced the growth of B16-F10 melanoma cells implanted in C57Bl/6 mice. Our data highlight the utility of a syngeneic B16-F10 p53+/+ mouse melanoma model for assessing existing and novel p53-MDM2/MDM4 inhibitors and in identifying new combination therapies that can efficiently eliminate tumors in vivo. Significance The MDM2 inhibitor Navtemadlin arrests mouse tumor growth and potentiates radiotherapy. Our results support a threshold model for apoptosis induction that requires a high, prolonged p53 signaling for cancer cells to become apoptotic.
Collapse
Affiliation(s)
- Katrine Ingelshed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Pavitra Kannan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Linnéa Påvénius
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Hacheney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Long Jiang
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Silke Eisinger
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Danai Lianoudaki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Dilraj Lama
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Francisca Castillo
- Center for Molecular Medicine, Stockholm, Sweden
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Bosdotter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Omayma Al-Radi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nicolas Fritz
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo J. Villablanca
- Center for Molecular Medicine, Stockholm, Sweden
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Mikael C. I. Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Wermeling
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - David P. Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Corresponding Authors: Saikiran K. Sedimbi and David P. Lane, Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Nobels Väg 16, Stockholm, SE-17177, Sweden. Phone: +46 852 448 6452; E-mail: ; Phone: +46 852 448 6452; E-mail:
| | - Saikiran K. Sedimbi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Corresponding Authors: Saikiran K. Sedimbi and David P. Lane, Department of Microbiology Tumor and Cell Biology, Karolinska Institute, Nobels Väg 16, Stockholm, SE-17177, Sweden. Phone: +46 852 448 6452; E-mail: ; Phone: +46 852 448 6452; E-mail:
| |
Collapse
|
11
|
Austin E, Huang A, Wang JY, Cohen M, Heilman E, Maverakis E, Michl J, Jagdeo J. Red Light Phototherapy Using Light-Emitting Diodes Inhibits Melanoma Proliferation and Alters Tumor Microenvironments. Front Oncol 2022; 12:928484. [PMID: 35847848 PMCID: PMC9278815 DOI: 10.3389/fonc.2022.928484] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 01/26/2023] Open
Abstract
Background Total annual cancer rates have decreased due to improved treatment and prevention. However, the incidence of melanoma is rising, and not all patients respond to immune and targeted approaches. Therefore, we sought to determine the efficacy of red light (RL) phototherapy in preclinical models of melanoma. Methods Melanoma cells (A375, B16F10, MNT-1) were irradiated with RL. Melanoma proliferation, apoptosis, oxidative stress, and p53 phosphorylation were measured in vitro. In C57BL/6 mice, phototherapy safety, B16F10 tumor growth, and immunocyte infiltration were assessed following RL. Results In vitro, 640 J/cm2 RL decreased cellular proliferation without increasing apoptosis, while 1280 J/cm2 increased apoptosis. RL increased intracellular reactive oxygen species generation and p53 phosphorylation. In animal models, 2560 J/cm2 RL significantly prevented melanoma growth and increased the expression of CD103+ dendritic cells. 1280 and 1920 J/cm2 RL decreased tumor volume, but not significantly. RL did not cause skin inflammation or erythema in normal skin. Conclusion RL represents a potentially safe and effective melanoma therapeutic. RL prevented tumor growth and increased the expression of immune markers, such as CD103, that are associated with favorable melanoma outcomes. Further research is needed to determine the optimal clinical treatment regimen for melanoma using RL.
Collapse
Affiliation(s)
- Evan Austin
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States,Department of Dermatology, University of California (UC) Davis Medical Center, Sacramento, CA, United States
| | - Alisen Huang
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Jennifer Y. Wang
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Marc Cohen
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Edward Heilman
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Emanual Maverakis
- Department of Dermatology, University of California (UC) Davis Medical Center, Sacramento, CA, United States
| | - Josef Michl
- Department of Pathology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Jared Jagdeo
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States,Department of Dermatology, University of California (UC) Davis Medical Center, Sacramento, CA, United States,*Correspondence: Jared Jagdeo,
| |
Collapse
|
12
|
Bellavia MC, Nyiranshuti L, Latoche JD, Ho KV, Fecek RJ, Taylor JL, Day KE, Nigam S, Pun M, Gallazzi F, Edinger RS, Storkus WJ, Patel RB, Anderson CJ. PET Imaging of VLA-4 in a New BRAF V600E Mouse Model of Melanoma. Mol Imaging Biol 2022; 24:425-433. [PMID: 34694528 PMCID: PMC9183947 DOI: 10.1007/s11307-021-01666-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/05/2023]
Abstract
PURPOSE Despite unprecedented responses to immune checkpoint inhibitors and targeted therapy in melanoma, a major subset of patients progresses and have few effective salvage options. We have previously demonstrated robust, selective uptake of the peptidomimetic LLP2A labeled with Cu-64 ([64Cu]-LLP2A) for positron emission tomography (PET) imaging in subcutaneous and metastatic models of B16F10 murine melanoma. LLP2A binds with high affinity to very late antigen-4 (VLA-4, integrin α4β1), a transmembrane protein overexpressed in melanoma and other cancers that facilitates tumor growth and metastasis. Yet B16F10 fails to faithfully reflect human melanoma biology, as it lacks certain oncogenic driver mutations, including BRAF mutations found in ≥ 50 % of clinical specimens. Here, we evaluated the PET tracer [64Cu]-CB-TE1A1P-PEG4-LLP2A ([64Cu]-LLP2A) in novel, translational BRAFV600E mutant melanoma models differing in VLA-4 expression-BPR (VLA-4-) and BPRα (VLA-4+). PROCEDURES BPR cells were transduced with α4 (CD49d) to overexpress intact cell surface VLA-4 (BPRα). The binding affinity of [64Cu]-LLP2A to BPR and BPRα cells was determined by saturation binding assays. [64Cu]-LLP2A internalization into B16F10, BPR, and BPRα cells was quantified via a plate-based assay. Tracer biodistribution and PET/CT imaging were evaluated in mice bearing subcutaneous BPR and BPRα tumors. RESULTS [64Cu]-LLP2A demonstrated high binding affinity to BPRα (Kd = 1.4 nM) but indeterminate binding to BPR cells. VLA-4+ BPRα and B16F10 displayed comparable time-dependent [64Cu]-LLP2A internalization, whereas BPR internalization was undetectable. PET/CT showed increased tracer uptake in BPRα tumors vs. BPR tumors in vivo, which was validated by significantly greater (p < 0.0001) BPRα tumor uptake in biodistribution analyses. CONCLUSIONS [64Cu]-LLP2A discriminates BPRα (VLA-4+) vs. BPR (VLA-4-) melanomas in vivo, supporting translation of these BRAF-mutated melanoma models via prospective imaging and theranostic studies. These results extend the utility of LLP2A to selectively target clinically relevant and therapy-resistant tumor variants toward its use for therapeutic patient care.
Collapse
Affiliation(s)
- Michael C Bellavia
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lea Nyiranshuti
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- RayzeBio Inc., San Diego, CA, 92121, USA
| | - Joseph D Latoche
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Khanh-Van Ho
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Ronald J Fecek
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Microbiology and Immunology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, 15601, USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kathryn E Day
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Shubhanchi Nigam
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Eurofins Scientific, Philadelphia, PA, 19355, USA
| | - Michael Pun
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Fabio Gallazzi
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Robert S Edinger
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Walter J Storkus
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ravi B Patel
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Carolyn J Anderson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA.
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Radiology, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
13
|
Kim H, Hwang E, Park BC, Kim SJ. Novel potential NOX2 inhibitors, Dudleya brittonii water extract and polygalatenoside A inhibit intracellular ROS generation and growth of melanoma. Biomed Pharmacother 2022; 150:112967. [PMID: 35430393 DOI: 10.1016/j.biopha.2022.112967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 11/02/2022] Open
Abstract
Reactive oxygen species (ROS) are key regulators of the proliferation, metastasis, and drug resistance of melanoma, which accounts for 60% of skin cancer deaths. In a previous study, we developed Dudleya brittonii water extract (DBWE) with antioxidant activity, but the mechanism of action and bioactive substances of DBWE have not been fully identified. This study showed altered NADPH oxidase 2 (NOX2) expression and selective inhibition of cytosolic ROS but not mitochondrial ROS in B16-F10 melanoma cells, suggesting the NOX2 inhibitory potential of DBWE. In addition, DBWE inhibited mitochondrial activity, lipid metabolism, and cell cycle in B16-F10 cells. The anti-melanoma effect of DBWE was abrogated by the addition of ROS, and there was no significant change in the melanogenesis pathway. Polygalatenoside A was identified as a candidate bioactive substance in the DBWE aqueous fraction through mass spectrometry, and the DBWE-like anti-melanoma effect was confirmed. These data suggest that DBWE and polygalatenoside A have the potential to prevent and treat melanoma.
Collapse
Affiliation(s)
- Hyungkuen Kim
- Division of Cosmetics and Biotechnology, College of Life and Health Sciences, Hoseo University, Baebang, Asan, Chungnam 31499, Republic of Korea
| | - Eunmi Hwang
- Division of Cosmetics and Biotechnology, College of Life and Health Sciences, Hoseo University, Baebang, Asan, Chungnam 31499, Republic of Korea
| | - Byung-Chul Park
- Graduate School of International Agricultural Technology, Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea.
| | - Sung-Jo Kim
- Division of Cosmetics and Biotechnology, College of Life and Health Sciences, Hoseo University, Baebang, Asan, Chungnam 31499, Republic of Korea.
| |
Collapse
|
14
|
Murphy JM, Jeong K, Ahn EYE, Lim STS. Nuclear focal adhesion kinase induces APC/C activator protein CDH1-mediated cyclin-dependent kinase 4/6 degradation and inhibits melanoma proliferation. J Biol Chem 2022; 298:102013. [PMID: 35525274 PMCID: PMC9163754 DOI: 10.1016/j.jbc.2022.102013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of cyclin-dependent kinases (CDKs) can promote unchecked cell proliferation and cancer progression. Although focal adhesion kinase (FAK) contributes to regulating cell cycle progression, the exact molecular mechanism remains unclear. Here, we found that FAK plays a key role in cell cycle progression potentially through regulation of CDK4/6 protein expression. We show that FAK inhibition increased its nuclear localization and induced G1 arrest in B16F10 melanoma cells. Mechanistically, we demonstrate nuclear FAK associated with CDK4/6 and promoted their ubiquitination and proteasomal degradation through recruitment of CDC homolog 1 (CDH1), an activator and substrate recognition subunit of the anaphase-promoting complex/cyclosome E3 ligase complex. We found the FAK N-terminal FERM domain acts as a scaffold to bring CDK4/6 and CDH1 within close proximity. However, overexpression of nonnuclear-localizing mutant FAK FERM failed to function as a scaffold for CDK4/6 and CDH1. Furthermore, shRNA knockdown of CDH1 increased CDK4/6 protein expression and blocked FAK inhibitor-induced reduction of CDK4/6 in B16F10 cells. In vivo, we show that pharmacological FAK inhibition reduced B16F10 tumor size, correlating with increased FAK nuclear localization and decreased CDK4/6 expression compared with vehicle controls. In patient-matched healthy skin and melanoma biopsies, we found FAK was mostly inactive and nuclear localized in healthy skin, whereas melanoma lesions showed increased active cytoplasmic FAK and elevated CDK4 expression. Taken together, our data demonstrate that FAK inhibition blocks tumor proliferation by inducing G1 arrest, in part through decreased CDK4/6 protein stability by nuclear FAK.
Collapse
Affiliation(s)
- James M Murphy
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kyuho Jeong
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Eun-Young Erin Ahn
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ssang-Taek Steve Lim
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
15
|
De Beck L, Awad RM, Basso V, Casares N, De Ridder K, De Vlaeminck Y, Gnata A, Goyvaerts C, Lecocq Q, San José-Enériz E, Verhulst S, Maes K, Vanderkerken K, Agirre X, Prosper F, Lasarte JJ, Mondino A, Breckpot K. Inhibiting Histone and DNA Methylation Improves Cancer Vaccination in an Experimental Model of Melanoma. Front Immunol 2022; 13:799636. [PMID: 35634329 PMCID: PMC9134079 DOI: 10.3389/fimmu.2022.799636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has improved the treatment of malignant skin cancer of the melanoma type, yet overall clinical response rates remain low. Combination therapies could be key to meet this cogent medical need. Because epigenetic hallmarks represent promising combination therapy targets, we studied the immunogenic potential of a dual inhibitor of histone methyltransferase G9a and DNA methyltransferases (DNMTs) in the preclinical B16-OVA melanoma model. Making use of tumor transcriptomic and functional analyses, methylation-targeted epigenetic reprogramming was shown to induce tumor cell cycle arrest and apoptosis in vitro coinciding with transient tumor growth delay and an IFN-I response in immune-competent mice. In consideration of a potential impact on immune cells, the drug was shown not to interfere with dendritic cell maturation or T-cell activation in vitro. Notably, the drug promoted dendritic cell and, to a lesser extent, T-cell infiltration in vivo, yet failed to sensitize tumor cells to programmed cell death-1 inhibition. Instead, it increased therapeutic efficacy of TCR-redirected T cell and dendritic cell vaccination, jointly increasing overall survival of B16-OVA tumor-bearing mice. The reported data confirm the prospect of methylation-targeted epigenetic reprogramming in melanoma and sustain dual G9a and DNMT inhibition as a strategy to tip the cancer-immune set-point towards responsiveness to active and adoptive vaccination against melanoma.
Collapse
Affiliation(s)
- Lien De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Veronica Basso
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Noelia Casares
- Immunology and Immunotherapy Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
| | - Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Alessandra Gnata
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Edurne San José-Enériz
- Hemato-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
| | - Stefaan Verhulst
- Liver Cell Biology Research Group, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ken Maes
- Laboratory of Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Center for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Xabier Agirre
- Hemato-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
- Laboratory of Cancer Epigenetics, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Felipe Prosper
- Hemato-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
- Laboratory of Cancer Epigenetics, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Juan José Lasarte
- Immunology and Immunotherapy Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
16
|
Takeda T, Tsubaki M, Kato N, Genno S, Ichimura E, Enomoto A, Imano M, Satou T, Nishida S. Sorafenib treatment of metastatic melanoma with c-Kit aberration reduces tumor growth and promotes survival. Oncol Lett 2021; 22:827. [PMID: 34691254 DOI: 10.3892/ol.2021.13089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Melanomas are highly malignant tumors that readily metastasize and have poor prognosis. Targeted therapy is a cornerstone of treatment for patients with melanoma. Although c-Kit gene aberration has found in 5-10% of melanoma cases, research on c-Kit inhibitors for melanoma with c-Kit aberration have been disappointing. Sorafenib is a tyrosine kinase inhibitor, whose targets include c-Kit, platelet derived growth factor receptor (PDGFR), VEGFR and RAF. The present study aimed to examine the effect of sorafenib on metastatic melanoma with c-Kit aberration. Cell viability was assessed via trypan blue assay. Migration and invasion were analyzed using cell culture inserts. The anti-metastatic effects and antitumour activity of sorafenib were determined in an in vivo model. Protein expression was detected via western blotting, and the expression of MMP and very late antigen (VLA) was detected via reverse transcription-quantitative PCR. It was identified that sorafenib decreased cell viability, migration and invasion in vitro. Furthermore, sorafenib inhibited metastasis and tumor growth in vivo. Mechanistically, sorafenib inhibited c-Kit, PDGFR, VEGFR, B-Raf and c-Raf phosphorylation both in vitro and in vivo. In addition, sorafenib reduced the expression levels of MMPs and VLA. Importantly, there was a significant effect of sorafenib treatment on overall survival in mice. Collectively, this study suggests that sorafenib may serve as a novel therapeutic option for melanoma with c-Kit dysregulation.
Collapse
Affiliation(s)
- Tomoya Takeda
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Masanobu Tsubaki
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Natsuki Kato
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Shuji Genno
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Eri Ichimura
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Aya Enomoto
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University School of Medicine, Osakasayama, Osaka 589-0014, Japan
| | - Takao Satou
- Department of Pathology, Kindai University School of Medicine, Osakasayama, Osaka 589-0014, Japan
| | - Shozo Nishida
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|
17
|
Qin SS, Han BJ, Williams A, Jackson KM, Jewell R, Chacon AC, Lord EM, Linehan DC, Kim M, Reuben A, Gerber SA, Prieto PA. Intertumoral Genetic Heterogeneity Generates Distinct Tumor Microenvironments in a Novel Murine Synchronous Melanoma Model. Cancers (Basel) 2021; 13:cancers13102293. [PMID: 34064795 PMCID: PMC8151632 DOI: 10.3390/cancers13102293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Metastatic melanoma patients may present with multiple, simultaneous metastases that are genetically different. This intertumoral heterogeneity can cause these tumors to respond differently to the same systemic therapy. Progression of any one tumor, even when others regress, eventually leads to therapy termination. The mechanism underlying these mixed responses remains unknown due to a lack of clinically representative animal models. In a novel murine model of synchronous melanoma that recapitulates human intertumoral heterogeneity, we show that intertumoral genetic heterogeneity leads to the simultaneous generation of distinct tumor immune microenvironments within the same mouse. Furthermore, each tumor can independently regulate local PD-1 (programmed cell death protein 1) and PD-L1 (PD-1 ligand) expressions, an immunosuppressive axis targeted by popular checkpoint immunotherapies. This model is useful for furthering the study of intertumoral heterogeneity and of lesion-specific therapeutic responses. Abstract Metastatic melanoma portends a poor prognosis and patients may present with multiple, simultaneous tumors. Despite recent advances in systemic immunotherapy, a majority of patients fail to respond, or exhibit lesion-specific responses wherein some metastases respond as others progress within the same patient. While intertumoral heterogeneity has been clinically associated with these mixed lesion-specific therapeutic responses, no clear mechanism has been identified, largely due to the scarcity of preclinical models. We developed a novel murine synchronous melanoma model that recapitulates this intertumoral genetic and microenvironmental heterogeneity. We show that genetic differences between tumors are sufficient to generate distinct tumor immune microenvironments (TIME) simultaneously in the same mouse. Furthermore, these TIMEs lead to the independent regulation of PD-1/PD-L1 (programmed cell death protein 1/PD-1 ligand), a popular axis targeted by immune checkpoint therapy, in response to ongoing anti-tumor immunity and the presence of interferon-gamma. Currently, therapeutic selection for metastatic melanoma patients is guided by a single biopsy, which may not represent the immune status of all tumors. As a result, patients can display heterogeneous lesion-specific responses. Further investigations into this synchronous melanoma model will provide mechanistic insight into the effects of intertumoral heterogeneity and guide therapeutic selection in this challenging patient population.
Collapse
Affiliation(s)
- Shuyang S. Qin
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA; (S.S.Q.); (B.J.H.); (E.M.L.); (M.K.); (S.A.G.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
| | - Booyeon J. Han
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA; (S.S.Q.); (B.J.H.); (E.M.L.); (M.K.); (S.A.G.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
| | - Alyssa Williams
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Katherine M. Jackson
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Rachel Jewell
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Alexander C. Chacon
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Edith M. Lord
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA; (S.S.Q.); (B.J.H.); (E.M.L.); (M.K.); (S.A.G.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
| | - David C. Linehan
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA; (S.S.Q.); (B.J.H.); (E.M.L.); (M.K.); (S.A.G.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
| | - Alexandre Reuben
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA;
| | - Scott A. Gerber
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA; (S.S.Q.); (B.J.H.); (E.M.L.); (M.K.); (S.A.G.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Peter A. Prieto
- Center for Tumor Immunology Research, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA; (A.W.); (K.M.J.); (R.J.); (A.C.C.); (D.C.L.)
- Department of Surgery, University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
- Correspondence: ; Tel.: +1-(585)-275-1611
| |
Collapse
|
18
|
Fibroblast MMP14-Dependent Collagen Processing Is Necessary for Melanoma Growth. Cancers (Basel) 2021; 13:cancers13081984. [PMID: 33924099 PMCID: PMC8074311 DOI: 10.3390/cancers13081984] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Matrix metalloproteinases (MMPs) were considered as targets for the treatment of various cancers. However, initial trials using broad inhibitors to MMPs have failed, partly attributed to the contrasting functions of these proteases acting as tumor promoters and suppressors, among other reasons. Our data now suggest that specific inhibition of MMP14 might represent a more specific approach, as loss of this protease in fibroblasts resulted in reduced growth of grafted melanomas. Here, we found that deletion of MMP14 in fibroblasts generates a matrix-rich environment that reduces tumor vascularization and melanoma cell proliferation. In in vitro and ex vivo assays, we showed that the latter is mediated by stiffening of the tissue due to collagen accumulation. Additionally, in vivo, we show that independently of MMP14 deletion, a collagen-rich stiff matrix inhibits the growth of melanomas. Abstract Skin homeostasis results from balanced synthesis and degradation of the extracellular matrix in the dermis. Deletion of the proteolytic enzyme MMP14 in dermal fibroblasts (MMP14Sf−/−) leads to a fibrotic skin phenotype with the accumulation of collagen type I, resulting from impaired proteolysis. Here, we show that melanoma growth in these mouse fibrotic dermal samples was decreased, paralleled by reduced tumor cell proliferation and vessel density. Using atomic force microscopy, we found increased peritumoral matrix stiffness of early but not late melanomas in the absence of fibroblast-derived MMP14. However, total collagen levels were increased at late melanoma stages in MMP14Sf−/− mice compared to controls. In ex vivo invasion assays, melanoma cells formed smaller tumor islands in MMP14Sf−/− skin, indicating that MMP14-dependent matrix accumulation regulates tumor growth. In line with these data, in vitro melanoma cell growth was inhibited in high collagen 3D spheroids or stiff substrates. Most importantly, in vivo induction of fibrosis using bleomycin reduced melanoma tumor growth. In summary, we show that MMP14 expression in stromal fibroblasts regulates melanoma tumor progression by modifying the peritumoral matrix and point to collagen accumulation as a negative regulator of melanoma.
Collapse
|
19
|
Coppo R, Orso F, Virga F, Dalmasso A, Baruffaldi D, Nie L, Clapero F, Dettori D, Quirico L, Grassi E, Defilippi P, Provero P, Valdembri D, Serini G, Sadeghi MM, Mazzone M, Taverna D. ESDN inhibits melanoma progression by blocking E-selectin expression in endothelial cells via STAT3. Cancer Lett 2021; 510:13-23. [PMID: 33862151 DOI: 10.1016/j.canlet.2021.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/10/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
An interactive crosstalk between tumor and stroma cells is essential for metastatic melanoma progression. We evidenced that ESDN/DCBLD2/CLCP1 plays a crucial role in endothelial cells during the spread of melanoma. Precisely, increased extravasation and metastasis formation were revealed in ESDN-null mice injected with melanoma cells, even if the primary tumor growth, vessel permeability, and angiogenesis were not enhanced. Interestingly, improved adhesion of melanoma cells to ESDN-depleted endothelial cells was observed, due to the presence of higher levels of E-selectin transcripts/proteins in ESDN-defective cells. In accordance with these results, anticorrelation was observed between ESDN and E-selectin in human endothelial cells. Most importantly, our data revealed that cimetidine, an E-selectin inhibitor, was able to block cell adhesion, extravasation, and metastasis formation in ESDN-null mice, underlying a major role of ESDN in E-selectin transcription upregulation, which according to our data, may presumably be linked to STAT3. Based on our results, we propose a protective role for ESDN during the spread of melanoma and reveal its therapeutic potential.
Collapse
Affiliation(s)
- Roberto Coppo
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Orso
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federico Virga
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; VIB Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Alberto Dalmasso
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Desirée Baruffaldi
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lei Nie
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA; Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Fabiana Clapero
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 10060, Candiolo, Torino, Italy
| | - Daniela Dettori
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lorena Quirico
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elena Grassi
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paola Defilippi
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paolo Provero
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Milano, Italy
| | - Donatella Valdembri
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 10060, Candiolo, Torino, Italy; Department of Oncology, University of Torino School of Medicine, 10060, Candiolo, Torino, Italy
| | - Guido Serini
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 10060, Candiolo, Torino, Italy; Department of Oncology, University of Torino School of Medicine, 10060, Candiolo, Torino, Italy
| | - Mehran M Sadeghi
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA; Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Massimiliano Mazzone
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; VIB Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Daniela Taverna
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy; Dept. Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
20
|
Lee MS, Lim SH, Yu AR, Hwang CY, Kang I, Yeo EJ. Carfilzomib in Combination with Bortezomib Enhances Apoptotic Cell Death in B16-F1 Melanoma Cells. BIOLOGY 2021; 10:biology10020153. [PMID: 33671902 PMCID: PMC7918982 DOI: 10.3390/biology10020153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary The incidence rate of metastatic melanoma has been rapidly increasing worldwide and its 5-year survival rate is very low. Due to partial responses, various side effects, and resistance to any known cancer therapeutics, more potent and safer therapeutics are needed to increase the survival rate of patients with melanoma. Since proteasome inhibitors, such as bortezomib and carfilzomib, have been suggested as treatments for various cancers, we investigated their potential for the treatment of melanoma by studying their molecular mechanisms of action in B16-F1 melanoma cells. In this study, we found that both bortezomib and carfilzomib lead to apoptosis via ER stress as well as ROS accumulation and MMP loss in melanoma cells. Bortezomib and carfilzomib synergistically reduced B16-F1 tumor growth in vitro and in a C57BL/6 xenograft mouse model. Therefore, a combination therapy with carfilzomib and bortezomib at submaximal concentrations may reduce their side effects and be beneficial for melanoma treatment. Abstract Proteasome inhibitors, such as bortezomib (BZ) and carfilzomib (CFZ), have been suggested as treatments for various cancers. To utilize BZ and/or CFZ as effective therapeutics for treating melanoma, we studied their molecular mechanisms using B16-F1 melanoma cells. Flow cytometry of Annexin V-fluorescein isothiocyanate-labeled cells indicated apoptosis induction by treatment with BZ and CFZ. Apoptosis was evidenced by the activation of various caspases, including caspase 3, 8, 9, and 12. Treatment with BZ and CFZ induced endoplasmic reticulum (ER) stress, as indicated by an increase in eIF2α phosphorylation and the expression of ER stress-associated proteins, including GRP78, ATF6α, ATF4, XBP1, and CCAAT/enhancer-binding protein homologous protein. The effects of CFZ on ER stress and apoptosis were lower than that of BZ. Nevertheless, CFZ and BZ synergistically induced ER stress and apoptosis in B16-F1 cells. Furthermore, the combinational pharmacological interactions of BZ and CFZ against the growth of B16-F1 melanoma cells were assessed by calculating the combination index and dose-reduction index with the CompuSyn software. We found that the combination of CFZ and BZ at submaximal concentrations could obtain dose reduction by exerting synergistic inhibitory effects on cell growth. Moreover, this drug combination reduced tumor growth in C57BL/6 syngeneic mice. Taken together, these results suggest that CFZ in combination with BZ may be a beneficial and potential strategy for melanoma treatment.
Collapse
Affiliation(s)
- Min Seung Lee
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (M.S.L.); (S.H.L.)
| | - So Hyun Lim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (M.S.L.); (S.H.L.)
| | - Ah-Ran Yu
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; (A.-R.Y.); (C.Y.H.)
| | - Chi Yeon Hwang
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; (A.-R.Y.); (C.Y.H.)
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (I.K.); (E.-J.Y.); Tel.: +82-29-610-922 (I.K.); +82-32-899-6050 (E.-J.Y.); Fax: +82-29-656-349 (I.K.); +82-32-899-6039 (E.-J.Y.)
| | - Eui-Ju Yeo
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (M.S.L.); (S.H.L.)
- Correspondence: (I.K.); (E.-J.Y.); Tel.: +82-29-610-922 (I.K.); +82-32-899-6050 (E.-J.Y.); Fax: +82-29-656-349 (I.K.); +82-32-899-6039 (E.-J.Y.)
| |
Collapse
|
21
|
Rivera HM, Muñoz EN, Osuna D, Florez M, Carvajal M, Gómez LA. Reciprocal Changes in miRNA Expression with Pigmentation and Decreased Proliferation Induced in Mouse B16F1 Melanoma Cells by L-Tyrosine and 5-Bromo-2'-Deoxyuridine. Int J Mol Sci 2021; 22:ijms22041591. [PMID: 33562431 PMCID: PMC7914888 DOI: 10.3390/ijms22041591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Many microRNAs have been identified as critical mediators in the progression of melanoma through its regulation of genes involved in different cellular processes such as melanogenesis, cell cycle control, and senescence. However, microRNAs’ concurrent participation in syngeneic mouse B16F1 melanoma cells simultaneously induced decreased proliferation and differential pigmentation by exposure to 5-Brd-2′-dU (5’Bromo-2-deoxyuridine) and L-Tyr (L-Tyrosine) respectively, is poorly understood. Aim: To evaluate changes in the expression of microRNAs and identify which miRNAs in-network may contribute to the functional bases of phenotypes of differential pigmentation and reduction of proliferation in B16F1 melanoma cells exposed to 5-Brd-2′-dU and L-Tyr. Methods: Small RNAseq evaluation of the expression profiles of miRNAs in B16F1 melanoma cells exposed to 5-Brd-2′-dU (2.5 μg/mL) and L-Tyr (5 mM), as well as the expression by qRT-PCR of some molecular targets related to melanogenesis, cell cycle, and senescence. By bioinformatic analysis, we constructed network models of regulation and co-expression of microRNAs. Results: We confirmed that stimulation or repression of melanogenesis with L-Tyr or 5-Brd-2′-dU, respectively, generated changes in melanin concentration, reduction in proliferation, and changes in expression of microRNAs 470-3p, 470-5p, 30d-5p, 129-5p, 148b-3p, 27b-3p, and 211-5p, which presented patterns of coordinated and reciprocal co-expression, related to changes in melanogenesis through their putative targets Mitf, Tyr and Tyrp1, and control of cell cycle and senescence: Cyclin D1, Cdk2, Cdk4, p21, and p27. Conclusions: These findings provide insights into the molecular biology of melanoma of the way miRNAs are coordinated and reciprocal expression that may operate in a network as molecular bases for understanding changes in pigmentation and decreased proliferation induced in B16F1 melanoma cells exposed to L-Tyr and 5-Brd-2′-dU.
Collapse
Affiliation(s)
- Hernán Mauricio Rivera
- Department of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (H.M.R.); (E.N.M.)
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public Health Research, National Institute of Health, Bogotá 111321, Colombia
| | - Esther Natalia Muñoz
- Department of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (H.M.R.); (E.N.M.)
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public Health Research, National Institute of Health, Bogotá 111321, Colombia
| | - Daniel Osuna
- Science Department, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (D.O.); (M.F.); (M.C.)
| | - Mauro Florez
- Science Department, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (D.O.); (M.F.); (M.C.)
| | - Michael Carvajal
- Science Department, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (D.O.); (M.F.); (M.C.)
| | - Luis Alberto Gómez
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public Health Research, National Institute of Health, Bogotá 111321, Colombia
- Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
22
|
Carreira B, Acúrcio RC, Matos AI, Peres C, Pozzi S, Vaskovich‐Koubi D, Kleiner R, Bento M, Satchi‐Fainaro R, Florindo HF. Nanomedicines as Multifunctional Modulators of Melanoma Immune Microenvironment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ana I. Matos
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Mariana Bento
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| |
Collapse
|
23
|
Abstract
Since the first resection of melanoma by Hunter in 1787, efforts to treat patients with this deadly malignancy have been ongoing. Initial work to understand melanoma biology for therapeutics development began with the employment of isolated cancer cells grown in cell cultures. However, these models lack in vivo interactions with the tumor microenvironment. Melanoma cell line transplantation into suitable animals such as mice has been informative and useful for testing therapeutics as a preclinical model. Injection of freshly isolated patient melanomas into immunodeficient animals has shown the capacity to retain the genetic heterogeneity of the tumors, which is lost during the long-term culture of melanoma cells. Upon advancement of technology, genetically engineered animals have been generated to study the spontaneous development of melanomas in light of newly discovered genetic aberrations associated with melanoma formation. Culturing melanoma cells in a matrix generate tumor spheroids, providing an in vitro environment that promotes the heterogeneity commonplace with human melanoma and displaces the need for animal care facilities. Advanced 3D cultures have been created simulating the structure and cellularity of human skin to permit in vitro testing of therapeutics on melanomas expressing the same phenotype as demonstrated in vivo. This review will discuss these models and their relevance to the study of melanomagenesis, growth, metastasis, and therapy.
Collapse
Affiliation(s)
- Randal K Gregg
- Department of Basic Medical Sciences, DeBusk College of Osteopathic Medicine at Lincoln Memorial University-Knoxville, Knoxville, TN, USA.
| |
Collapse
|
24
|
Pratt EC, Isaac E, Stater EP, Yang G, Ouerfelli O, Pillarsetty N, Grimm J. Synthesis of the PET Tracer 124I-Trametinib for MAPK/ERK Kinase Distribution and Resistance Monitoring. J Nucl Med 2020; 61:1845-1850. [PMID: 32444378 PMCID: PMC9364901 DOI: 10.2967/jnumed.120.241901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/16/2020] [Indexed: 01/19/2023] Open
Abstract
Trametinib is an extremely potent allosteric inhibitor of mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated kinase (ERK) (MEK) 1/2, which has been approved for treatment of metastatic melanoma and anaplastic thyroid cancer in patients with confirmed BRAFV600E/K mutations. Though trametinib is highly efficacious, adverse side effects, including skin, gastrointestinal, and hepatic toxicity, are dose-limiting and can lead to treatment termination. Development of a noninvasive tool to visualize and quantify the delivery and distribution of trametinib (either as a single agent or in combination with other therapeutics) to tumors and organs would be helpful in assessing therapeutic index, personalizing individual dose, and potentially predicting resistance to therapy. Methods: To address these issues, we have developed a radiolabeled trametinib and evaluated the in vitro and in vivo properties. 123I-, 124I-, and 131I-trametinib, pure tracer analogs to trametinib, were synthesized in more than 95% purity, with an average yield of 69.7% and more than 100 GBq/μmol specific activity. Results: Overall, 124I-trametinib uptake in a panel of cancer cell lines can be blocked with cold trametinib, confirming specificity of the radiotracer in vitro and in vivo. 124I-trametinib was taken up at higher rates in KRAS and BRAF mutant cell lines than in wild-type KRAS cancer cell lines. In vivo, biodistribution revealed high uptake in the liver 2 h after injection, followed by clearance through the gastrointestinal tract over 4 d. Importantly, uptake higher than expected was observed in the lung and heart for up to 24 h. Peak uptake in the skin and gastrointestinal tract was observed between 6 and 24 h, whereas in B16F10 melanoma-bearing mice peak tumor concentrations were achieved between 24 and 48 h. Tumor uptake relative to muscle and skin was relatively low, peaking at 3.4- to 8.1-fold by 72 h, respectively. The biodistribution of 124I-trametinib was significantly reduced in mice on trametinib therapy, providing a quantitative method to observe MEK inhibition in vivo. Conclusion:124I-trametinib serves as an in vivo tool to personalize the dose instead of using the current single-fixed-dose scheme and, when combined with radiomic data, to monitor the emergence of therapy resistance. In addition, the production of iodinated trametinib affords researchers the ability to measure drug distribution for improved drug delivery studies.
Collapse
Affiliation(s)
- Edwin C. Pratt
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elizabeth Isaac
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evan P. Stater
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Guangbin Yang
- Organic Synthesis Core, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Ouathek Ouerfelli
- Organic Synthesis Core, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Nagavarakishore Pillarsetty
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan Grimm
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
25
|
Mahogunin Ring Finger 1 Is Required for Genomic Stability and Modulates the Malignant Phenotype of Melanoma Cells. Cancers (Basel) 2020; 12:cancers12102840. [PMID: 33019669 PMCID: PMC7599452 DOI: 10.3390/cancers12102840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 01/20/2023] Open
Abstract
Simple Summary Melanoma, the most aggressive skin cancer, accounts for the majority of deaths due to this disease. Therefore, identification of genes/proteins involved in melanoma genesis and/or progression is urgent. Mutations abrogating expression of Mahogunin Ring Finger 1 (MGRN1) in mice cause complex phenotypes with hyperpigmentation, and known MGRN1 interactors are important regulators of cell shape and movement. This suggests that MGRN1 may modulate the malignant phenotype of melanoma cells. Analysis of MGRN1-KO mouse melanocytes and melanoma cells showed that lack of MGRN1 leads to cell cycle defects and to a more differentiated, less aggressive phenotype, with increased adhesion to various matrices, decreased motility and high genomic instability. The higher aggressivity of MGRN1-expressing melanoma cells was confirmed in an in vivo mouse melanoma model and is consistent with higher survival of human melanoma patients expressing low levels of MGRN1. Therefore, MGRN1 appears an important determinant of the malignant phenotype of melanoma. Abstract The mouse mahoganoid mutation abrogating Mahogunin Ring Finger-1 (MGRN1) E3 ubiquitin ligase expression causes hyperpigmentation, congenital heart defects and neurodegeneration. To study the pathophysiology of MGRN1 loss, we compared Mgrn1-knockout melanocytes with genetically matched controls and melan-md1 (mahoganoid) melanocytes. MGRN1 knockout induced a more differentiated and adherent phenotype, decreased motility, increased the percentage of cells in the S phase of the cell cycle and promoted genomic instability, as shown by stronger γH2AX labelling, increased burden of DNA breaks and higher abundance of aneuploid cells. Lack of MGRN1 expression decreased the ability of melanocytes to cope with DNA breaks generated by oxidizing agents or hydroxyurea-induced replicative stress, suggesting a contribution of genomic instability to the mahoganoid phenotype. MGRN1 knockout in B16-F10 melanoma cells also augmented pigmentation, increased cell adhesion to collagen, impaired 2D and 3D motility and caused genomic instability. Tumors formed by Mgrn1-KO B16-F10 cells had lower mitotic indices, fewer Ki67-positive cells and showed a trend towards smaller size. In short-term lung colonization assays Mgrn1-KO cells showed impaired colonization potential. Moreover, lower expression of MGRN1 is significantly associated with better survival of human melanoma patients. Therefore, MGRN1 might be an important phenotypic determinant of melanoma cells.
Collapse
|
26
|
Bognar Z, Cseh AM, Fekete K, Antus C, Bognar R, Tapodi A, Ramadan FHJ, Sumegi B, Gallyas F. Amiodarone's major metabolite, desethylamiodarone inhibits proliferation of B16-F10 melanoma cells and limits lung metastasis formation in an in vivo experimental model. PLoS One 2020; 15:e0239088. [PMID: 32977329 PMCID: PMC7518930 DOI: 10.1371/journal.pone.0239088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/30/2020] [Indexed: 12/27/2022] Open
Abstract
Previously, we demonstrated the in vitro anti-tumor effects of desethylamiodarone (DEA) in bladder and cervix cancer cell lines. In the present study, we intended to establish its potentiality in B16-F10 metastatic melanoma cells in vitro and in vivo. We assessed cell proliferation, apoptosis and cell cycle by using sulforhodamine B assay, Muse™ Annexin V & Dead Cell and Muse® Cell Cycle assays, respectively. We determined colony formation after crystal violet staining. For studying mechanistic aspects, immunoblotting analysis was performed. We used a C57BL/6 experimental lung metastasis model for demonstrating in vivo anti-metastatic potential of DEA. DEA inhibited in vitro proliferation and colony formation, and in vivo lung metastasizing properties of B16-F10 cells. It arrested the cells in G0/G1 phase of their cycle likely via p21 in a p53-dependent fashion, and induced caspase mediated apoptosis likely via inversely regulating Bcl-2 and Bax levels, and reducing Akt and ERK1/2 activation. In this study, we provided in vitro and in vivo experimental evidences for DEA’s potentiality in the therapy of metastatic melanomas. Since DEA is the major metabolite of amiodarone, a worldwide used antiarrhythmic drug, safety concerns could be resolved more easily for it than for a novel pharmacological agent.
Collapse
Affiliation(s)
- Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- * E-mail:
| | - Anna Maria Cseh
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Csenge Antus
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Rita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Antal Tapodi
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Fadi H. J. Ramadan
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- MTA-PTE Nuclear-Mitochondrial Interactions Research Group, Pecs, Hungary
- Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- MTA-PTE Nuclear-Mitochondrial Interactions Research Group, Pecs, Hungary
- Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| |
Collapse
|
27
|
COL2A1 Is a Novel Biomarker of Melanoma Tumor Repopulating Cells. Biomedicines 2020; 8:biomedicines8090360. [PMID: 32962144 PMCID: PMC7555262 DOI: 10.3390/biomedicines8090360] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/09/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Soft 3D-fibrin-gel selected tumor repopulating cells (TRCs) from the B16F1 melanoma cell line exhibit extraordinary self-renewal and tumor-regeneration capabilities. However, their biomarkers and gene regulatory features remain largely unknown. Here, we utilized the next-generation sequencing-based RNA sequencing (RNA-seq) technique to discover novel biomarkers and active gene regulatory features of TRCs. Systems biology analysis of RNA-seq data identified differentially expressed gene clusters, including the cell adhesion cluster, which subsequently identified highly specific and novel biomarkers, such as Col2a1, Ncam1, F11r, and Negr1. We validated the expression of these genes by real-time qPCR. The expression level of Col2a1 was found to be relatively low in TRCs but twenty-fold higher compared to the parental control cell line, thus making the biomarker very specific for TRCs. We validated the COL2A1 protein by immunofluorescence microscopy, showing a higher expression of COL2A1 in TRCs compared to parental control cells. KEGG pathway analysis showed the JAK/STAT, hypoxia, and Akt signaling pathways to be active in TRCs. Besides, the aerobic glycolysis pathway was found to be very active, indicating a typical Warburg Effect on highly tumorigenic cells. Together, our study revealed highly specific biomarkers and active cell signaling pathways of melanoma TRCs that can potentially target and neutralize TRCs.
Collapse
|
28
|
Duval KEA, Vernice NA, Wagner RJ, Fiering SN, Petryk JD, Lowry GJ, Tau SS, Yin J, Houde GR, Chaudhry AS, Hoopes PJ. Immunogenetic effects of low dose (CEM43 30) magnetic nanoparticle hyperthermia and radiation in melanoma cells. Int J Hyperthermia 2020; 36:37-46. [PMID: 31795829 PMCID: PMC6943912 DOI: 10.1080/02656736.2019.1627433] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: In this in vitro study we have used an RNA quantification technique, nanoString, and a conventional protein analysis technique (Western Blot) to assess the genetic and protein expression of B16 murine melanoma cells following a modest magnetic nanoparticle hyperthermia (mNPH) dose equivalent to 30 minutes @ 43°C (CEM43 30) and/or a clinically relevant 8 Gy radiation dose. Methods: Melanoma cells with mNPs(2.5 μg Fe/106 cells) were pelleted and exposed to an alternating magnetic field (AMF) to generate the targeted thermal dose. Thermal dose was accurately monitored by a fiber optic probe and automatically maintained at CEM43 30. All cells were harvested 24 hours after treatment. Results: The mNPH dose demonstrated notable elevations in the thermotolerance/immunogenic HSP70 gene and a number of chemoattractant and toll-like receptor gene pathways. The 8 Gy dose also upregulated a number of important immune and cytotoxic genetic and protein pathways. However, the mNPH/radiation combination was the most effective stimulator of a wide variety of immune and cytotoxic genes including HSP70, cancer regulating chemokines CXCL10, CXCL11, the T-cell trafficking chemokine CXCR3, innate immune activators TLR3, TLR4, the MDM2 and mTOR negative regulator of p53, the pro-apoptotic protein PUMA, and the cell death receptor Fas. Importantly a number of the genetic changes were accurately validated by protein expression changes, i.e., HSP70, p-mTOR, p-MDM2. Conclusion: These results not only show that low dose mNPH and radiation independently increase the expression of important immune and cytotoxic genes but that the effect is greatly enhanced when they are used in combination.
Collapse
Affiliation(s)
- Kayla E A Duval
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | | | - Robert J Wagner
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - James D Petryk
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - Steven S Tau
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - John Yin
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Georgia R Houde
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
29
|
Odnokoz O, Yu P, Peck AR, Sun Y, Kovatich AJ, Hooke JA, Hu H, Mitchell EP, Rui H, Fuchs SY. Malignant cell-specific pro-tumorigenic role of type I interferon receptor in breast cancers. Cancer Biol Ther 2020; 21:629-636. [PMID: 32378445 DOI: 10.1080/15384047.2020.1750297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Within the microenvironment of solid tumors, stress associated with deficit of nutrients and oxygen as well as tumor-derived factors triggers the phosphorylation-dependent degradation of the IFNAR1 chain of type I interferon (IFN1) receptor and ensuing suppression of the IFN1 pathway. Here we sought to examine the importance of these events in malignant mammary cells. Expression of non-degradable IFNAR1S526A mutant in mouse mammary adenocarcinoma cells stimulated the IFN1 pathway yet did not affect growth of these cells in vitro or ability to form subcutaneous tumors in the syngeneic mice. Remarkably, these cells exhibited a notably accelerated growth when transplanted orthotopically into mammary glands. Importantly, in human patients with either ER+ or ER- breast cancers, high levels of IFNAR1 were associated with poor prognosis. We discuss the putative mechanisms underlying the pro-tumorigenic role of IFNAR1 in malignant breast cells.
Collapse
Affiliation(s)
- Olena Odnokoz
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - Pengfei Yu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - Amy R Peck
- Department of Pathology, Medical College of Wisconsin , Milwaukee, WI, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin , Milwaukee, WI, USA
| | - Albert J Kovatich
- John P. Murtha Cancer Center Research Program, Uniformed Services University and Walter Reed National Military Medical Center , Bethesda, MD, USA
| | - Jeffrey A Hooke
- John P. Murtha Cancer Center Research Program, Uniformed Services University and Walter Reed National Military Medical Center , Bethesda, MD, USA
| | - Hai Hu
- Chan Soon-Shiong Institute of Molecular Medicine , Windber, PA, USA
| | - Edith P Mitchell
- Department of Medical Oncology, Thomas Jefferson University , Philadelphia, PA, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin , Milwaukee, WI, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA, USA
| |
Collapse
|
30
|
Zhuo M, Gorgun FM, Tyler DS, Englander EW. Hypoxia potentiates the capacity of melanoma cells to evade cisplatin and doxorubicin cytotoxicity via glycolytic shift. FEBS Open Bio 2020; 10:789-801. [PMID: 32134564 PMCID: PMC7193165 DOI: 10.1002/2211-5463.12830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/27/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
The hypoxic environment within solid tumors impedes the efficacy of chemotherapeutic treatments. Here, we demonstrate that hypoxia augments the capacity of melanoma cells to withstand cisplatin and doxorubicin cytotoxicity. We show that B16F10 cells derived from spontaneously formed melanoma and YUMM1.7 cells, engineered to recapitulate human‐relevant melanoma driver mutations, profoundly differ in their vulnerabilities to cisplatin and doxorubicin. The differences are manifested in magnitude of proliferative arrest and cell death rates, extent of mtDNA depletion, and impairment of mitochondrial respiration. In both models, cytotoxicity is mitigated by hypoxia, which augments glycolytic metabolism. Collectively, the findings implicate metabolic reprogramming in drug evasion and suggest that melanoma tumors with distinct genetic makeup may have differential drug vulnerabilities, highlighting the importance of precision anticancer treatments.
Collapse
Affiliation(s)
- Ming Zhuo
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| | - Falih M. Gorgun
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| | - Douglas S. Tyler
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| | - Ella W. Englander
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
31
|
Markman JL, Porritt RA, Wakita D, Lane ME, Martinon D, Noval Rivas M, Luu M, Posadas EM, Crother TR, Arditi M. Loss of testosterone impairs anti-tumor neutrophil function. Nat Commun 2020; 11:1613. [PMID: 32235862 PMCID: PMC7109066 DOI: 10.1038/s41467-020-15397-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
In men, the incidence of melanoma rises rapidly after age 50, and nearly two thirds of melanoma deaths are male. The immune system is known to play a key role in controlling the growth and spread of malignancies, but whether age- and sex-dependent changes in immune cell function account for this effect remains unknown. Here, we show that in castrated male mice, neutrophil maturation and function are impaired, leading to elevated metastatic burden in two models of melanoma. Replacement of testosterone effectively normalized the tumor burden in castrated male mice. Further, the aberrant neutrophil phenotype was also observed in prostate cancer patients receiving androgen deprivation therapy, highlighting the evolutionary conservation and clinical relevance of the phenotype. Taken together, these results provide a better understanding of the role of androgen signaling in neutrophil function and the impact of this biology on immune control of malignancies. It is known that there are sex differences in the incidence and prognosis of certain cancers, including melanoma. In this study, the authors utilize a melanoma model to reveal that castrated mice have a higher metastatic burden associated with androgen dependent impaired neutrophil function.
Collapse
Affiliation(s)
- Janet L Markman
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rebecca A Porritt
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Daiko Wakita
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Malcolm E Lane
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Daisy Martinon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Sciences, Infectious and Immunologic Disease Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Science, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Michael Luu
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Edwin M Posadas
- Urologic Oncology Program/Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Center Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Timothy R Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Sciences, Infectious and Immunologic Disease Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Biomedical Science, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,Department of Biomedical Sciences, Infectious and Immunologic Disease Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,Department of Biomedical Science, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
32
|
Cheng ZY, Hsiao YT, Huang YP, Peng SF, Huang WW, Liu KC, Hsia TC, Way TD, Chung JG. Casticin Induces DNA Damage and Affects DNA Repair Associated Protein Expression in Human Lung Cancer A549 Cells (Running Title: Casticin Induces DNA Damage in Lung Cancer Cells). Molecules 2020; 25:E341. [PMID: 31952105 PMCID: PMC7024307 DOI: 10.3390/molecules25020341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022] Open
Abstract
Casticin was obtained from natural plants, and it has been shown to exert biological functions; however, no report concerns the induction of DNA damage and repair in human lung cancer cells. The objective of this study was to investigate the effects and molecular mechanism of casticin on DNA damage and repair in human lung cancer A549 cells. Cell viability was determined by flow cytometric assay. The DNA damage was evaluated by 4',6-diamidino-2-phenylindole (DAPI) staining and electrophoresis which included comet assay and DNA gel electrophoresis. The protein levels associated with DNA damage and repair were analyzed by western blotting. The expression and translocation of p-H2A.X were observed by confocal laser microscopy. Casticin reduced total viable cell number and induced DNA condensation, fragmentation, and damage in A549 cells. Furthermore, casticin increased p-ATM at 6 h and increased p-ATR and BRCA1 at 6-24 h treatment but decreased p-ATM at 24-48 h, as well as decreased p-ATR and BRCA1 at 48 h. Furthermore, casticin decreased p-p53 at 6-24 h but increased at 48 h. Casticin increased p-H2A.X and MDC1 at 6-48 h treatment. In addition, casticin increased PARP (cleavage) at 6, 24, and 48 h treatment, DNA-PKcs and MGMT at 48 h in A549 cells. Casticin induced the expressions and nuclear translocation of p-H2AX in A549 cells by confocal laser microscopy. Casticin reduced cell number through DNA damage and condensation in human lung cancer A549 cells.
Collapse
Affiliation(s)
- Zheng-Yu Cheng
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Kuo-Ching Liu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan;
| | - Te-Chun Hsia
- Department of Respiratory Therapy, China Medical University, Taichung 404, Taiwan;
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Tzong-Der Way
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| |
Collapse
|
33
|
Mitofusins modulate the increase in mitochondrial length, bioenergetics and secretory phenotype in therapy-induced senescent melanoma cells. Biochem J 2019; 476:2463-2486. [PMID: 31431479 PMCID: PMC6735661 DOI: 10.1042/bcj20190405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 01/03/2023]
Abstract
Cellular senescence is an endpoint of chemotherapy, and targeted therapies in melanoma and the senescence-associated secretory phenotype (SASP) can affect tumor growth and microenvironment, influencing treatment outcomes. Metabolic interventions can modulate the SASP, and an enhanced mitochondrial energy metabolism supports resistance to therapy in melanoma cells. Herein, we assessed the mitochondrial function of therapy-induced senescent melanoma cells obtained after exposing the cells to temozolomide (TMZ), a methylating chemotherapeutic agent. Senescence induction in melanoma was accompanied by a substantial increase in mitochondrial basal, ATP-linked, and maximum respiration rates and in coupling efficiency, spare respiratory capacity, and respiratory control ratio. Further examinations revealed an increase in mitochondrial mass and length. Alterations in mitochondrial function and morphology were confirmed in isolated senescent cells, obtained by cell-size sorting. An increase in mitofusin 1 and 2 (MFN1 and 2) expression and levels was observed in senescent cells, pointing to alterations in mitochondrial fusion. Silencing mitofusin expression with short hairpin RNA (shRNA) prevented the increase in mitochondrial length, oxygen consumption rate and secretion of interleukin 6 (IL-6), a component of the SASP, in melanoma senescent cells. Our results represent the first in-depth study of mitochondrial function in therapy-induced senescence in melanoma. They indicate that senescence increases mitochondrial mass, length and energy metabolism; and highlight mitochondria as potential pharmacological targets to modulate senescence and the SASP.
Collapse
|
34
|
Patel P, Sun L, Robbins Y, Clavijo PE, Friedman J, Silvin C, Van Waes C, Cook J, Mitchell J, Allen C. Enhancing direct cytotoxicity and response to immune checkpoint blockade following ionizing radiation with Wee1 kinase inhibition. Oncoimmunology 2019; 8:e1638207. [PMID: 31646086 DOI: 10.1080/2162402x.2019.1638207] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/31/2022] Open
Abstract
Tumor cells activate the G2/M cell cycle checkpoint in response to ionizing radiation (IR) and effector immune cell-derived granzyme B to facilitate repair and survival. Wee1 kinase inhibition reverses the ability of tumor cells to pause at G2/M. Here, we hypothesized that AZD1775, a small molecule inhibitor of Wee1 kinase, could sensitize tumor cells to IR and T-lymphocyte killing and improve responses to combination IR and programmed death (PD)-axis immune checkpoint blockade (ICB). Multiple models of head and neck carcinoma, lung carcinoma and melanoma were used in vitro and in vivo to explore this hypothesis. AZD1775 reversed G2/M cell cycle checkpoint activation following IR, inducing cell death. Combination IR and AZD1775 induced accumulation of DNA damage in M-phase cells and was rescued with nucleoside supplementation, indicating mitotic catastrophe. Combination treatment enhanced control of syngeneic MOC1 tumors in vivo, and on-target effects of systemic AZD1775 could be localized with targeted IR. Combination treatment enhanced granzyme B-dependent T-lymphocyte killing through reversal of additive G2/M cell cycle block induced by IR and granzyme B. Combination IR and AZ1775-enhanced CD8+ cell-dependent MOC1 tumor growth control and rate of complete rejection of established tumors in the setting of PD-axis ICB. Functional assays demonstrated increased tumor antigen-specific immune responses in sorted T-lymphocytes. The combination of IR and AZD1775 not only lead to enhanced tumor-specific cytotoxicity, it also enhanced susceptibility to T-lymphocyte killing and responses to PD-axis ICB. These data provide the pre-clinical rationale for the combination of these therapies in the clinical trial setting.
Collapse
Affiliation(s)
- Priya Patel
- Translational Tumor Immunology Program, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Lily Sun
- Translational Tumor Immunology Program, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Yvette Robbins
- Translational Tumor Immunology Program, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Paul E Clavijo
- Translational Tumor Immunology Program, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Jay Friedman
- Translational Tumor Immunology Program, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Silvin
- Tumor Biology Section, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Carter Van Waes
- Tumor Biology Section, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - John Cook
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James Mitchell
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clint Allen
- Translational Tumor Immunology Program, National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD, USA.,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
35
|
Couto GK, Segatto NV, Oliveira TL, Seixas FK, Schachtschneider KM, Collares T. The Melding of Drug Screening Platforms for Melanoma. Front Oncol 2019; 9:512. [PMID: 31293965 PMCID: PMC6601395 DOI: 10.3389/fonc.2019.00512] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022] Open
Abstract
The global incidence of cancer is rising rapidly and continues to be one of the leading causes of death in the world. Melanoma deserves special attention since it represents one of the fastest growing types of cancer, with advanced metastatic forms presenting high mortality rates due to the development of drug resistance. The aim of this review is to evaluate how the screening of drugs and compounds for melanoma has been performed over the last seven decades. Thus, we performed literature searches to identify melanoma drug screening methods commonly used by research groups during this timeframe. In vitro and in vivo tests are essential for the development of new drugs; however, incorporation of in silico analyses increases the possibility of finding more suitable candidates for subsequent tests. In silico techniques, such as molecular docking, represent an important and necessary first step in the screening process. However, these techniques have not been widely used by research groups to date. Our research has shown that the vast majority of research groups still perform in vitro and in vivo tests, with emphasis on the use of in vitro enzymatic tests on melanoma cell lines such as SKMEL and in vivo tests using the B16 mouse model. We believe that the union of these three approaches (in silico, in vitro, and in vivo) is essential for improving the discovery and development of new molecules with potential antimelanoma action. This workflow would provide greater confidence and safety for preclinical trials, which will translate to more successful clinical trials and improve the translatability of new melanoma treatments into clinical practice while minimizing the unnecessary use of laboratory animals under the principles of the 3R's.
Collapse
Affiliation(s)
- Gabriela Klein Couto
- Research Group in Molecular and Cellular Oncology, Postgraduate Program in Biochemistry and Bioprospecting, Cancer Biotechnology Laboratory, Center for Technological Development, Federal University of Pelotas, Pelotas, Brazil
| | - Natália Vieira Segatto
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| | - Thaís Larré Oliveira
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| | - Fabiana Kömmling Seixas
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Tiago Collares
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| |
Collapse
|
36
|
Paskaš S, Krajnović T, Basile MS, Dunđerović D, Cavalli E, Mangano K, Mammana S, Al-Abed Y, Nicoletti F, Mijatović S, Maksimović-Ivanić D. Senescence as a main mechanism of Ritonavir and Ritonavir-NO action against melanoma. Mol Carcinog 2019; 58:1362-1375. [PMID: 30997718 DOI: 10.1002/mc.23020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022]
Abstract
The main focus of this study is exploring the effect and mechanism of two HIV-protease inhibitors: Ritonavir and Ritonavir-nitric oxide (Ritonavir-NO) on in vitro growth of melanoma cell lines. NO modification significantly improved the antitumor potential of Ritonavir, as the IC50 values of Ritonavir-NO were approximately two times lower than IC50 values of the parental compound. Our results showed for the first time, that both compounds induced senescence in primary and metastatic melanoma cell lines. This transformation was manifested as a change in cell morphology, enlargement of nuclei, increased cellular granulation, upregulation of β-galactosidase activity, lipofuscin granules appearance, higher production of reactive oxygen species and persistent inhibition of proliferation. The expression of p53, as one of the key regulators of senescence, was upregulated after 48 hours of Ritonavir-NO treatment only in metastatic B16F10 cells, ranking it as a late-response event. The development of senescent phenotype was consistent with the alteration of the cytoskeleton-as we observed diminished expression of vinculin, α-actin, and β-tubulin. Permanent inhibition of S6 protein by Ritonavir-NO, but not Ritonavir, could be responsible for a stronger antiproliferative potential of the NO-modified compound. Taken together, induction of senescent phenotype may provide an excellent platform for developing therapeutic approaches based on selective killing of senescent cells.
Collapse
Affiliation(s)
- Svetlana Paskaš
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Tamara Krajnović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Maria S Basile
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Duško Dunđerović
- Institute of Pathology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Eugenio Cavalli
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Santa Mammana
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| |
Collapse
|
37
|
5-Arylidene(chromenyl-methylene)-thiazolidinediones: Potential New Agents against Mutant Oncoproteins K-Ras, N-Ras and B-Raf in Colorectal Cancer and Melanoma. ACTA ACUST UNITED AC 2019; 55:medicina55040085. [PMID: 30935124 PMCID: PMC6524019 DOI: 10.3390/medicina55040085] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/05/2019] [Accepted: 03/28/2019] [Indexed: 01/16/2023]
Abstract
Background and objectives: Cancer represents the miscommunication between and within the body cells. The mutations of the oncogenes encoding the MAPK pathways play an important role in the development of tumoral diseases. The mutations of KRAS and BRAF oncogenes are involved in colorectal cancer and melanoma, while the NRAS mutations are associated with melanoma. Thiazolidine-2,4-dione is a versatile scaffold in medicinal chemistry and a useful tool in the development of new antitumoral compounds. The aim of our study was to predict the pharmacokinetic/pharmacodynamic properties, the drug-likeness and lead-likeness of two series of synthetic 5-arylidene(chromenyl-methylene)-thiazolidinediones, the molecular docking on the oncoproteins K-Ras, N-Ras and B-Raf, and to investigate the cytotoxicity of the compounds, in order to select the best structural profile for potential anticancer agents. Materials and Methods: In our paper we studied the cytotoxicity of two series of thiazolidine-2,4-dione derivatives, their ADME-Tox properties and the molecular docking on a mutant protein of K-Ras, two isoforms of N-Ras and an isoform of B-Raf with 16 mutations. Results: The heterocyclic compounds strongly interact with K-Ras and N-Ras right after their posttranslational processing and/or compete with GDP for the nucleotide-binding site of the two GTPases. They are less active against the GDP-bound states of the two targets. All derivatives have a similar binding pattern in the active site of B-Raf. Conclusions: The data obtained encourage the further investigation of the 5-arylidene(chromenyl-methylene)-thiazolidinediones as potential new agents against the oncoproteins K-Ras, N-Ras and B-Raf.
Collapse
|
38
|
Li K, Xiao G, Richardson JJ, Tardy BL, Ejima H, Huang W, Guo J, Liao X, Shi B. Targeted Therapy against Metastatic Melanoma Based on Self-Assembled Metal-Phenolic Nanocomplexes Comprised of Green Tea Catechin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801688. [PMID: 30886799 PMCID: PMC6402403 DOI: 10.1002/advs.201801688] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/09/2018] [Indexed: 02/05/2023]
Abstract
The targeted therapy of metastatic melanoma is an important yet challenging goal that has received only limited attention to date. Herein, green tea polyphenols, (-)-epigallocatechin-3-gallate (EGCG), and lanthanide metal ions (Sm3+) are used as building blocks to engineer self-assembled SmIII-EGCG nanocomplexes with synergistically enhanced tumor inhibitory properties. These nanocomplexes have negligible systemic toxic effects on healthy cells but cause a significant reduction in the viability of melanoma cells by efficiently regulating their metabolic pathways. Moreover, the wound-induced migration of melanoma cells can be efficiently inhibited by SmIII-EGCG, which is a key criterion for metastatic melanoma therapy. In a mouse melanoma tumor model, SmIII-EGCG is directly compared with a clinical anticancer drug, 5-fluorouracil and shows remarkable tumor inhibition. Moreover, the targeted therapy of SmIII-EGCG is shown to prevent metastatic lung melanoma from spreading to main organs with no adverse side effects on the body weight or organs. These in vivo results demonstrate significant advantages of SmIII-EGCG over its clinical counterpart. The results suggest that these green tea-based, self-assembled nanocomplexes possess all of the key traits of a clinically promising candidate to address the challenges associated with the treatment of advanced stage metastatic melanoma.
Collapse
Affiliation(s)
- Ke Li
- Department of Biomass Chemistry and EngineeringSichuan UniversityChengdu610065China
- Laboratory of EthnopharmacologyRegenerative Medicine Research CenterWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Gao Xiao
- Wyss Institute for Biologically Inspired EngineeringJohn A. Paulson School of Engineering and Applied SciencesHarvard UniversityBostonMA02115USA
- Department of Environmental Science and EngineeringCollege of Environment and ResourcesFuzhou UniversityFuzhou350108China
| | - Joseph J. Richardson
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology and Department of Chemical and Biomolecular EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Blaise L. Tardy
- Department of Bioproducts and BiosystemsSchool of Chemical EngineeringAalto UniversityP. O. Box 1630000076Finland
| | - Hirotaka Ejima
- Department of Materials EngineeringThe University of Tokyo7‐3‐1 HongoBunkyo‐kuTokyo113‐8656Japan
| | - Wen Huang
- Laboratory of EthnopharmacologyRegenerative Medicine Research CenterWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Junling Guo
- Department of Biomass Chemistry and EngineeringSichuan UniversityChengdu610065China
- Wyss Institute for Biologically Inspired EngineeringJohn A. Paulson School of Engineering and Applied SciencesHarvard UniversityBostonMA02115USA
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
| | - Xuepin Liao
- Department of Biomass Chemistry and EngineeringSichuan UniversityChengdu610065China
| | - Bi Shi
- Department of Biomass Chemistry and EngineeringSichuan UniversityChengdu610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
| |
Collapse
|
39
|
Calabrese EJ. The additive to background assumption in cancer risk assessment: A reappraisal. ENVIRONMENTAL RESEARCH 2018; 166:175-204. [PMID: 29890424 DOI: 10.1016/j.envres.2018.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 05/21/2023]
Abstract
The assumption that chemical and radiation induced cancers act in a manner that is additive to background was proposed in the mid-1970s. It was adopted by the U.S. Environmental Protection Agency (EPA) in 1986 and then subsequently by other regulatory agencies worldwide for cancer risk assessment. It ensured that cancer risks at low doses act in a linear fashion. The additive to background process assumes that the mechanism(s) resulting in induced (i.e., treatment related) and spontaneous (i.e., control group) cancers are identical. This assumption could not be properly evaluated due to inadequate mechanistic data when it was proposed in the 1970s. Using the findings of modern molecular toxicology, including oncogene activation/mutation, gene regulation, and molecular pathway analyses, the additive to background assumption was evaluated in the present paper. Based on published studies with 45 carcinogens over 13 diverse mammalian models and for a broad range of tumor types compelling evidence indicates that carcinogen-induced tumors are mediated in general via mechanisms that are not identical to those affecting the occurrence of the same type of spontaneous tumors in appropriate control groups. These findings, which challenge a fundamental assumption of the additive to background concept, have significant implications for cancer risk assessment policy, regulatory agency practices, as well as fundamental concepts of cancer biology.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA 01003, United States.
| |
Collapse
|
40
|
Csoboz B, Gombos I, Tatrai E, Tovari J, Kiss AL, Horvath I, Vigh L. Chemotherapy induced PRL3 expression promotes cancer growth via plasma membrane remodeling and specific alterations of caveolae-associated signaling. Cell Commun Signal 2018; 16:51. [PMID: 30157875 PMCID: PMC6116440 DOI: 10.1186/s12964-018-0264-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The outcome of cancer therapy is greatly defined by the ability of a tumor cell to evade treatment and re-establish its bulk mass after medical interventions. Consequently, there is an urgent need for the characterization of molecules affecting tumor reoccurrence. The phosphatase of regenerating liver 3 (PRL3) protein was recently emerged among the targets that could affect such a phenomenon. METHODS The expression induction of PRL3 in melanoma cells treated with chemotherapeutic agents was assessed by western blotting. The effect of PRL3 expression on cancer growth was investigated both in vitro and in vivo. The association of PRL3 with the caveolae structures of the plasma membrane was analyzed by detergent free raft purification. The effect of PRL3 expression on the membrane organization was assayed by electron microscopy and by membrane biophysical measurements. Purification of the plasma membrane fraction and co-immunoprecipitation were used to evaluate the altered protein composition of the plasma membrane upon PRL3 expression. RESULTS Here, we identified PRL3 as a genotoxic stress-induced oncogene whose expression is significantly increased by the presence of classical antitumor therapeutics. Furthermore, we successfully connected the presence of this oncogene with increased tumor growth, which implies that tumor cells can utilize PRL3 effects as a survival strategy. We further demonstrated the molecular mechanism that is connected with the pro-growth action of PRL3, which is closely associated with its localization to the caveolae-type lipid raft compartment of the plasma membrane. In our study, PRL3 was associated with distinct changes in the plasma membrane structure and in the caveolar proteome, such as the dephosphorylation of integrin β1 at Thr788/Thr789 and the increased partitioning of Rac1 to the plasma membrane. These alterations at the plasma membrane were further associated with the elevation of cyclin D1 in the nucleus. CONCLUSIONS This study identifies PRL3 as an oncogene upregulated in cancer cells upon exposure to anticancer therapeutics. Furthermore, this work contributes to the existing knowledge on PRL3 function by characterizing its association with the caveolae-like domains of the plasma membrane and their resident proteins.
Collapse
Affiliation(s)
- Balint Csoboz
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvari Krt. 62, Szeged, 6726, Hungary.
| | - Imre Gombos
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvari Krt. 62, Szeged, 6726, Hungary
| | - Eniko Tatrai
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, 1094, Hungary
| | - Jozsef Tovari
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, 1094, Hungary
| | - Anna L Kiss
- Department of Anatomy, Histology and Embryology, Semmelweis University Budapest, Budapest, 1094, Hungary
| | - Ibolya Horvath
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvari Krt. 62, Szeged, 6726, Hungary
| | - Laszlo Vigh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvari Krt. 62, Szeged, 6726, Hungary.
| |
Collapse
|
41
|
Sun L, Moore E, Berman R, Clavijo PE, Saleh A, Chen Z, Van Waes C, Davies J, Friedman J, Allen CT. WEE1 kinase inhibition reverses G2/M cell cycle checkpoint activation to sensitize cancer cells to immunotherapy. Oncoimmunology 2018; 7:e1488359. [PMID: 30288354 DOI: 10.1080/2162402x.2018.1488359] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Indexed: 01/29/2023] Open
Abstract
Intrinsic resistance to cytotoxic T-lymphocyte (CTL) killing limits responses to immune activating anti-cancer therapies. Here, we established that activation of the G2/M cell cycle checkpoint results in tumor cell cycle pause and protection from granzyme B-induced cell death. This was reversed with WEE1 kinase inhibition, leading to enhanced CTL killing of antigen-positive tumor cells. Similarly, but at a later time point, cell cycle pause following TNFα exposure was reversed with WEE1 kinase inhibition, leading to CTL transmembrane TNFα-dependent induction of apoptosis and necroptosis in bystander antigen-negative tumor cells. Results were reproducible in models of oral cavity carcinoma, melanoma and colon adenocarcinoma harboring variable Tp53 genomic alterations. WEE1 kinase inhibition sensitized tumors to PD-1 mAb immune checkpoint blockade in vivo, resulting in CD8+-dependent rejection of established tumors harboring antigen-positive or mixed antigen-positive and negative tumor cells. Together, these data describe activation of the G2/M cell cycle checkpoint in response to early and late CTL products as a mechanism of resistance to CTL killing, and provide pre-clinical rationale for the clinical combination of agents that inhibit cell cycle checkpoints and activate anti-tumor immunity.
Collapse
Affiliation(s)
- Lillian Sun
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Ellen Moore
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Rose Berman
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Paul E Clavijo
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Anthony Saleh
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - John Davies
- Experimental Transplantation and Immunology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Jay Friedman
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Clint T Allen
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| |
Collapse
|
42
|
Li Y, Yue Z, Yu H, Liu X, Tao L, Zhu Z, Fan F, Shen C, Wang A, Chen W, Lu Y. A spontaneous metastatic mathematical model in mice for screening anti-metastatic agents. J Pharmacol Toxicol Methods 2018; 92:57-66. [DOI: 10.1016/j.vascn.2018.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 03/08/2018] [Accepted: 03/10/2018] [Indexed: 10/17/2022]
|
43
|
Moore EC, Sun L, Clavijo PE, Friedman J, Harford JB, Saleh AD, Van Waes C, Chang EH, Allen CT. Nanocomplex-based TP53 gene therapy promotes anti-tumor immunity through TP53- and STING-dependent mechanisms. Oncoimmunology 2018; 7:e1404216. [PMID: 29900037 PMCID: PMC5993490 DOI: 10.1080/2162402x.2017.1404216] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/30/2017] [Accepted: 11/06/2017] [Indexed: 11/17/2022] Open
Abstract
Loss or mutation of TP53 has been linked to alterations in anti-tumor immunity as well as dysregulation of cell cycle and apoptosis. We explored immunologic effects and mechanisms following restoration of wild-type human TP53 cDNA in murine oral cancer cells using the therapeutic nanocomplex scL-53. We demonstrated scL-53 induces dose-dependent expression of TP53 and induction of apoptosis and immunogenic cell death. We further demonstrated both TP53-dependent and independent induction of tumor cell immunogenicity through the use of blocking mAbs, nanocomplex loaded with DNA plasmid with or without TP53 cDNA, empty nanocomplex and siRNA knockdown techniques. TP53-independent immune modulation was observed following treatment with nanocomplex loaded with DNA plasmid lacking TP53 cDNA and abrogated in STING-deficient tumor cells, supporting the presence of a cytoplasmic DNA sensing, STING-dependent type-I IFN response. Cooperatively, TP53- and STING-dependent alterations sensitized tumor cells to CTL-mediated lysis, which was further enhanced following reversal of adaptive immune resistance with PD-1 mAb. In vivo, combination scL-53 and PD-1 mAb resulted in growth control or rejection of established tumors that was abrogated in mice depleted of CD8+ cells or in STING deficient mice. Cumulatively, this work demonstrates 1) a direct anti-tumor effects of functional TP53; 2) non-redundant TP53- and STING-dependent induction of tumor cell immunogenicity following scL-53 treatment; and 3) that adaptive immune resistance following scL-53 treatment can be reversed with PD-based immune checkpoint blockade, resulting in the rejection or control of syngeneic murine tumors. These data strongly support the clinical combination of scL-53 and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ellen C Moore
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Lillian Sun
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Paul E Clavijo
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Jay Friedman
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | | | - Anthony D Saleh
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Esther H Chang
- SynerGene Therapeutics, Potomac, MD, USA.,Georgetown University Medical Center, Washington, DC, USA
| | - Clint T Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| |
Collapse
|
44
|
Kalal BS, Fathima F, Pai VR, Sanjeev G, Krishna CM, Upadhya D. Inhibition of ERK1/2 or AKT Activity Equally Enhances Radiation Sensitization in B16F10 Cells. World J Oncol 2018; 9:21-28. [PMID: 29581812 PMCID: PMC5862079 DOI: 10.14740/wjon1088w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/28/2018] [Indexed: 12/11/2022] Open
Abstract
Background The aim of the study was to evaluate the radiation sensitizing ability of ERK1/2, PI3K-AKT and JNK inhibitors in highly radiation resistant and metastatic B16F10 cells which carry wild-type Ras and Braf. Methods Mouse melanoma cell line B16F10 was exposed to 1.0, 2.0 and 3.0 Gy of electron beam radiation. Phosphorylated ERK1/2, AKT and JNK levels were estimated by ELISA. Cells were exposed to 2.0 and 3.0 Gy of radiation with or without prior pharmacological inhibition of ERK1/2, AKT as well as JNK pathways. Cell death induced by radiation as well as upon inhibition of these pathways was measured by TUNEL assay using flow cytometry. Results Exposure of B16F10 cells to 1.0, 2.0 and 3.0 Gy of electron beam irradiation triggered an increase in all the three phosphorylated proteins compared to sham-treated and control groups. B16F10 cells pre-treated with either ERK1/2 or AKT inhibitors equally enhanced radiation-induced cell death at 2.0 as well as 3.0 Gy (P < 0.001), while inhibition of JNK pathway increased radiation-induced cell death to a lesser extent. Interestingly combined inhibition of ERK1/2 or AKT pathways did not show additional cell death compared to individual ERK1/2 or AKT inhibition. This indicates that ERK1/2 or AKT mediates radiation resistance through common downstream molecules in B16F10 cells. Conclusions Even without activating mutations in Ras or Braf genes, ERK1/2 and AKT play a critical role in B16F10 cell survival upon radiation exposure and possibly act through common downstream effector/s.
Collapse
Affiliation(s)
- Bhuvanesh Sukhlal Kalal
- Department of Biochemistry, Yenepoya Medical College, Yenepoya University, Mangalore, India.,Yenepoya Research Centre, Yenepoya University, Mangalore, India
| | - Faraz Fathima
- Yenepoya Research Centre, Yenepoya University, Mangalore, India
| | - Vinitha Ramanath Pai
- Department of Biochemistry, Yenepoya Medical College, Yenepoya University, Mangalore, India
| | - Ganesh Sanjeev
- Department of Physics, Mangalore University, Mangalore, India
| | | | - Dinesh Upadhya
- Yenepoya Research Centre, Yenepoya University, Mangalore, India.,Department of Anatomy, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
45
|
Liu Q, Das M, Liu Y, Huang L. Targeted drug delivery to melanoma. Adv Drug Deliv Rev 2018; 127:208-221. [PMID: 28939379 DOI: 10.1016/j.addr.2017.09.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
Melanoma derived from melanocytes is the most aggressive genre of skin cancer. Although the considerable advancement in the study of human cancer biology and drug discovery, most advanced melanoma patients are inevitably unable to be cured. With the emergence of nanotechnology, the use of nano-carriers is widely expected to alter the landscape of melanoma treatment. In this review, we will discuss melanoma biology, current treatment options, mechanisms behind drug resistance, and nano-based solutions for effective anti-cancer therapy, followed by challenges and perspectives in both pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Cordeiro MF, Marmitt LP, Horn AP. Subcutaneous injection of multipotent mesenchymal stromal cells admixed with melanoma cells in mice favors tumor incidence and growth: a systematic review and meta-analysis. Arch Dermatol Res 2018; 310:231-240. [DOI: 10.1007/s00403-018-1819-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/20/2022]
|
47
|
Del Castillo Velasco‐Herrera M, van der Weyden L, Nsengimana J, Speak AO, Sjöberg MK, Bishop DT, Jönsson G, Newton‐Bishop J, Adams DJ. Comparative genomics reveals that loss of lunatic fringe (LFNG) promotes melanoma metastasis. Mol Oncol 2018; 12:239-255. [PMID: 29193607 PMCID: PMC5792739 DOI: 10.1002/1878-0261.12161] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/16/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Metastasis is the leading cause of death in patients with advanced melanoma, yet the somatic alterations that aid tumour cell dissemination and colonisation are poorly understood. Here, we deploy comparative genomics to identify and validate clinically relevant drivers of melanoma metastasis. To do this, we identified a set of 976 genes whose expression level was associated with a poor outcome in patients from two large melanoma cohorts. Next, we characterised the genomes and transcriptomes of mouse melanoma cell lines defined as weakly metastatic, and their highly metastatic derivatives. By comparing expression data between species, we identified lunatic fringe (LFNG), among 28 genes whose expression level is predictive of poor prognosis and whose altered expression is associated with a prometastatic phenotype in mouse melanoma cells. CRISPR/Cas9-mediated knockout of Lfng dramatically enhanced the capability of weakly metastatic melanoma cells to metastasise in vivo, a phenotype that could be rescued with the Lfng cDNA. Notably, genomic alterations disrupting LFNG are found exclusively in human metastatic melanomas sequenced as part of The Cancer Genome Atlas. Using comparative genomics, we show that LFNG expression plays a functional role in regulating melanoma metastasis.
Collapse
Affiliation(s)
| | | | - Jeremie Nsengimana
- Leeds Institute of Cancer and PathologySt James's University HospitalUniversity of LeedsUK
| | - Anneliese O. Speak
- Experimental Cancer GeneticsWellcome Trust Sanger InstituteHinxtonCambridgeUK
| | - Marcela K. Sjöberg
- Experimental Cancer GeneticsWellcome Trust Sanger InstituteHinxtonCambridgeUK
- Departamento de Biología Celular y MolecularFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - David Timothy Bishop
- Leeds Institute of Cancer and PathologySt James's University HospitalUniversity of LeedsUK
| | - Göran Jönsson
- Division of Oncology and PathologyDepartment of Clinical SciencesSkåne University HospitalLund UniversitySweden
| | - Julia Newton‐Bishop
- Leeds Institute of Cancer and PathologySt James's University HospitalUniversity of LeedsUK
| | - David J. Adams
- Experimental Cancer GeneticsWellcome Trust Sanger InstituteHinxtonCambridgeUK
| |
Collapse
|
48
|
Berridge MV, Neuzil J. The mobility of mitochondria: Intercellular trafficking in health and disease. Clin Exp Pharmacol Physiol 2017; 44 Suppl 1:15-20. [DOI: 10.1111/1440-1681.12764] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/16/2022]
Affiliation(s)
| | - Jiri Neuzil
- School of Medical Science Griffith University Southport Australia
- Institute of Biotechnology Czech Academy of Sciences Prague‐West Czech Republic
| |
Collapse
|
49
|
Anti-tumor effects of differentiation-inducing factor-1 in malignant melanoma: GSK-3-mediated inhibition of cell proliferation and GSK-3-independent suppression of cell migration and invasion. Biochem Pharmacol 2017; 138:31-48. [DOI: 10.1016/j.bcp.2017.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/09/2017] [Indexed: 01/15/2023]
|
50
|
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: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/24/2017] [Indexed: 12/19/2022]
|