1
|
Ghajar-Rahimi G, Yusuf N. Updates in clinical trial-explored chemopreventive agents for cutaneous melanoma: mechanisms affecting melanocytes. Melanoma Manag 2025; 12:2505400. [PMID: 40376946 DOI: 10.1080/20450885.2025.2505400] [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: 01/30/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025] Open
Abstract
Cutaneous melanoma is a highly aggressive skin cancer with rising incidence, driven by risk factors such as ultraviolet exposure, genetic predisposition, and immunosuppression. While surgical excision remains the primary treatment, interest in chemoprevention strategies is growing. Numerous natural and synthetic agents have shown preclinical promise, but evaluating their effectiveness is challenging due to their systemic effects on multiple cell types. This review provides a focused examination of the melanocyte-specific mechanisms of select agents that have been tested in clinical trials for melanoma chemoprevention. We discuss various molecular and cellular mechanisms driving the anti-melanoma properties of nonsteroidal anti-inflammatory drugs, statins, sulforaphane, vitamin D, and N-acetylcysteine. Despite promising preclinical and early clinical data, challenges remain regarding precise mechanisms, optimal dosing, long-term safety, and patient selection. Future research should focus on refining melanoma prevention strategies through well-designed clinical trials and personalized approaches integrating genetic and molecular risk factors.
Collapse
Affiliation(s)
- Gelare Ghajar-Rahimi
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nabiha Yusuf
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
2
|
Martins M, Veiga F, Paiva-Santos AC, Pires PC. Drug Repurposing and Nanotechnology for Topical Skin Cancer Treatment: Redirecting toward Targeted and Synergistic Antitumor Effects. ACS Pharmacol Transl Sci 2025; 8:308-338. [PMID: 39974652 PMCID: PMC11833728 DOI: 10.1021/acsptsci.4c00679] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Skin cancer represents a major health concern due to its rising incidence and limited treatment options. Current treatments (surgery, chemotherapy, radiotherapy, immunotherapy, and targeted therapy) often entail high costs, patient inconvenience, significant adverse effects, and limited therapeutic efficacy. The search for novel treatment options is also marked by the high capital investment and extensive development involved in the drug discovery process. In response to these challenges, repurposing existing drugs for topical application and optimizing their delivery through nanotechnology could be the answer. This innovative strategy aims to combine the advantages of the known pharmacological background of commonly used drugs to expedite therapeutic development, with nanosystem-based formulations, which among other advantages allow for improved skin permeation and retention and overall higher therapeutic efficacy and safety. The present review provides a critical analysis of repurposed drugs such as doxycycline, itraconazole, niclosamide, simvastatin, leflunomide, metformin, and celecoxib, formulated into different nanosystems, namely, nanoemulsions and nanoemulgels, nanodispersions, solid lipid nanoparticles, nanostructured lipid carriers, polymeric nanoparticles, hybrid lipid-polymer nanoparticles, hybrid electrospun nanofibrous scaffolds, liposomes and liposomal gels, ethosomes and ethosomal gels, and aspasomes, for improved outcomes in the battle against skin cancer. Enhanced antitumor effects on melanoma and nonmelanoma research models are highlighted, with some nanoparticles even showing intrinsic anticancer properties, leading to synergistic effects. The explored research findings highly evidence the potential of these approaches to complement the currently available therapeutic strategies in the hope that these treatments might one day reach the pharmaceutical market.
Collapse
Affiliation(s)
- Maria Martins
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia C. Pires
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- RISE-Health,
Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
3
|
Liang J, Yu D, Luo C, Bennett C, Jedrychowski M, Gygi SP, Widlund HR, Puigserver P. Epigenetic suppression of PGC1α (PPARGC1A) causes collateral sensitivity to HMGCR-inhibitors within BRAF-treatment resistant melanomas. Nat Commun 2023; 14:3251. [PMID: 37277330 PMCID: PMC10241879 DOI: 10.1038/s41467-023-38968-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
While targeted treatment against BRAF(V600E) improve survival for melanoma patients, many will see their cancer recur. Here we provide data indicating that epigenetic suppression of PGC1α defines an aggressive subset of chronic BRAF-inhibitor treated melanomas. A metabolism-centered pharmacological screen further identifies statins (HMGCR inhibitors) as a collateral vulnerability within PGC1α-suppressed BRAF-inhibitor resistant melanomas. Lower PGC1α levels mechanistically causes reduced RAB6B and RAB27A expression, whereby their combined re-expression reverses statin vulnerability. BRAF-inhibitor resistant cells with reduced PGC1α have increased integrin-FAK signaling and improved extracellular matrix detached survival cues that helps explain their increased metastatic ability. Statin treatment blocks cell growth by lowering RAB6B and RAB27A prenylation that reduces their membrane association and affects integrin localization and downstream signaling required for growth. These results suggest that chronic adaptation to BRAF-targeted treatments drive novel collateral metabolic vulnerabilities, and that HMGCR inhibitors may offer a strategy to treat melanomas recurring with suppressed PGC1α expression.
Collapse
Affiliation(s)
- Jiaxin Liang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Deyang Yu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Chi Luo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Parthenon Therapeutics, Boston, MA, 02135, USA
| | - Christopher Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Atavistik Bio, Cambridge, MA, 02139, USA
| | - Mark Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Fuentes-Fayos AC, G-García ME, Pérez-Gómez JM, Montero-Hidalgo AJ, Martín-Colom J, Doval-Rosa C, Blanco-Acevedo C, Torres E, Toledano-Delgado Á, Sánchez-Sánchez R, Peralbo-Santaella E, Ortega-Salas RM, Jiménez-Vacas JM, Tena-Sempere M, López M, Castaño JP, Gahete MD, Solivera J, Luque RM. Metformin and simvastatin exert additive antitumour effects in glioblastoma via senescence-state: clinical and translational evidence. EBioMedicine 2023; 90:104484. [PMID: 36907105 PMCID: PMC10024193 DOI: 10.1016/j.ebiom.2023.104484] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/13/2023] [Accepted: 02/03/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Glioblastoma is one of the most devastating and incurable cancers due to its aggressive behaviour and lack of available therapies, being its overall-survival from diagnosis ∼14-months. Thus, identification of new therapeutic tools is urgently needed. Interestingly, metabolism-related drugs (e.g., metformin/statins) are emerging as efficient antitumour agents for several cancers. Herein, we evaluated the in vitro/in vivo effects of metformin and/or statins on key clinical/functional/molecular/signalling parameters in glioblastoma patients/cells. METHODS An exploratory-observational-randomized retrospective glioblastoma patient cohort (n = 85), human glioblastoma/non-tumour brain human cells (cell lines/patient-derived cell cultures), mouse astrocytes progenitor cell cultures, and a preclinical xenograft glioblastoma mouse model were used to measure key functional parameters, signalling-pathways and/or antitumour progression in response to metformin and/or simvastatin. FINDINGS Metformin and simvastatin exerted strong antitumour actions in glioblastoma cell cultures (i.e., proliferation/migration/tumoursphere/colony-formation/VEGF-secretion inhibition and apoptosis/senescence induction). Notably, their combination additively altered these functional parameters vs. individual treatments. These actions were mediated by the modulation of key oncogenic signalling-pathways (i.e., AKT/JAK-STAT/NF-κB/TGFβ-pathways). Interestingly, an enrichment analysis uncovered a TGFβ-pathway activation, together with AKT inactivation, in response to metformin + simvastatin combination, which might be linked to an induction of the senescence-state, the associated secretory-phenotype, and to the dysregulation of spliceosome components. Remarkably, the antitumour actions of metformin + simvastatin combination were also observed in vivo [i.e., association with longer overall-survival in human, and reduction in tumour-progression in a mouse model (reduced tumour-size/weight/mitosis-number, and increased apoptosis)]. INTERPRETATION Altogether, metformin and simvastatin reduce aggressiveness features in glioblastomas, being this effect significantly more effective (in vitro/in vivo) when both drugs are combined, offering a clinically relevant opportunity that should be tested for their use in humans. FUNDING Spanish Ministry of Science, Innovation and Universities; Junta de Andalucía; CIBERobn (CIBER is an initiative of Instituto de Salud Carlos III, Spanish Ministry of Health, Social Services and Equality).
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain.
| | - Miguel E G-García
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Antonio J Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Julia Martín-Colom
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Carlos Doval-Rosa
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Encarnación Torres
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Álvaro Toledano-Delgado
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Pathology Service, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Esther Peralbo-Santaella
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Flow Cytometry Unit, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004, Cordoba, Spain
| | - Rosa M Ortega-Salas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Pathology Service, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Manuel Tena-Sempere
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Miguel López
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain; NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Juan Solivera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain.
| |
Collapse
|
5
|
Andersson CR, Ye J, Blom K, Fryknäs M, Larsson R, Nygren P. Assessment in vitro of interactions between anti-cancer drugs and noncancer drugs commonly used by cancer patients. Anticancer Drugs 2023; 34:92-102. [PMID: 36066384 PMCID: PMC9760465 DOI: 10.1097/cad.0000000000001344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/02/2022]
Abstract
Cancer patients often suffer from cancer symptoms, treatment complications and concomitant diseases and are, therefore, often treated with several drugs in addition to anticancer drugs. Whether such drugs, here denoted as 'concomitant drugs', have anticancer effects or interact at the tumor cell level with the anticancer drugs is not very well known. The cytotoxic effects of nine concomitant drugs and their interactions with five anti-cancer drugs commonly used for the treatment of colorectal cancer were screened over broad ranges of drug concentrations in vitro in the human colon cancer cell line HCT116wt. Seven additional tyrosine kinase inhibitors were included to further evaluate key findings as were primary cultures of tumor cells from patients with colorectal cancer. Cytotoxic effects were evaluated using the fluorometric microculture cytotoxicity assay (FMCA) and interaction analysis was based on Bliss independent interaction analysis. Simvastatin and loperamide, included here as an opioid agonists, were found to have cytotoxic effects on their own at reasonably low concentrations whereas betamethasone, enalapril, ibuprofen, metformin, metoclopramide, metoprolol and paracetamol were inactive also at very high concentrations. Drug interactions ranged from antagonistic to synergistic over the concentrations tested with a more homogenous pattern of synergy between simvastatin and protein kinase inhibitors in HCT116wt cells. Commonly used concomitant drugs are mostly neither expected to have anticancer effects nor to interact significantly with anticancer drugs frequently used for the treatment of colorectal cancer.
Collapse
Affiliation(s)
| | - Jiawei Ye
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Medical Laboratory Sciences, School of Medicine, Southeast University, Nanjing, China
| | - Kristin Blom
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mårten Fryknäs
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter Nygren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Sharma S, Dayal S, Aggarwal K, Gowda V.M V, Kaur R. Scrutinising the role of simvastatin in a patient of Pachyonychia Congenita with
KRT6A
gene mutation. Australas J Dermatol 2022; 63:e145-e149. [DOI: 10.1111/ajd.13835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Shruti Sharma
- Pt BD Sharma Postgraduate Institute of Medical Sciences Rohtak India
| | - Surabhi Dayal
- Pt BD Sharma Postgraduate Institute of Medical Sciences Rohtak India
| | - Kamal Aggarwal
- Pt BD Sharma Postgraduate Institute of Medical Sciences Rohtak India
| | - Varsha Gowda V.M
- Pt BD Sharma Postgraduate Institute of Medical Sciences Rohtak India
| | - Rupinder Kaur
- Pt BD Sharma Postgraduate Institute of Medical Sciences Rohtak India
| |
Collapse
|
7
|
Abstract
Melanoma is the most lethal skin cancer that originates from the malignant transformation of melanocytes. Although melanoma has long been regarded as a cancerous malignancy with few therapeutic options, increased biological understanding and unprecedented innovations in therapies targeting mutated driver genes and immune checkpoints have substantially improved the prognosis of patients. However, the low response rate and inevitable occurrence of resistance to currently available targeted therapies have posed the obstacle in the path of melanoma management to obtain further amelioration. Therefore, it is necessary to understand the mechanisms underlying melanoma pathogenesis more comprehensively, which might lead to more substantial progress in therapeutic approaches and expand clinical options for melanoma therapy. In this review, we firstly make a brief introduction to melanoma epidemiology, clinical subtypes, risk factors, and current therapies. Then, the signal pathways orchestrating melanoma pathogenesis, including genetic mutations, key transcriptional regulators, epigenetic dysregulations, metabolic reprogramming, crucial metastasis-related signals, tumor-promoting inflammatory pathways, and pro-angiogenic factors, have been systemically reviewed and discussed. Subsequently, we outline current progresses in therapies targeting mutated driver genes and immune checkpoints, as well as the mechanisms underlying the treatment resistance. Finally, the prospects and challenges in the development of melanoma therapy, especially immunotherapy and related ongoing clinical trials, are summarized and discussed.
Collapse
Affiliation(s)
- Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China.
| |
Collapse
|
8
|
Yu WY, Hill ST, Chan ER, Pink JJ, Cooper K, Leachman S, Lund AW, Kulkarni R, Bordeaux JS. Computational Drug Repositioning Identifies Statins as Modifiers of Prognostic Genetic Expression Signatures and Metastatic Behavior in Melanoma. J Invest Dermatol 2021; 141:1802-1809. [PMID: 33417917 DOI: 10.1016/j.jid.2020.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/02/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022]
Abstract
Despite advances in melanoma treatment, more than 70% of patients with distant metastasis die within 5 years. Proactive treatment of early melanoma to prevent metastasis could save lives and reduce overall healthcare costs. Currently, there are no treatments specifically designed to prevent early melanoma from progressing to metastasis. We used the Connectivity Map to conduct an in silico drug screen and identified 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins) as a drug class that might prevent melanoma metastasis. To confirm the in vitro effect of statins, RNA sequencing was completed on A375 cells after treatment with fluvastatin to describe changes in the melanoma transcriptome. Statins induced differential expression in genes associated with metastasis and are used in commercially available prognostic tests for melanoma metastasis. Finally, we completed a chart review of 475 patients with melanoma. Patients taking statins were less likely to have metastasis at the time of melanoma diagnosis in both univariate and multivariate analyses (24.7% taking statins vs. 37.6% not taking statins, absolute risk reduction = 12.9%, P = 0.038). These findings suggest that statins might be useful as a treatment to prevent melanoma metastasis. Prospective trials are required to verify our findings and to determine the mechanism of metastasis prevention.
Collapse
Affiliation(s)
- Wesley Y Yu
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA.
| | - Sheena T Hill
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - E Ricky Chan
- Institute for Computational Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - John J Pink
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kevin Cooper
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Sancy Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Rajan Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Jeremy S Bordeaux
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Kobayashi K, Baba K, Kambayashi S, Okuda M. Effect of simvastatin on cell proliferation and Ras activation in canine tumour cells. Vet Comp Oncol 2020; 19:99-108. [PMID: 32779819 DOI: 10.1111/vco.12644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/24/2022]
Abstract
Statins are inhibitors of the mevalonate cascade that is responsible for cholesterol biosynthesis and the formation of intermediate metabolites, farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP) used in the prenylation of proteins. Although statins are widely used in the treatment of hypercholesterolemia, recent studies suggest that they also inhibit proliferation of tumour cells by reducing prenylation of small GTP-binding proteins, such as, Ras. This study aimed to evaluate the effect of simvastatin on cell proliferation and Ras activation in various canine tumour cell lines, including hemangiosarcoma (HSA), melanoma, and lymphoma cell lines. Simvastatin inhibited cell proliferation of all cell lines tested in a concentration- and time-dependent manner, but the susceptibilities were different amongst the cell lines. Simvastatin induced apoptotic cell death via activation of caspase-3 and cell cycle arrest. The cytotoxic effects of simvastatin were attenuated by GGPP and FPP. Simvastatin decreased the amount of prenylated Ras and GTP-bound Ras in HSA and melanoma cell lines, but not in lymphoma cell lines. These results indicate that simvastatin induces cytotoxic effects through the depletion of GGPP and FPP in a variety of canine tumour cells, whereas multiple mechanisms are involved in the effects. Further study is required to elucidate the underlying mechanisms of simvastatin-induced cytotoxic effects in a variety of canine tumour cells.
Collapse
Affiliation(s)
- Kosuke Kobayashi
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Kenji Baba
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Satoshi Kambayashi
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Masaru Okuda
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
10
|
von Schuckmann LA, Khosrotehrani K, Ghiasvand R, Hughes MCB, van der Pols JC, Malt M, Smithers BM, Green AC. Statins may reduce disease recurrence in patients with ulcerated primary melanoma. Br J Dermatol 2020; 183:1049-1055. [PMID: 32133622 DOI: 10.1111/bjd.19012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Statins may restrict the cellular functions required for melanoma growth and metastasis. OBJECTIVES To determine whether long-term statin use commenced before diagnosis of a primary melanoma is associated with reduced risk of melanoma recurrence. METHODS We prospectively followed a cohort of patients newly diagnosed between 2010 and 2014 with localized tumour-stage T1b to T4b melanoma in Queensland, Australia. We used Cox regression analyses to examine associations between long-term statin use and melanoma recurrence for the entire cohort, and then separately by sex and by presence of ulceration, due to evidence of effect modification. RESULTS Among 700 patients diagnosed with stage T1b to T4b primary melanoma (mean age 62 years, 59% male, 28% with ulcerated tumours), 94 patients (13%) developed melanoma recurrence within 2 years. Long-term statin users (n = 204, 29%) had a significantly lower risk of disease recurrence than nonusers [adjusted hazard ratio (HRadj ) 0·55, 95% confidence Interval (CI) 0·32-0·97] regardless of statin subtype or potency. Compared with nonusers of statins, risk of recurrence was significantly decreased in male statin users (HRadj 0·39, 95% CI 0·19-0·79) but not in female statin users (HRadj 0·82, 95% CI 0·29-2·27) and in statin users with ulcerated (HRadj 0·17, 95% CI 0·05-0·52) but not nonulcerated (HRadj 0·91, 95% CI 0·46-1·81) primary melanoma. CONCLUSIONS Statins commenced before melanoma diagnosis may reduce the risk of melanoma recurrence, especially in men and in those with ulcerated tumours. Clinical trial evaluation of the potential role of statins in improving the prognosis of high-risk melanoma is warranted.
Collapse
Affiliation(s)
- L A von Schuckmann
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,School of Public Health, The University of Queensland, Herston, QLD, Australia
| | - K Khosrotehrani
- Experimental Dermatology Group, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia.,Department of Dermatology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - R Ghiasvand
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - M C B Hughes
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - J C van der Pols
- School of Exercise and Nutrition Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - M Malt
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - B M Smithers
- Queensland Melanoma Project, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - A C Green
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,CRUK Manchester and Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
11
|
Cholesterol and beyond - The role of the mevalonate pathway in cancer biology. Biochim Biophys Acta Rev Cancer 2020; 1873:188351. [PMID: 32007596 DOI: 10.1016/j.bbcan.2020.188351] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/14/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer is a multifaceted global disease. Transformation of a normal to a malignant cell takes several steps, including somatic mutations, epigenetic alterations, metabolic reprogramming and loss of cell growth control. Recently, the mevalonate pathway has emerged as a crucial regulator of tumor biology and a potential therapeutic target. This pathway controls cholesterol production and posttranslational modifications of Rho-GTPases, both of which are linked to several key steps of tumor progression. Inhibitors of the mevalonate pathway induce pleiotropic antitumor-effects in several human malignancies, identifying the pathway as an attractive candidate for novel therapies. In this review, we will provide an overview about the role and regulation of the mevalonate pathway in certain aspects of cancer initiation and progression and its potential for therapeutic intervention in oncology.
Collapse
|
12
|
Ji RC, Eshita Y, Kobayashi T, Hidano S, Kamiyama N, Onishi Y. Role of simvastatin in tumor lymphangiogenesis and lymph node metastasis. Clin Exp Metastasis 2018; 35:785-796. [PMID: 30255290 DOI: 10.1007/s10585-018-9940-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/20/2018] [Indexed: 12/27/2022]
Abstract
Lymphangiogenesis plays a crucial role in promoting cancer metastasis to sentinel lymph nodes (LNs) and beyond. Increasing data have shown that simvastatin, a cholesterol-lowering medication for the prevention of cardiovascular diseases, is involved in tumor growth and dissemination, and endothelial functions. This study aimed to investigate the potential effect of simvastatin on lymphatic formation and LN metastasis. Tumor models were established by subcutaneous injection of B16-F10 melanoma cells into mouse hind footpads. Simvastatin was administered (0.2 µg/g, intraperitoneal injection, IP) every other day for a total of eight times. Tissue samples were removed and examined by immunohistochemical staining and reverse transcription-polymerase chain reaction (RT-PCR) techniques. The lymphatics of LN, skin, liver, and lung exhibited morphological changes, and LN weight and metastatic area of the tumor group treated with simvastatin was lower than that of the untreated tumor group. Analysis of lymphatic size, area fraction, and lymphatic vessel density showed tissue specificity and variation to melanoma carcinogenesis in the simvastatin-treated group compared with the untreated group. In addition, LNs and cutaneous tissues showed altered expression of lymphangiogenic factors and inflammatory cytokines such as VEGF-A/-C/-D and TNF-α. These findings indicated that simvastatin may modify lymphangiogenesis and tumor progression in malignant melanoma.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Faculty of Welfare and Health Science, Oita University, Oita, 870-1192, Japan.
| | - Yuki Eshita
- Hokkaido University Research Center for Zoonosis Control, Hokkaido, 001-0020, Japan.,Oita University Faculty of Medicine, Oita, 879-5593, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, 879-5593, Japan
| | - Shinya Hidano
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, 879-5593, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, 879-5593, Japan
| | | |
Collapse
|
13
|
Soran H, Ho JH, Durrington PN. Acquired low cholesterol: diagnosis and relevance to safety of low LDL therapeutic targets. Curr Opin Lipidol 2018; 29:318-326. [PMID: 29746303 DOI: 10.1097/mol.0000000000000526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Acquired hypocholesterolaemia occurs more commonly than inherited hypocholesterolaemia but has received little attention in the literature. In this review, we discuss the causes and underlying mechanisms of acquired hypocholesterolaemia and its relevance to safety of therapeutically induced decreased LDL cholesterol levels. RECENT FINDINGS Hypocholesterolaemia is increasingly identified as cholesterol testing becomes more widespread in the assessment of cardiovascular risk. Lower therapeutic targets for LDL cholesterol are also being achieved more regularly with the introduction of more intensive cholesterol-lowering regimens. Acquired hypocholesterolaemia may be the presenting feature of treatable diseases. Understanding its mechanisms may also provide new treatment approaches for neoplastic disease, such as breast cancer, and infections, such as tuberculosis. SUMMARY When hypocholesterolaemia is discovered, it is important to identify its cause. Further research into the pathogenesis of hypocholesterolaemia may provide new therapies for primary diseases underlying it.
Collapse
Affiliation(s)
- Handrean Soran
- Lipoprotein Research Group, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jan Hoong Ho
- Lipoprotein Research Group, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Paul N Durrington
- Lipoprotein Research Group, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
| |
Collapse
|
14
|
Atorvastatin decreases steroid production in H295R cells and in major endocrine tissues of male rats. Arch Toxicol 2018; 92:1703-1715. [DOI: 10.1007/s00204-018-2187-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/13/2018] [Indexed: 12/13/2022]
|
15
|
Tatè R, Zona E, De Cicco R, Trotta V, Urciuoli M, Morelli A, Baiano S, Carnuccio R, Fuggetta MP, Morelli F. Simvastatin inhibits the expression of stemness-related genes and the metastatic invasion of human cancer cells via destruction of the cytoskeleton. Int J Oncol 2017; 51:1851-1859. [DOI: 10.3892/ijo.2017.4158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/15/2017] [Indexed: 11/06/2022] Open
|
16
|
Sakellakis M, Akinosoglou K, Kostaki A, Spyropoulou D, Koutras A. Statins and risk of breast cancer recurrence. BREAST CANCER-TARGETS AND THERAPY 2016; 8:199-205. [PMID: 27853392 PMCID: PMC5104306 DOI: 10.2147/bctt.s116694] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background The primary end point of our study was to test whether the concurrent use of a statin is related to a lower risk of recurrence and increased relapse-free survival in patients with early breast cancer. Materials and methods We reviewed 610 female patients with stage I, II, or III breast cancer who had been surgically treated and who had subsequently received at least adjuvant chemotherapy in order to prevent recurrence. Results Among the 610 patients with breast cancer, 83 (13.6%) were receiving a statin on a chronic basis for other medical purposes. Overall, statin users displayed longer mean relapse-free survival (16.6 vs 10.2 years, P=0.028). After data had been adjusted for patient and disease characteristics, statin users maintained a lower risk of recurrence. This favorable outcome in statin users was particularly evident when we included only younger patients in the analysis (20 vs 10 years, P=0.006). Conclusion Statins may be linked to a favorable outcome in early breast cancer patients, especially in younger age-groups.
Collapse
Affiliation(s)
- Minas Sakellakis
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| | - Karolina Akinosoglou
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| | - Anastasia Kostaki
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - Despina Spyropoulou
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| | - Angelos Koutras
- Department of Medicine, Division of Oncology, University Hospital, Patras Medical School, Patras
| |
Collapse
|
17
|
Vanderlaan RD, Manlhiot C, Edwards LB, Conway J, McCrindle BW, Dipchand AI. Risk factors for specific causes of death following pediatric heart transplant: An analysis of the registry of the International Society of Heart and Lung Transplantation. Pediatr Transplant 2015; 19:896-905. [PMID: 26381803 DOI: 10.1111/petr.12594] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2015] [Indexed: 11/27/2022]
Abstract
We sought to determine temporal changes in COD and identify COD-specific risk factors in pediatric primary HTx recipients. Using the ISHLT registry, time-dependent hazard of death after pediatric HTx, stratified by COD, was analyzed by multiphasic parametric hazard modeling with multivariable regression models for risk factor analysis. The proportion of pediatric HTx deaths from each of cardiovascular cause, allograft vasculopathy, and malignancy increased over time, while all other COD decreased post-HTx. Pre-HTx ECMO was associated with increased risk of death from graft failure (HR 2.43; p < 0.001), infection (HR 2.85; p < 0.001), and MOF (HR 2.22; p = 0.001), while post-HTx ECMO was associated with death from cerebrovascular events/bleed (HR 2.55; p = 0.001). CHD was associated with deaths due to pulmonary causes (HR 1.78; p = 0.007) or infection (HR 1.72; p < 0.001). Non-adherence was a significant risk factor for all cardiac COD, notably graft failure (HR 1.66; p = 0.001) and rejection (HR 1.89; p < 0.001). Risk factors related to specific COD are varied across different temporal phases post-HTx. Increased understanding of these factors will assist in risk stratification, guide anticipatory clinical decisions, and potentially improve patient survival.
Collapse
Affiliation(s)
- R D Vanderlaan
- Department of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - C Manlhiot
- Department of Pediatrics, Labatt Family Heart Center, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - J Conway
- Department of Pediatrics, Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada
| | - B W McCrindle
- Department of Pediatrics, Labatt Family Heart Center, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - A I Dipchand
- Department of Pediatrics, Labatt Family Heart Center, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Simvastatin decreases steroid production in the H295R cell line and decreases steroids and FSH in female rats. Reprod Toxicol 2015; 58:174-83. [PMID: 26476359 DOI: 10.1016/j.reprotox.2015.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 09/20/2015] [Accepted: 10/09/2015] [Indexed: 12/17/2022]
Abstract
Endocrine modulating effects of Simvastatin (SV) and its metabolite, Simvastatin β-hydroxy acid (SVA), were investigated in H295R cells and in female Sprague-Dawley (SPRD) rats. H295R cells were exposed to SV and SVA concentrations from 0 to 10μM for 48h. Four groups of SPRD rats received 0 (CT), 1.3 (L), 5.0 (M), and 20.0 (H)mg SV/kg bw/day for 14 days. 10 Steroids were investigated in H295R growth media, and in tissues and plasma from rats using GC-MS/MS. Plasma LH and FSH were quantified by ELISA. In the H295R assay, SV and SVA particularly decreased progestagens with IC50-values from 0.10-0.13μM for SV and from 0.019-0.055μM for SVA. In rats, SV decreased progestagens in ovaries, brain and plasma, and plasma FSH in the M (72.4% decrease) and H group (76.6% decrease). Because progestagens and gonadotropins are major players in fertility, administration of SV might exert negative effects on female reproduction.
Collapse
|
19
|
Desai SS, Kharade SS, Parekh VI, Iyer S, Agarwal SK. Pro-oncogenic Roles of HLXB9 Protein in Insulinoma Cells through Interaction with Nono Protein and Down-regulation of the c-Met Inhibitor Cblb (Casitas B-lineage Lymphoma b). J Biol Chem 2015; 290:25595-608. [PMID: 26342078 DOI: 10.1074/jbc.m115.661413] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Indexed: 12/21/2022] Open
Abstract
Pancreatic islet β-cells that lack the MEN1-encoded protein menin develop into tumors. Such tumors express the phosphorylated isoform of the β-cell differentiation transcription factor HLXB9. It is not known how phospho-HLXB9 acts as an oncogenic factor in insulin-secreting β-cell tumors (insulinomas). In this study we investigated the binding partners and target genes of phospho-HLXB9 in mouse insulinoma MIN6 β-cells. Co-immunoprecipitation coupled with mass spectrometry showed a significant association of phospho-HLXB9 with the survival factor p54nrb/Nono (54-kDa nuclear RNA-binding protein, non-POU-domain-containing octamer). Endogenous phospho-HLXB9 co-localized with endogenous Nono in the nucleus. Overexpression of HLXB9 decreased the level of overexpressed Nono but not endogenous Nono. Anti-phospho-HLXB9 chromatin immunoprecipitation followed by deep sequencing (ChIP-Seq) identified the c-Met inhibitor, Cblb, as a direct phospho-HLXB9 target gene. Phospho-HLXB9 occupied the promoter of Cblb and reduced the expression of Cblb mRNA. Cblb overexpression or HLXB9 knockdown decreased c-Met protein and reduced cell migration. Also, increased phospho-HLXB9 coincided with reduced Cblb and increased c-Met in insulinomas of two mouse models of menin loss. These data provide mechanistic insights into the role of phospho-HLXB9 as a pro-oncogenic factor by interacting with a survival factor and by promoting the oncogenic c-Met pathway. These mechanisms have therapeutic implications for reducing β-cell proliferation in insulinomas by inhibiting phospho-HLXB9 or its interaction with Nono and modulating the expression of its direct (Cblb) or indirect (c-Met) targets. Our data also implicate the use of pro-oncogenic activities of phospho-HLXB9 in β-cell expansion strategies to alleviate β-cell loss in diabetes.
Collapse
Affiliation(s)
- Shruti S Desai
- From the Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Sampada S Kharade
- From the Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Vaishali I Parekh
- From the Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Sucharitha Iyer
- From the Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Sunita K Agarwal
- From the Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
20
|
TORRES CRISTIANG, OLIVARES ARACELI, STOORE CAROLL. Simvastatin exhibits antiproliferative effects on spheres derived from canine mammary carcinoma cells. Oncol Rep 2015; 33:2235-44. [DOI: 10.3892/or.2015.3850] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/19/2015] [Indexed: 11/05/2022] Open
|
21
|
Smit MA, Maddalo G, Greig K, Raaijmakers LM, Possik PA, van Breukelen B, Cappadona S, Heck AJR, Altelaar AFM, Peeper DS. ROCK1 is a potential combinatorial drug target for BRAF mutant melanoma. Mol Syst Biol 2014; 10:772. [PMID: 25538140 PMCID: PMC4300494 DOI: 10.15252/msb.20145450] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Treatment of BRAF mutant melanomas with specific BRAF inhibitors leads to tumor remission. However, most patients eventually relapse due to drug resistance. Therefore, we designed an integrated strategy using (phospho)proteomic and functional genomic platforms to identify drug targets whose inhibition sensitizes melanoma cells to BRAF inhibition. We found many proteins to be induced upon PLX4720 (BRAF inhibitor) treatment that are known to be involved in BRAF inhibitor resistance, including FOXD3 and ErbB3. Several proteins were down-regulated, including Rnd3, a negative regulator of ROCK1 kinase. For our genomic approach, we performed two parallel shRNA screens using a kinome library to identify genes whose inhibition sensitizes to BRAF or ERK inhibitor treatment. By integrating our functional genomic and (phospho)proteomic data, we identified ROCK1 as a potential drug target for BRAF mutant melanoma. ROCK1 silencing increased melanoma cell elimination when combined with BRAF or ERK inhibitor treatment. Translating this to a preclinical setting, a ROCK inhibitor showed augmented melanoma cell death upon BRAF or ERK inhibition in vitro. These data merit exploration of ROCK1 as a target in combination with current BRAF mutant melanoma therapies.
Collapse
Affiliation(s)
- Marjon A Smit
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gianluca Maddalo
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Utrecht, The Netherlands Netherlands Proteomics Centre, Utrecht, The Netherlands Center for Biomembrane Research, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Kylie Greig
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Linsey M Raaijmakers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Utrecht, The Netherlands Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Patricia A Possik
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bas van Breukelen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Utrecht, The Netherlands Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Salvatore Cappadona
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Utrecht, The Netherlands Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Utrecht, The Netherlands Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - A F Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, Utrecht University, Utrecht, The Netherlands Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Statins in oncological research: from experimental studies to clinical practice. Crit Rev Oncol Hematol 2014; 92:296-311. [PMID: 25220658 DOI: 10.1016/j.critrevonc.2014.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 06/03/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
Statins, 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors are commonly used drugs in the treatment of dyslipidemias, primarily raised cholesterol. Recently, many epidemiological and preclinical studies pointed to anti-tumor properties of statins, including anti-proliferative activities, apoptosis, decreased angiogenesis and metastasis. These processes play an important role in carcinogenesis and, therefore, the role of statins in cancer disease is being seriously discussed among oncologists. Anti-neoplastic properties of statins combined with an acceptable toxicity profile in the majority of individuals support their further development as anti-tumor drugs. The mechanism of action, current preclinical studies and clinical efficacy of statins are reviewed in this paper. Moreover, promising results have been reported regarding the statins' efficacy in some cancer types, especially in esophageal and colorectal cancers, and hepatocellular carcinoma. Statins' hepatotoxicity has traditionally represented an obstacle to the prescription of this class of drugs and this issue is also discussed in this review.
Collapse
|
23
|
Pisanti S, Picardi P, Ciaglia E, D'Alessandro A, Bifulco M. Novel prospects of statins as therapeutic agents in cancer. Pharmacol Res 2014; 88:84-98. [PMID: 25009097 DOI: 10.1016/j.phrs.2014.06.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/25/2014] [Accepted: 06/25/2014] [Indexed: 02/07/2023]
Abstract
Statins are well known competitive inhibitors of hydroxymethylglutaryl-CoA reductase enzyme (HMG-CoA reductase), thus traditionally used as cholesterol-lowering agents. In recent years, more and more effects of statins have been revealed. Nowadays alterations of lipid metabolism have been increasingly recognized as a hallmark of cancer cells. Consequently, much attention has been directed toward the potential of statins as therapeutic agents in the oncological field. Accumulated in vitro and in vivo clinical evidence point out the role of statins in a variety of human malignancies, in regulating tumor cell growth and anti-tumor immune response. Herein, we summarize and discuss, in light of the most recent observations, the anti-tumor effects of statins, underpinning the detailed mode of action and looking for their true significance in cancer prevention and treatment, to determine if and in which case statin repositioning could be really justified for neoplastic diseases.
Collapse
Affiliation(s)
- Simona Pisanti
- Department of Medicine and Surgery, University of Salerno, Italy; Department of Pharmacy, University of Salerno, Italy.
| | - Paola Picardi
- Department of Medicine and Surgery, University of Salerno, Italy; Department of Pharmacy, University of Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine and Surgery, University of Salerno, Italy; Department of Pharmacy, University of Salerno, Italy
| | - Alba D'Alessandro
- Department of Medicine and Surgery, University of Salerno, Italy; Department of Pharmacy, University of Salerno, Italy
| | - Maurizio Bifulco
- Department of Medicine and Surgery, University of Salerno, Italy; Department of Pharmacy, University of Salerno, Italy.
| |
Collapse
|