1
|
Bédouelle E, Nguyen JM, Varey E, Khammari A, Dreno B. Should Targeted Therapy Be Continued in BRAF-Mutant Melanoma Patients after Complete Remission? Dermatology 2021; 238:517-526. [PMID: 34818219 DOI: 10.1159/000518718] [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: 03/05/2021] [Accepted: 07/03/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Targeted therapy is used to treat patients with a BRAF-mutated metastatic melanoma and is continued until disease progression or severe toxicity. No robust data on the management of patients achieving a complete remission (CR) are available. MAIN OBJECTIVE To determine the relapse rate in the first year after targeted therapy discontinuation in patients in CR. SECONDARY OBJECTIVES To determine the relapse rates throughout the follow-up and to identify prognostic factors for relapse at 1 year. METHODS A retrospective, monocentric observational study was conducted in patients with advanced melanoma included in the RIC-Mel database who discontinued targeted therapy after achieving a CR confirmed by CT scan and PET/CT scan. RESULTS Twenty-nine patients were included. Seventeen (58.6%) patients were treated with BRAF inhibitor (BRAFi) alone and 12 (41.4%) with a BRAFi combined with a MEK inhibitor (BRAFi + MEKi). The median treatment duration was 9.7 months. The relapse rates after discontinuation were 69% at 12 months (BRAFi: 70.6%; BRAFi + MEKi: 66.7%) and 76% at 36 months (BRAFi: 76.5%; BRAFi + MEKi: 75%). A non-significant trend towards a higher risk of relapse was found in women (p = 0.1; RR 3.36; 95% CI 0.77-17.07), in patients with an LDH level greater than the upper limits of normal (p = 0.58; RR 2.43; 95% CI 0.10-56.71), and when more than two metastatic sites were involved (p = 0.19; RR 4.6; 95% CI 0.46-46.51). After relapse, targeted therapy was resumed in 17 patients (7 with BRAFi; 10 with BRAFi + MEKi) with a response rate of 53%. CONCLUSIONS This real-life study provided long-term data in patients who discontinued targeted therapy after CR. Most patients experienced a relapse in the first year after targeted therapy discontinuation, of whom 50% were in the first 3 months. After targeted therapy resumption, 53% of relapsing patients achieved an objective response. Patients should be followed during the first year after treatment discontinuation. In addition, patients with less than 3 metastatic sites, a baseline LDH level with normal ranges, men, and patients responding rapidly to treatment would be more likely to maintain a CR after treatment discontinuation.
Collapse
Affiliation(s)
- Eve Bédouelle
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| | | | - Emilie Varey
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| | - Amir Khammari
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| | - Brigitte Dreno
- Department of Dermatology, CHU Nantes, CIC 1413, CRCINA Inserm U 1232, Nantes University, Nantes, France
| |
Collapse
|
2
|
Huang H, Yi J, Park S, Zhang H, Kim E, Park S, Kwon W, Jang S, Zhang X, Chen H, Choi SK, Kim SH, Liu K, Dong Z, Lee MH, Ryoo Z, Kim MO. Costunolide suppresses melanoma growth via the AKT/mTOR pathway in vitro and in vivo. Am J Cancer Res 2021; 11:1410-1427. [PMID: 33948365 PMCID: PMC8085867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/20/2021] [Indexed: 06/12/2023] Open
Abstract
Melanoma is the most common type of skin cancer and its incidence is rapidly increasing. AKT, and its related signaling pathways, are highly activated in many cancers including lung, colon, and esophageal cancers. Costunolide (CTD) is a sesquiterpene lactone that has been reported to possess neuroprotective, anti-inflammatory, and anti-cancer properties. However, the target and mechanism underlying its efficacy in melanoma have not been identified. In this study, we elucidated the mechanism behind the anti-cancer effect of CTD in melanoma in vitro and in vivo by identifying CTD as an AKT inhibitor. We first verified that p-AKT and AKT are highly expressed in melanoma patient tissues and cell lines. CTD significantly inhibited the proliferation, migration, and invasion of melanoma cells including SK-MEL-5, SK-MEL-28, and A375 that are overexpressed p-AKT and AKT proteins. We investigated the mechanism of CTD using a computational docking modeling, pull-down, and site directed mutagenesis assay. CTD directly bound to AKT thereby arresting cell cycle at the G1 phase, and inducing the apoptosis of melanoma cells. In addition, CTD regulated the G1 phase and apoptosis biomarkers, and inhibited the expression of AKT/mTOR/GSK3b/p70S6K/4EBP cascade proteins. After reducing AKT expression in melanoma cells, cell growth was significantly decreased and CTD did not showed further inhibitory effects. Furthermore, CTD administration suppressed tumor growth and weight in cell-derived xenograft mice models in vivo without body weight loss and inhibited the expression of Ki-67, p-AKT, and p70S6K in tumor tissues. In summary, our study implied that CTD inhibited melanoma progression in vitro and in vivo. In this study, we reported that CTD could affect melanoma growth by targeting AKT. Therefore, CTD has considerable potential as a drug for melanoma therapy.
Collapse
Affiliation(s)
- Hai Huang
- Department of Animal Science and Biotechnology, Kyungpook National UniversitySangju-si, Gyeongsang buk-do 37224, Republic of Korea
| | - Junkoo Yi
- Gyeongbuk Livestock Research InstituteYeongju 36052, South Korea
| | - Song Park
- Core Protein Resources Center, DGISTDaegu, Republic of Korea
- Department of Brian and Cognitive Sciences, DGISTDaegu, Republic of Korea
| | - Haibo Zhang
- Department of Animal Science and Biotechnology, Kyungpook National UniversitySangju-si, Gyeongsang buk-do 37224, Republic of Korea
| | - Eungyung Kim
- Department of Animal Science and Biotechnology, Kyungpook National UniversitySangju-si, Gyeongsang buk-do 37224, Republic of Korea
| | - Sijun Park
- Shool of Life Science, Kyungpook National UniversityDaegu, Republic of Korea
| | - Wookbong Kwon
- Shool of Life Science, Kyungpook National UniversityDaegu, Republic of Korea
- Core Protein Resources Center, DGISTDaegu, Republic of Korea
| | - Soyoung Jang
- Shool of Life Science, Kyungpook National UniversityDaegu, Republic of Korea
| | - Xiujuan Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Hanyong Chen
- The Hormel Institute, University of MinnesotaAustin, Minnesota, USA
| | - Seong-Kyoon Choi
- Core Protein Resources Center, DGISTDaegu, Republic of Korea
- Division of Biotechnology, DGISTDaegu, Republic of Korea
| | - Sung-hyun Kim
- Department of Bio-Medical Analysis, Korea Polytechnic CollegeChungnam, Korea
| | - Kangddong Liu
- Basic Medical College, Zhengzhou University ZhengzhouZhengzhou, Henan, China
| | - Zigang Dong
- Basic Medical College, Zhengzhou University ZhengzhouZhengzhou, Henan, China
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin UniversityNaju, Jeollanamdo 58245, Republic of Korea
| | - Zaeyoung Ryoo
- Shool of Life Science, Kyungpook National UniversityDaegu, Republic of Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National UniversitySangju-si, Gyeongsang buk-do 37224, Republic of Korea
| |
Collapse
|
3
|
de Oliveira ÉA, Goding CR, Maria-Engler SS. Organotypic Models in Drug Development "Tumor Models and Cancer Systems Biology for the Investigation of Anticancer Drugs and Resistance Development". Handb Exp Pharmacol 2021; 265:269-301. [PMID: 32548785 DOI: 10.1007/164_2020_369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The landscape of cancer treatment has improved over the past decades, aiming to reduce systemic toxicity and enhance compatibility with the quality of life of the patient. However, at the therapeutic level, metastatic cancer remains hugely challenging, based on the almost inevitable emergence of therapy resistance. A small subpopulation of cells able to survive drug treatment termed the minimal residual disease may either harbor resistance-associated mutations or be phenotypically resistant, allowing them to regrow and become the dominant population in the therapy-resistant tumor. Characterization of the profile of minimal residual disease represents the key to the identification of resistance drivers that underpin cancer evolution. Therapeutic regimens must, therefore, be dynamic and tailored to take into account the emergence of resistance as tumors evolve within a complex microenvironment in vivo. This requires the adoption of new technologies based on the culture of cancer cells in ways that more accurately reflect the intratumor microenvironment, and their analysis using omics and system-based technologies to enable a new era in the diagnostics, classification, and treatment of many cancer types by applying the concept "from the cell plate to the patient." In this chapter, we will present and discuss 3D model building and use, and provide comprehensive information on new genomic techniques that are increasing our understanding of drug action and the emergence of resistance.
Collapse
Affiliation(s)
- Érica Aparecida de Oliveira
- Skin Biology and Melanoma Lab, Department of Clinical Chemistry and Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Silvya Stuchi Maria-Engler
- Skin Biology and Melanoma Lab, Department of Clinical Chemistry and Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
Mutation-oriented profiling of autoinhibitory kinase conformations predicts RAF inhibitor efficacies. Proc Natl Acad Sci U S A 2020; 117:31105-31113. [PMID: 33229534 PMCID: PMC7733820 DOI: 10.1073/pnas.2012150117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Kinase-targeted therapies have the potential to improve the survival of patients with cancer. However, the cancer-specific spectrum of kinase alterations exhibits distinct functional properties and requires mutation-oriented drug treatments. Besides post-translational modifications and diverse intermolecular interactions of kinases, it is the distinct disease mutation which reshapes full-length kinase conformations, affecting their activity. Oncokinase mutation profiles differ between cancer types, as it was shown for BRAF in melanoma and non-small-cell lung cancers. Here, we present the target-oriented application of a kinase conformation (KinCon) reporter platform for live-cell measurements of autoinhibitory kinase activity states. The bioluminescence-based KinCon biosensor allows the tracking of conformation dynamics of full-length kinases in intact cells and real time. We show that the most frequent BRAF cancer mutations affect kinase conformations and thus the engagement and efficacy of V600E-specific BRAF inhibitors (BRAFi). We illustrate that the patient mutation harboring KinCon reporters display differences in the effectiveness of the three clinically approved BRAFi vemurafenib, encorafenib, and dabrafenib and the preclinical paradox breaker PLX8394. We confirmed KinCon-based drug efficacy predictions for BRAF mutations other than V600E in proliferation assays using patient-derived lung cancer cell lines and by analyzing downstream kinase signaling. The systematic implementation of such conformation reporters will allow to accelerate the decision process for the mutation-oriented RAF-kinase cancer therapy. Moreover, we illustrate that the presented kinase reporter concept can be extended to other kinases which harbor patient mutations. Overall, KinCon profiling provides additional mechanistic insights into full-length kinase functions by reporting protein-protein interaction (PPI)-dependent, mutation-specific, and drug-driven changes of kinase activity conformations.
Collapse
|
5
|
Xue YN, Xue YN, Wang ZC, Mo YZ, Wang PY, Tan WQ. A Novel Signature of 23 Immunity-Related Gene Pairs Is Prognostic of Cutaneous Melanoma. Front Immunol 2020; 11:576914. [PMID: 33193373 PMCID: PMC7604355 DOI: 10.3389/fimmu.2020.576914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/29/2020] [Indexed: 01/11/2023] Open
Abstract
In this study, we aimed to identify an immune-related signature for predicting prognosis in cutaneous melanoma (CM). Sample data from The Cancer Genome Atlas (TCGA; n = 460) were used to develop a prognostic signature with 23 immune-related gene pairs (23 IRGPs) for CM. Patients were divided into high- and low-risk groups using the TCGA and validation datasets GSE65904 (n = 214), GSE59455 (n = 141), and GSE22153 (n = 79). The ability of the 23-IRGP signature to predict CM was precise, with the stratified high-risk groups showing a poor prognosis, and it had a significant predictive power when used for immune microenvironment and biological analyses. We subsequently established a novel promising prognostic model in CM to determine the association between the immune microenvironment and CM patient results. This approach may be used to discover signatures in other diseases while avoiding the technical biases associated with other platforms.
Collapse
Affiliation(s)
- Ya-Nan Xue
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Nan Xue
- Department of Biological Science, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Zheng-Cai Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Zhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Pin-Yan Wang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Xing C, Tian H, Zhang Y, Guo K, Tang Y, Wang Q, Lv L, Wang L. DDX39 Overexpression Predicts a Poor Prognosis and Promotes Aggressiveness of Melanoma by Cooperating With SNAIL. Front Oncol 2020; 10:1261. [PMID: 32903487 PMCID: PMC7435017 DOI: 10.3389/fonc.2020.01261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the prognostic value and molecular mechanism of DDX39 and its effector SNAIL in melanoma. First, overexpression of DDX39 in melanoma, which was identified by database analysis, was further validated in patient tissues. Cell growth, cell cycle, cell migration, and cell invasion assays were then performed to evaluate the effects of downregulated DDX39 on the melanoma cell proliferation and aggressiveness. The same approaches were also used to reveal the cooperation of the transcription factor SNAIL with DDX39 to promote the aggressiveness of melanoma cells. We found that the expression of DDX39 was significantly upregulated in melanoma tissue compared to pigmented nevus tissue, and it was positively correlated with the clinical stage defined by the American Joint Committee on Cancer (AJCC) and the prognosis. Downregulation of DDX39 in melanoma cells was found to significantly inhibit cell proliferation, increase G2/M cell cycle arrest, enhance caspase-mediated cell apoptosis, and suppress cell invasion and migration. In addition, we demonstrated that the overexpression of SNAIL could restore the cell growth and aggressiveness impaired by DDX39 RNA interference. Immunohistochemical staining also showed a positive correlation between DDX39 overexpression and SNAIL overexpression in melanoma tissues, suggesting that SNAIL is one of the effectors activated by DDX39. In summary, the overexpression of DDX39 and SNAIL was positively related to the poor prognosis of melanoma patients and the increased aggressiveness of melanoma cells. Our study provides valuable evidence regarding the prognostic value of DDX39 and SNAIL as well as their potential as novel therapeutic targets for treating melanoma patients.
Collapse
Affiliation(s)
- Chengjuan Xing
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Tian
- Department of Emergency Medicine, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
| | - Yini Zhang
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kun Guo
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Tang
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qimin Wang
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li Lv
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lifen Wang
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Boichard A, Richard SB, Kurzrock R. The Crossroads of Precision Medicine and Therapeutic Decision-Making: Use of an Analytical Computational Platform to Predict Response to Cancer Treatments. Cancers (Basel) 2020; 12:cancers12010166. [PMID: 31936627 PMCID: PMC7017109 DOI: 10.3390/cancers12010166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/21/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022] Open
Abstract
Metastatic cancer is a medical challenge that has been historically resistant to treatments. One area of leverage in cancer care is the development of molecularly-driven combination therapies, offering the possibility to overcome resistance. The selection of optimized treatments based on the complex molecular features of a patient’s tumor may be rendered easier by using a computer-assisted program. We used the PreciGENE® platform that uses multi-pathway molecular analysis to identify personalized therapeutic options. These options are ranked using a predictive score reflecting the degree to which a therapy or combination of therapies matches the patient’s biomarker profile. We searched PubMed from February 2010 to June 2017 for all patients described as exceptional responders who also had molecular data available. Altogether, 70 patients with cancer who had received 202 different treatment lines and who had responded (stable disease ≥12 months/partial or complete remission) to ≥1 regimen were curated. We demonstrate that an algorithm reflecting the degree to which patients were matched to the drugs administered correctly ranked the response to the regimens with a sensitivity of 84% and a specificity of 77%. The difference in matching score between successful and unsuccessful treatment lines was significant (median, 65% versus 0%, p-value <0.0001).
Collapse
Affiliation(s)
- Amélie Boichard
- Center for Personalized Cancer Therapy, University of California Moores Cancer Center, La Jolla, CA 92093, USA;
- Correspondence:
| | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California Moores Cancer Center, La Jolla, CA 92093, USA;
| |
Collapse
|
8
|
Ren H, Hu Y, Xie T, Jin C, Hu Y, Yang B. Effect of gefitinib on serum EGFR and CYFRA21-1 in patients with advanced non-small cell lung cancer. Oncol Lett 2019; 18:4167-4175. [PMID: 31516615 PMCID: PMC6732996 DOI: 10.3892/ol.2019.10762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/01/2019] [Indexed: 12/26/2022] Open
Abstract
Changes of epidermal growth factor receptor (EGFR) and cytokeratin fragment antigen 21-1 (CYFRA21-1) in patients with advanced non-small cell lung cancer (NSCLC) before and after gefitinib treatment were observed to explore the significance of such changes. A total of 175 patients with advanced NSCLC who were admitted to Hubei Cancer Hospital from July 2012 to October 2015 were collected and divided into two groups: the control group (85 patients who received conventional chemotherapy) and the experimental group (90 patients treated with gefitinib combined with chemotherapy). The serum expression levels of EGFR and CYFRA21-1 were detected by enzyme-linked immunosorbent assay (ELISA). The therapeutic efficacy and 3-year survival of the two groups were compared, and the factors affecting the survival of the patients were analyzed. The total effective rate and local effective rate of the experimental group were significantly higher than those of the control group (P<0.05). Before treatment, no significant difference was detected in the levels of EGFR and CYFRA21-1 between the two groups (P>0.05). After treatment, the expression levels of EGFR and CYFRA21-1 in the two groups were significantly lower than those before treatment (P<0.05). According to the 3-year survival rate, the experimental group was divided into the survival group and the non-survival group. Single factor analysis was performed on the general data, showing that the influencing factors of the survival include the KPS score, smoking history, number of lesions, pathological stage, EGFR, and CYFRA21-1. Gefitinib can bring significantly improved therapeutic efficacy, lower expression levels of EGFR and CYFRA21-1, and longer survival time for patients with advanced NSCLC. Indicators including confirmed smoking history, a KPS score less than or equal to 60 points, multiple lesions, pathological stage IV, high expression of EGFR and CYFRA21-1, are important factors affecting the survival of patient with advanced NSCLC.
Collapse
Affiliation(s)
- Hui Ren
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| | - Yang Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| | - Tao Xie
- Department of Head and Neck Radiotherapy, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| | - Caibao Jin
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| | - Yanping Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| | - Bin Yang
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
9
|
Röck R, Mayrhofer JE, Torres-Quesada O, Enzler F, Raffeiner A, Raffeiner P, Feichtner A, Huber RG, Koide S, Taylor SS, Troppmair J, Stefan E. BRAF inhibitors promote intermediate BRAF(V600E) conformations and binary interactions with activated RAS. SCIENCE ADVANCES 2019; 5:eaav8463. [PMID: 31453322 PMCID: PMC6693913 DOI: 10.1126/sciadv.aav8463] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 07/09/2019] [Indexed: 05/05/2023]
Abstract
Oncogenic BRAF mutations initiate tumor formation by unleashing the autoinhibited kinase conformation and promoting RAS-decoupled proliferative RAF-MEK-ERK signaling. We have engineered luciferase-based biosensors to systematically track full-length BRAF conformations and interactions affected by tumorigenic kinase mutations and GTP loading of RAS. Binding of structurally diverse αC-helix-OUT BRAF inhibitors (BRAFi) showed differences in specificity and efficacy by shifting patient mutation-containing BRAF reporters from the definitive opened to more closed conformations. Unexpectedly, BRAFi engagement with the catalytic pocket of V600E-mutated BRAF stabilized an intermediate and inactive kinase conformation that enhanced binary RAS:RAF interactions, also independently of RAF dimerization in melanoma cells. We present evidence that the interference with RAS interactions and nanoclustering antagonizes the sequential formation of drug-induced RAS:RAF tetramers. This suggests a previously unappreciated allosteric effect of anticancer drug-driven intramolecular communication between the kinase and RAS-binding domains of mutated BRAF, which may further promote paradoxical kinase activation and drug resistance mechanisms.
Collapse
Affiliation(s)
- Ruth Röck
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Johanna E. Mayrhofer
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Omar Torres-Quesada
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Florian Enzler
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Andrea Raffeiner
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Philipp Raffeiner
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Andreas Feichtner
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Roland G. Huber
- Bioinformatics Institute (BII), Agency for Science Technology and Research (A*STAR), Singapore 138671, Singapore
| | - Shohei Koide
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine and Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Susan S. Taylor
- Department of Pharmacology, Department of Chemistry and Biochemistry, and Howard Hughes Medical Institute, University of California, San Diego, San Diego, CA 92093, USA
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Innsbruck Medical University, Innrain 66, 6020 Innsbruck, Austria
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| |
Collapse
|
10
|
Bergeron C, Moran C, Coroiu A, Körner A. Development and initial validation of the Self-Efficacy for Skin Self-Examination Scale in a Canadian sample of patients with melanoma. Eur J Oncol Nurs 2019; 40:78-84. [PMID: 31229210 DOI: 10.1016/j.ejon.2019.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE Melanoma is the deadliest type of skin cancer, although survival rates are high if detected early. Skin self-examination (SSE) is a health behaviour that can lead to early detection of melanoma and more positive health outcomes. Self-efficacy for SSE is a potential predictor of engaging in skin self-exams. However, no standardized measures of self-efficacy for SSE are currently available. The present study reports on the development and initial validation of a measure assessing self-efficacy for SSE. METHOD Based on a literature review, including previous studies assessing this construct, the research team developed 9 items capturing confidence in one's ability to conduct SSE. Items were subsequently revised by the research team and two dermatologists, resulting in the elimination of 4 items and modifications to the response options. The final 5-item Self-Efficacy for SSE scale was administered to a sample of 242 melanoma patients recruited from local hospitals. We assessed the scale's factor structure through exploratory factor analysis and the internal consistency with Cronbach's alpha. Scale scores were correlated with intentions to perform SSE, physician support for SSE, skin cancer-specific distress, general distress, and sociodemographic variables to assess convergent and divergent validity. RESULTS The scale was found to be unifactorial with adequate model fit, have acceptable internal consistency (Cronbach alpha = 0.74), and initial evidence of convergent and divergent validity, as indicated by the scale correlating with physicians' support for SSE (r = 0.30, p < 0.01), intention to perform SSE (rs = 0.21, p < 0.01) and a weak correlation with general distress (r = 0.15, p < 0.05). CONCLUSIONS Pending future research corroborating these findings, this measure has potential for use by nurses and other health professionals in follow-up care to identify individuals at risk for low self-efficacy for SSE requiring more targeted educational SSE interventions.
Collapse
Affiliation(s)
- Catherine Bergeron
- Department of Educational and Counselling Psychology, McGill University, Montreal, Canada.
| | - Chelsea Moran
- Department of Psychology, University of Calgary, Calgary, Canada
| | - Adina Coroiu
- Department of Educational and Counselling Psychology, McGill University, Montreal, Canada
| | - Annett Körner
- Department of Educational and Counselling Psychology, McGill University, Montreal, Canada; Department of Oncology, McGill University, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Louise Granofsky Psychosocial Oncology Program, Segal Cancer Centre, Montreal, Canada; Psychosocial Oncology Program, McGill University Health Centre, Montreal, Canada
| |
Collapse
|
11
|
Lindsay CR, Jamal-Hanjani M, Forster M, Blackhall F. KRAS: Reasons for optimism in lung cancer. Eur J Cancer 2018; 99:20-27. [PMID: 29894909 DOI: 10.1016/j.ejca.2018.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/21/2018] [Accepted: 05/13/2018] [Indexed: 01/07/2023]
Abstract
Despite being the most frequent gain-of-function genetic alteration in human cancer, KRAS mutation has to date offered only limited potential as a prognostic and predictive biomarker. Results from the phase III SELECT-1 trial in non-small cell lung cancer (NSCLC) recently added to a number of historical and more contemporary disappointments in targeting KRAS mutant disease, including farnesyl transferase inhibition and synthetic lethality partners such as STK33. This narrative review uses the context of these previous failures to demonstrate how the knowledge gained from these experiences can be used as a platform for exciting advances in NSCLC on the horizon. It now seems clear that mutational subtype (most commonly G12C) of individual mutations is of greater relevance than the categorical evaluation of KRAS mutation presence or otherwise. A number of direct small molecules targeted to these subtypes are in development and have shown promising biological activity, with some in the late stages of preclinical validation.
Collapse
Affiliation(s)
- C R Lindsay
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK.
| | - M Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK; Department of Oncology, University College of London Hospital and UCL Cancer Institute, London, UK
| | - M Forster
- Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK; Department of Oncology, University College of London Hospital and UCL Cancer Institute, London, UK
| | - F Blackhall
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK
| |
Collapse
|
12
|
Herraiz C, Jiménez-Cervantes C, Sánchez-Laorden B, García-Borrón JC. Functional interplay between secreted ligands and receptors in melanoma. Semin Cell Dev Biol 2018; 78:73-84. [PMID: 28676423 DOI: 10.1016/j.semcdb.2017.06.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022]
Abstract
Melanoma, the most aggressive form of skin cancer, results from the malignant transformation of melanocytes located in the basement membrane separating the epidermal and dermal skin compartments. Cutaneous melanoma is often initiated by solar ultraviolet radiation (UVR)-induced mutations. Melanocytes intimately interact with keratinocytes, which provide growth factors and melanocortin peptides acting as paracrine regulators of proliferation and differentiation. Keratinocyte-derived melanocortins activate melanocortin-1 receptor (MC1R) to protect melanocytes from the carcinogenic effect of UVR. Accordingly, MC1R is a major determinant of susceptibility to melanoma. Despite extensive phenotypic heterogeneity and high mutation loads, the molecular basis of melanomagenesis and the molecules mediating the crosstalk between melanoma and stromal cells are relatively well understood. Mutations of intracellular effectors of receptor tyrosine kinase (RTK) signalling, notably NRAS and BRAF, are major driver events more frequent than mutations in RTKs. Nevertheless, melanomas often display aberrant signalling from RTKs such as KIT, ERRB1-4, FGFR, MET and PDGFR, which contribute to disease progression and resistance to targeted therapies. Progress has also been made to unravel the role of the tumour secretome in preparing the metastatic niche. However, key aspects of the melanoma-stroma interplay, such as the molecular determinants of dormancy, remain poorly understood.
Collapse
Affiliation(s)
- Cecilia Herraiz
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia, and Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, El Palmar, Murcia, Spain
| | - Celia Jiménez-Cervantes
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia, and Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, El Palmar, Murcia, Spain
| | - Berta Sánchez-Laorden
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - José C García-Borrón
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia, and Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, El Palmar, Murcia, Spain.
| |
Collapse
|
13
|
Didier R, Mallavialle A, Ben Jouira R, Domdom MA, Tichet M, Auberger P, Luciano F, Ohanna M, Tartare-Deckert S, Deckert M. Targeting the Proteasome-Associated Deubiquitinating Enzyme USP14 Impairs Melanoma Cell Survival and Overcomes Resistance to MAPK-Targeting Therapies. Mol Cancer Ther 2018; 17:1416-1429. [PMID: 29703842 DOI: 10.1158/1535-7163.mct-17-0919] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/26/2018] [Accepted: 04/16/2018] [Indexed: 11/16/2022]
Abstract
Advanced cutaneous melanoma is one of the most challenging cancers to treat because of its high plasticity, metastatic potential, and resistance to treatment. New targeted therapies and immunotherapies have shown remarkable clinical efficacy. However, such treatments are limited to a subset of patients and relapses often occur, warranting validation of novel targeted therapies. Posttranslational modification of proteins by ubiquitin coordinates essential cellular functions, including ubiquitin-proteasome system (UPS) function and protein homeostasis. Deubiquitinating enzymes (DUB) have been associated to multiple diseases, including cancer. However, their exact involvement in melanoma development and therapeutic resistance remains poorly understood. Using a DUB trap assay to label cellular active DUBs, we have observed an increased activity of the proteasome-associated DUB, USP14 (Ubiquitin-specific peptidase 14) in melanoma cells compared with melanocytes. Our survey of public gene expression databases indicates that high expression of USP14 correlates with melanoma progression and with a poorer survival rate in metastatic melanoma patients. Knockdown or pharmacologic inhibition of USP14 dramatically impairs viability of melanoma cells irrespective of the mutational status of BRAF, NRAS, or TP53 and their transcriptional cell state, and overcomes resistance to MAPK-targeting therapies both in vitro and in human melanoma xenografted mice. At the molecular level, we find that inhibition of USP14 rapidly triggers accumulation of poly-ubiquitinated proteins and chaperones, mitochondrial dysfunction, ER stress, and a ROS production leading to a caspase-independent cell death. Our results provide a rationale for targeting the proteasome-associated DUB USP14 to treat and combat melanomas. Mol Cancer Ther; 17(7); 1416-29. ©2018 AACR.
Collapse
Affiliation(s)
- Robin Didier
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Aude Mallavialle
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Rania Ben Jouira
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Marie Angela Domdom
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Mélanie Tichet
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | | | | | | | - Sophie Tartare-Deckert
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| | - Marcel Deckert
- Inserm, U1065, Team Microenvironment, Signaling and Cancer, Centre Méditerranéen de Médecine Moléculaire (C3M) and Université Côte d'Azur, Nice, France. .,Equipe labellisée Ligue Contre le Cancer 2016, Nice, France
| |
Collapse
|
14
|
Russell J. Management of colorectal cancer patients with brain metastases. ANZ J Surg 2018; 88:126. [DOI: 10.1111/ans.14227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Jeremy Russell
- Department of Neurosurgery; Austin Hospital; Melbourne Victoria Australia
| |
Collapse
|
15
|
Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat Med 2018; 24:203-212. [DOI: 10.1038/nm.4472] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 12/15/2017] [Indexed: 02/08/2023]
|
16
|
Hermel DJ, Ott PA. Combining forces: the promise and peril of synergistic immune checkpoint blockade and targeted therapy in metastatic melanoma. Cancer Metastasis Rev 2017; 36:43-50. [PMID: 28181070 DOI: 10.1007/s10555-017-9656-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Both immune checkpoint inhibitors and molecularly targeted agents have dramatically improved clinical outcomes for patients with metastatic melanoma. These two therapeutic approaches harness distinct mechanistic pathways-on the one hand, monoclonal antibodies against the immune checkpoints CTLA-4 and PD-1/PD-L1 stimulate the T cell mediated host immune response, while targeted inhibitors of the proto-oncogenes BRAF and MEK disrupt constitutive kinase activity responsible for tumor growth. The prospect of combining these two treatment modalities has been proposed as a potential way to increase overall response rate, extend durability of the anti-tumor response, and circumvent the immune-mediated resistance to targeted therapy. This review explores the preclinical rationale-building upon a wealth of in vitro and in vivo studies-for improved anti-tumor efficacy from combined immune checkpoint inhibition and targeted therapy. In the process, we detail the early clinical trials that have assessed the compatibility of combining these two therapies and the unexpected challenges faced from studies showing increased toxicity from these regimens. Ultimately, with more clinical data expected to mature and accrue in the near future, we elucidate a potentially novel and promising strategy for patients with advanced melanoma.
Collapse
Affiliation(s)
- David J Hermel
- Resident Physician, University of Southern California, Los Angeles, CA, USA
| | - Patrick A Ott
- Melanoma Disease Center and Center for Immuno-Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA, 02215-5450, USA.
| |
Collapse
|
17
|
Abl kinase regulation by BRAF/ERK and cooperation with Akt in melanoma. Oncogene 2017; 36:4585-4596. [PMID: 28368422 PMCID: PMC5552414 DOI: 10.1038/onc.2017.76] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/08/2017] [Accepted: 02/22/2017] [Indexed: 12/18/2022]
Abstract
The melanoma incidence continues to increase, and the disease remains incurable for many due to its metastatic nature and high rate of therapeutic resistance. In particular, melanomas harboring BRAFV600E and PTEN mutations often are resistant to current therapies, including BRAF inhibitors (BRAFi) and immune checkpoint inhibitors. Abl kinases (Abl/Arg) are activated in melanomas and drive progression; however, their mechanism of activation has not been established. Here we elucidate a novel link between BRAFV600E/ERK signaling and Abl kinases. We demonstrate that BRAFV600E/ERK play a critical role in binding, phosphorylating and regulating Abl localization and Abl/Arg activation by Src family kinases. Importantly, Abl/Arg activation downstream of BRAFV600E has functional and biological significance, driving proliferation, invasion, as well as switch in epithelial-mesenchymal-transition transcription factor expression, which is known to be critical for melanoma cells to shift between differentiated and invasive states. Finally, we describe findings of high translational significance by demonstrating that Abl/Arg cooperate with PI3K/Akt/PTEN, a parallel pathway that is associated with intrinsic resistance to BRAFi and immunotherapy, as Abl/Arg and Akt inhibitors cooperate to prevent viability, cell cycle progression and in vivo growth of melanomas harboring mutant BRAF/PTEN. Thus, these data not only provide mechanistic insight into Abl/Arg regulation during melanoma development, but also pave the way for the development of new strategies for treating patients with melanomas harboring mutant BRAF/PTEN, which often are refractory to current therapies.
Collapse
|
18
|
Molecular Profiling of Clear Cell Ovarian Cancers: Identifying Potential Treatment Targets for Clinical Trials. Int J Gynecol Cancer 2017; 26:648-54. [PMID: 26937756 PMCID: PMC4841290 DOI: 10.1097/igc.0000000000000677] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Advanced stage/recurrent clear cell ovarian cancers (CCOCs) are characterized by a low response to chemotherapy and a poor prognosis. There is growing interest in investigating novel/molecular targeted therapies in patients with CCOC in histotype-specific trials. However, CCOCs are not a uniform entity and comprise a number of molecular subtypes and it is unlikely that a single approach to treatment will be appropriate for all patients. The aim of this study was to analyze the results of a multiplatform profiling panel in CCOCs to identify potential therapeutic targets. Patients and Methods Tumor profiling was performed on 521 CCOCs. They were grouped into pure (n = 422) and mixed (n = 99) CCOC for analysis. Testing included a combination of DNA sequencing (including next-generation sequencing) using a 46-gene panel, immunohistochemistry, fluorescent or chromogenic in situ hybridization, and RNA fragment analysis. Results The most common findings were in the PIK3CA/Akt/mTOR pathway, with 61% of all CCOCs showing a molecular alteration in one of these pathway components. Next-generation sequencing revealed PIK3CA mutations in 50% of pure CCOCs. Significant differences were observed between pure and mixed CCOCs with respect to hormone receptor expression (9% vs 34.7% for ER, 13.45 vs 26.4% for PR), cMET (24.1% vs 11.6%), PD-1 tumor infiltrating lymphocytes (48.1% vs 100%), expression of PD-L1 (7.4% vs 25%), and TOPO1 (41% vs 27.1%) on immunohistochemistry, whereas next-generation sequencing revealed significant differences in mutation frequency in PIK3CA (50% vs 18.5%), TP53 (18.1% vs 57.7%), KRAS (12.4% vs 3.7%), and cMET (1.9% vs 11.1%). Conclusions This large study confirms that the PIK3CA/Akt/mTOR pathway is commonly altered in CCOCs, and highlights the significant differences between pure and mixed CCOCs. Clear cell ovarian cancers are molecularly heterogeneous and there are a number of potential therapeutic targets which could be tested in clinical trials.
Collapse
|
19
|
Kwong A, Sanlorenzo M, Rappersberger K, Vujic I. Update on advanced melanoma treatments: small molecule targeted therapy, immunotherapy, and future combination therapies. Wien Med Wochenschr 2017; 169:314-322. [DOI: 10.1007/s10354-016-0535-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/12/2016] [Indexed: 12/15/2022]
|
20
|
Application of aluminum chloride phthalocyanine-loaded solid lipid nanoparticles for photodynamic inactivation of melanoma cells. Int J Pharm 2017; 518:228-241. [PMID: 28063902 DOI: 10.1016/j.ijpharm.2017.01.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/16/2016] [Accepted: 01/02/2017] [Indexed: 12/12/2022]
Abstract
Cutaneous melanoma is the most aggressive skin cancer and is particularly resistant to current therapeutic approaches. Photodynamic therapy (PDT) is a well-established photoprocess that is employed to treat some cancers, including non-melanoma skin cancer. Aluminum chloride phthalocyanine (ClAlPc) is used as a photosensitizer in PDT; however, its high hydrophobicity hampers its photodynamic activity under physiological conditions. The aim of this study was to produce solid lipid nanoparticles (SLN) containing ClAlPc using the direct emulsification method. ClAlPc-loaded SLNs (ClAlPc/SLNs) were characterized according to their particle size and distribution, zeta potential, morphology, encapsulation efficiency, stability, and phototoxic action in vitro in B16-F10 melanoma cells. ClAlPc/SLN had a mean diameter between 100 and 200nm, homogeneous size distribution (polydispersity index <0.3), negative zeta potential, and spherical morphology. The encapsulation efficiency was approximately 100%. The lipid crystallinity was investigated using X-ray diffraction and differential scanning calorimetry and indicated that ClAlPc was integrated into the SLN matrix. The ClAlPc/SLN formulations maintained their physicochemical stability without expelling the drug over a 24-month period. Compared to free ClAlPc, ClAlPc/SLN exerted outstanding phototoxicity effects in vitro against melanoma cells. Therefore, our results demonstrated that the ClAlPc/SLN described in the current study has the potential for use in further preclinical and clinical trials in PDT for melanoma treatment.
Collapse
|
21
|
Peippo M, Gardberg M, Lamminen T, Kaipio K, Carpén O, Heuser VD. FHOD1 formin is upregulated in melanomas and modifies proliferation and tumor growth. Exp Cell Res 2017; 350:267-278. [PMID: 27919746 DOI: 10.1016/j.yexcr.2016.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 01/14/2023]
Abstract
The functional properties of actin-regulating formin proteins are diverse and in many cases cell-type specific. FHOD1, a formin expressed predominantly in cells of mesenchymal lineage, bundles actin filaments and participates in maintenance of cell shape, migration and cellular protrusions. FHOD1 participates in cancer-associated epithelial to mesenchymal transition (EMT) in oral squamous cell carcinoma and breast cancer. The role of FHOD1 in melanomas has not been characterized. Here, we show that FHOD1 expression is typically strong in cutaneous melanomas and cultured melanoma cells while the expression is low or absent in benign nevi. By using shRNA to knockdown FHOD1 in melanoma cells, we discovered that FHOD1 depleted cells are larger, rounder and have smaller focal adhesions and inferior migratory capacity as compared to control cells. Importantly, we found FHOD1 depleted cells to have reduced colony-forming capacity and attenuated tumor growth in vivo, a finding best explained by the reduced proliferation rate caused by cell cycle arrest. Unexpectedly, FHOD1 depletion did not prevent invasive growth at the tumor margins. These results suggest that FHOD1 participates in key cellular processes that are dysregulated in malignancy, but may not be essential for melanoma cell invasion.
Collapse
Affiliation(s)
- Minna Peippo
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital, Turku, Finland; MediCity Research Laboratory, University of Turku, Finland.
| | - Maria Gardberg
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital, Turku, Finland.
| | - Tarja Lamminen
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital, Turku, Finland; MediCity Research Laboratory, University of Turku, Finland.
| | - Katja Kaipio
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital, Turku, Finland; MediCity Research Laboratory, University of Turku, Finland.
| | - Olli Carpén
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital, Turku, Finland; Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Vanina D Heuser
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital, Turku, Finland; MediCity Research Laboratory, University of Turku, Finland.
| |
Collapse
|
22
|
Gkountela S, Szczerba B, Donato C, Aceto N. Recent advances in the biology of human circulating tumour cells and metastasis. ESMO Open 2016; 1:e000078. [PMID: 27843628 PMCID: PMC5070257 DOI: 10.1136/esmoopen-2016-000078] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 01/22/2023] Open
Abstract
The development of a metastatic disease is recognised as the cause of death of over 90% of patients diagnosed with cancer. Understanding the biological features of metastasis has been hampered for a long time by the difficulties to study widespread cancerous lesions in patients, and by the absence of reliable methods to isolate viable metastatic cells during disease progression. These difficulties negatively impact on our ability to develop new agents that are tailored to block the spread of cancer. Yet, recent advances in specialised devices for the isolation of circulating tumour cells (CTCs), hand-in-hand with technologies that enable single cell resolution interrogation of their genome and transcriptome, are now paving the way to understanding those molecular mechanisms that drive the formation of metastasis. In this review, we aim to summarise some of the latest discoveries in CTC biology in the context of several types of cancer, and to highlight those findings that have a potential to improve the clinical management of patients with metastatic cancer.
Collapse
Affiliation(s)
- Sofia Gkountela
- Cancer Metastasis, Department of Biomedicine , University of Basel , Basel , Switzerland
| | - Barbara Szczerba
- Cancer Metastasis, Department of Biomedicine , University of Basel , Basel , Switzerland
| | - Cinzia Donato
- Cancer Metastasis, Department of Biomedicine , University of Basel , Basel , Switzerland
| | - Nicola Aceto
- Cancer Metastasis, Department of Biomedicine , University of Basel , Basel , Switzerland
| |
Collapse
|
23
|
Gkountela S, Aceto N. Stem-like features of cancer cells on their way to metastasis. Biol Direct 2016; 11:33. [PMID: 27457474 PMCID: PMC4960876 DOI: 10.1186/s13062-016-0135-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED More than 90 % of cancer-related deaths are due to the development of a systemic metastatic disease. Clearly, much remains to be understood about the biological principles that govern human cancer metastasis, aiming at the ambitious objective to decrease metastasis-related mortality in patients. For many years, research on metastasis has been conducted in great part on experimental mouse models, mainly because of the difficulties in sampling, longitudinal studies, and molecular interrogation of a human metastatic disease. However, recently, extraordinary advances in microfluidic technologies are allowing the isolation and characterization of human circulating tumor cells (CTCs) that escaped a primary tumor mass and are in the process of seeding a distant metastasis. Analysis of human CTCs has now revealed important features of cancer metastasis, such as the high metastatic potential of CTC-clusters compared to single CTCs, the dynamic expression of epithelial and mesenchymal markers on CTCs during treatment, and the possibility to culture CTCs from patients for a real-time and individualized testing of drug susceptibility. Nevertheless, several aspects of CTC biology remain unsolved, such as the characterization of the stem-like cell population among human CTCs. Here, we focus on describing the latest findings in the CTC field, and discuss them in the context of cancer stem cell biology. Defining the molecular features of those few metastasis-initiating, stem-like CTCs holds the exceptional promise to develop metastasis-tailored therapies for patients with cancer. REVIEWERS This article was reviewed by Elisa Cimetta, Luca Pellegrini and Sirio Dupont (nominated by LP).
Collapse
Affiliation(s)
- Sofia Gkountela
- Department of Biomedicine, Cancer Metastasis, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | - Nicola Aceto
- Department of Biomedicine, Cancer Metastasis, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| |
Collapse
|
24
|
Rajkumar S, Watson IR. Molecular characterisation of cutaneous melanoma: creating a framework for targeted and immune therapies. Br J Cancer 2016; 115:145-55. [PMID: 27336610 PMCID: PMC4947706 DOI: 10.1038/bjc.2016.195] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 05/16/2016] [Accepted: 05/18/2016] [Indexed: 12/12/2022] Open
Abstract
Large-scale genomic analyses of cutaneous melanoma have revealed insights into the aetiology and heterogeneity of this disease, as well as opportunities to further personalise treatment for patients with targeted and immune therapies. Herein, we review the proposed genomic classification of cutaneous melanoma from large-scale next-generation sequencing studies, including the largest integrative analysis of melanoma from The Cancer Genome Atlas (TCGA) Network. We examine studies that have identified molecular features of melanomas linked to immune checkpoint inhibitor response. In addition, we draw attention to low-frequency actionable mutations and highlight frequent non-coding mutations in melanoma where little is known about their biological function that may provide novel avenues for the development of treatment strategies for melanoma patients.
Collapse
Affiliation(s)
- Shivshankari Rajkumar
- Rosalind and Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montréal, QC, Canada H3A 1A3
| | - Ian R Watson
- Rosalind and Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montréal, QC, Canada H3A 1A3
| |
Collapse
|
25
|
The mitogen-activated protein kinase pathway plays a critical role in regulating immunological properties of BRAF mutant cutaneous melanoma cells. Melanoma Res 2016; 26:223-35. [DOI: 10.1097/cmr.0000000000000244] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Girotti MR, Gremel G, Lee R, Galvani E, Rothwell D, Viros A, Mandal AK, Lim KHJ, Saturno G, Furney SJ, Baenke F, Pedersen M, Rogan J, Swan J, Smith M, Fusi A, Oudit D, Dhomen N, Brady G, Lorigan P, Dive C, Marais R. Application of Sequencing, Liquid Biopsies, and Patient-Derived Xenografts for Personalized Medicine in Melanoma. Cancer Discov 2016; 6:286-99. [PMID: 26715644 DOI: 10.1158/2159-8290.cd-15-1336] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/23/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Targeted therapies and immunotherapies have transformed melanoma care, extending median survival from ∼9 to over 25 months, but nevertheless most patients still die of their disease. The aim of precision medicine is to tailor care for individual patients and improve outcomes. To this end, we developed protocols to facilitate individualized treatment decisions for patients with advanced melanoma, analyzing 364 samples from 214 patients. Whole exome sequencing (WES) and targeted sequencing of circulating tumor DNA (ctDNA) allowed us to monitor responses to therapy and to identify and then follow mechanisms of resistance. WES of tumors revealed potential hypothesis-driven therapeutic strategies for BRAF wild-type and inhibitor-resistant BRAF-mutant tumors, which were then validated in patient-derived xenografts (PDX). We also developed circulating tumor cell-derived xenografts (CDX) as an alternative to PDXs when tumors were inaccessible or difficult to biopsy. Thus, we describe a powerful technology platform for precision medicine in patients with melanoma. SIGNIFICANCE Although recent developments have revolutionized melanoma care, most patients still die of their disease. To improve melanoma outcomes further, we developed a powerful precision medicine platform to monitor patient responses and to identify and validate hypothesis-driven therapies for patients who do not respond, or who develop resistance to current treatments.
Collapse
Affiliation(s)
- Maria Romina Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Gabriela Gremel
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Rebecca Lee
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Elena Galvani
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Dominic Rothwell
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Amaya Viros
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Amit Kumar Mandal
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Kok Haw Jonathan Lim
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Grazia Saturno
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Simon J Furney
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Franziska Baenke
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Jane Rogan
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Jacqueline Swan
- Research Services, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Matthew Smith
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Alberto Fusi
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Deemesh Oudit
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Nathalie Dhomen
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Ged Brady
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Paul Lorigan
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom.
| |
Collapse
|
27
|
Huang SK, Hoon DSB. Liquid biopsy utility for the surveillance of cutaneous malignant melanoma patients. Mol Oncol 2016; 10:450-63. [PMID: 26778792 PMCID: PMC5307330 DOI: 10.1016/j.molonc.2015.12.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 01/01/2023] Open
Abstract
Cutaneous melanoma is one of the highest incident-rate cancers with increasing prevalence in Western societies. Despite the advent of new approved therapeutics, the 5-year overall survival rate of stage IV melanoma patients remains below 15%. Current treatments for late stage disease have shown higher efficacy when treated at a lower disease burden. Thus, blood-based biomarkers capable of detecting melanoma prior to clinically evident distant metastasis, will improve the treatment and outcomes for melanoma patients. To that end, effective treatment of melanoma necessitates identification of patients at risk for developing distant metastases. Furthermore, employing blood biomarkers that monitor cancer progression over the course of treatment is a promising solution to post-treatment drug resistance often developed in melanoma patients. Non-invasive blood biomarker assays allow for regular dynamic monitoring of disease. "Liquid Biopsy" of blood, which exploits circulating tumor cells (CTCs), cell-free circulating tumor DNA (ctDNA) and cell-free circulating microRNA (cmiRNA), has been shown to detect prognostic factors for relapse in AJCC stage III and stage IV melanoma patients. Moreover, molecular characterization of CTC and analysis of various forms of ctDNA present promising potential in development of individualized therapy for melanoma patients. New approaches such as massive parallel sequencing (MPS) provide a comprehensive view of the disease progression, allowing for the selection of therapeutic options for individual patients. With advancements of improving molecular assays, liquid biopsy analysis as a powerful, routine clinical assay for melanoma patients, is highly promising prospective.
Collapse
Affiliation(s)
- Sharon K Huang
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404, United States
| | - Dave S B Hoon
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404, United States.
| |
Collapse
|
28
|
Jayachandran A, Lo PH, Chueh AC, Prithviraj P, Molania R, Davalos-Salas M, Anaka M, Walkiewicz M, Cebon J, Behren A. Transketolase-like 1 ectopic expression is associated with DNA hypomethylation and induces the Warburg effect in melanoma cells. BMC Cancer 2016; 16:134. [PMID: 26907172 PMCID: PMC4763451 DOI: 10.1186/s12885-016-2185-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 02/16/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The metabolism of cancer cells is often reprogrammed by dysregulation of metabolic enzymes. Transketolase-like 1 (TKTL1) is a homodimeric transketolase linking the pentose-phosphate pathway with the glycolytic pathway. It is generally silenced at a transcriptional level in somatic tissues. However, in human cancers its expression is associated with the acquisition of a glycolytic phenotype (the Warburg effect) by cancer cells that contributes to the progression of malignant tumors. In melanoma, defective promoter methylation results in the expression of genes and their products that can affect the tumor cell's phenotype including the modification of immune and functional characteristics. The present study evaluates the role of TKTL1 as a mediator of disease progression in melanoma associated with a defective methylation phenotype. METHODS The expression of TKTL1 in metastatic melanoma tumors and cell lines was analysed by qRT-PCR and immunohistochemistry. The promoter methylation status of TKTL1 in melanoma cells was evaluated by quantitative methylation specific PCR. Using qRT-PCR, the effect of a DNA demethylating agent 5-aza-2'-deoxycytidine (5aza) on the expression of TKTL1 was examined. Biochemical and molecular analyses such as glucose consumption, lactate production, invasion, proliferation and cell cycle progression together with ectopic expression and siRNA mediated knockdown were used to investigate the role of TKTL1 in melanoma cells. RESULTS Expression of TKTL1 was highly restricted in normal adult tissues and was overexpressed in a subset of metastatic melanoma tumors and derived cell lines. The TKTL1 promoter was activated by hypomethylation and treatment with 5aza induced TKTL1 expression in melanoma cells. Augmented expression of TKTL1 in melanoma cells was associated with a glycolytic phenotype. Loss and gain of function studies revealed that TKTL1 contributed to enhanced invasion of melanoma cells. CONCLUSIONS Our data provide evidence for an important role of TKTL1 in aerobic glycolysis and tumor promotion in melanoma that may result from defective promoter methylation. This epigenetic change may enable the natural selection of tumor cells with a metabolic phenotype and thereby provide a potential therapeutic target for a subset of melanoma tumors with elevated TKTL1 expression.
Collapse
Affiliation(s)
- Aparna Jayachandran
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Cancer Medicine, Latrobe University, Melbourne, VIC, 3086, Australia.,School of Medicine and the Gallipoli Medical Research Foundation, The University of Queensland, Brisbane, QLD 4120, Australia
| | - Pu-Han Lo
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia
| | - Anderly C Chueh
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, 3010, Australia
| | - Prashanth Prithviraj
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ramyar Molania
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Mercedes Davalos-Salas
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Matthew Anaka
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Marzena Walkiewicz
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Jonathan Cebon
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Cancer Medicine, Latrobe University, Melbourne, VIC, 3086, Australia
| | - Andreas Behren
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia. .,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia. .,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia. .,School of Cancer Medicine, Latrobe University, Melbourne, VIC, 3086, Australia. .,Cancer Immuno-biology Laboratory, Olivia Newton-John Cancer Research Institute, Level 5, Olivia Newton-John Cancer and Wellness Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
29
|
Baenke F, Chaneton B, Smith M, Van Den Broek N, Hogan K, Tang H, Viros A, Martin M, Galbraith L, Girotti MR, Dhomen N, Gottlieb E, Marais R. Resistance to BRAF inhibitors induces glutamine dependency in melanoma cells. Mol Oncol 2016; 10:73-84. [PMID: 26365896 PMCID: PMC4717845 DOI: 10.1016/j.molonc.2015.08.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/07/2015] [Accepted: 08/09/2015] [Indexed: 12/12/2022] Open
Abstract
BRAF inhibitors can extend progression-free and overall survival in melanoma patients whose tumors harbor mutations in BRAF. However, the majority of patients eventually develop resistance to these drugs. Here we show that BRAF mutant melanoma cells that have developed acquired resistance to BRAF inhibitors display increased oxidative metabolism and increased dependency on mitochondria for survival. Intriguingly, the increased oxidative metabolism is associated with a switch from glucose to glutamine metabolism and an increased dependence on glutamine over glucose for proliferation. We show that the resistant cells are more sensitive to mitochondrial poisons and to inhibitors of glutaminolysis, suggesting that targeting specific metabolic pathways may offer exciting therapeutic opportunities to treat resistant tumors, or to delay emergence of resistance in the first-line setting.
Collapse
Affiliation(s)
- Franziska Baenke
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Barbara Chaneton
- Cancer Metabolism Research Unit, Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| | - Matthew Smith
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Niels Van Den Broek
- Cancer Metabolism Research Unit, Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| | - Kate Hogan
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Haoran Tang
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Amaya Viros
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Matthew Martin
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Laura Galbraith
- Cancer Metabolism Research Unit, Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| | - Maria R Girotti
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Nathalie Dhomen
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Eyal Gottlieb
- Cancer Metabolism Research Unit, Cancer Research UK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK.
| | - Richard Marais
- Molecular Oncology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK.
| |
Collapse
|
30
|
Fernández NB, Lorenzo D, Picco ME, Barbero G, Dergan-Dylon LS, Marks MP, García-Rivello H, Gimenez L, Labovsky V, Grumolato L, Lopez-Bergami P. ROR1 contributes to melanoma cell growth and migration by regulating N-cadherin expression via the PI3K/Akt pathway. Mol Carcinog 2015; 55:1772-1785. [PMID: 26509654 DOI: 10.1002/mc.22426] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/01/2015] [Accepted: 10/18/2015] [Indexed: 01/30/2023]
Abstract
The Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1) is primarily expressed by neural crest cells during embryogenesis. Following a complete downregulation after birth, ROR1 was shown to re-express in various types of cancers. Little is known about ROR1 expression and function in melanoma. Here we show that ROR1 is aberrantly expressed in both melanoma cell lines and tumors and that its expression associates with poor Post-Recurrence Survival of melanoma. Using gain- and loss-of-function approaches we found that ROR1 enhances both anchorage-dependent and -independent growth of melanoma cells. In addition, ROR1 decreases cell adhesion and increases cell motility and migration. Mechanistically, ROR1 was found to induce upregulation of Akt and the mesenquimal markers N-cadherin and vimentin. The regulation of N-cadherin by ROR1 relies on both Akt dependent and independent mechanisms. ROR1 does not affect Wnt canonical pathway but was found to be engaged in a positive feedback loop with Wnt5a. In summary, we show that ROR1 contributes to melanoma progression and is a candidate biomarker of poor prognosis. Although further studies are needed to confirm this possibility, the present work indicates that ROR1 is a good prospective target for melanoma cancer therapy. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Natalia Brenda Fernández
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniela Lorenzo
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Elisa Picco
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gastón Barbero
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, CONICET, Buenos Aires, Argentina
| | - Leonardo Sebastián Dergan-Dylon
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Paula Marks
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | | | - Vivian Labovsky
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luca Grumolato
- INSERM U982, Institute for Research and Innovation in Biomedicine, University of Rouen, France
| | - Pablo Lopez-Bergami
- Instituto de Medicina y Biología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina. .,Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
31
|
Cho JH, Robinson JP, Arave RA, Burnett WJ, Kircher DA, Chen G, Davies MA, Grossmann AH, VanBrocklin MW, McMahon M, Holmen SL. AKT1 Activation Promotes Development of Melanoma Metastases. Cell Rep 2015; 13:898-905. [PMID: 26565903 DOI: 10.1016/j.celrep.2015.09.057] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/31/2015] [Accepted: 09/18/2015] [Indexed: 10/22/2022] Open
Abstract
Metastases are the major cause of melanoma-related mortality. Previous studies implicating aberrant AKT signaling in human melanoma metastases led us to evaluate the effect of activated AKT1 expression in non-metastatic BRAF(V600E)/Cdkn2a(Null) mouse melanomas in vivo. Expression of activated AKT1 resulted in highly metastatic melanomas with lung and brain metastases in 67% and 17% of our mice, respectively. Silencing of PTEN in BRAF(V600E)/Cdkn2a(Null) melanomas cooperated with activated AKT1, resulting in decreased tumor latency and the development of lung and brain metastases in nearly 80% of tumor-bearing mice. These data demonstrate that AKT1 activation is sufficient to elicit lung and brain metastases in this context and reveal that activation of AKT1 is distinct from PTEN silencing in metastatic melanoma progression. These findings advance our knowledge of the mechanisms driving melanoma metastasis and may provide valuable insights for clinical management of this disease.
Collapse
Affiliation(s)
- Joseph H Cho
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - James P Robinson
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Rowan A Arave
- Department of Chemistry, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - William J Burnett
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - David A Kircher
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Guo Chen
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Allie H Grossmann
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Matthew W VanBrocklin
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA; Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Martin McMahon
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA; Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Sheri L Holmen
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA; Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA.
| |
Collapse
|
32
|
Luo C, Pietruska JR, Sheng J, Bronson RT, Hu MG, Cui R, Hinds PW. Expression of oncogenic BRAFV600E in melanocytes induces Schwannian differentiation in vivo. Pigment Cell Melanoma Res 2015; 28:603-6. [PMID: 26036358 DOI: 10.1111/pcmr.12384] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Chi Luo
- Graduate Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.,Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Jodie R Pietruska
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA.,Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Jinghao Sheng
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA.,Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Roderick T Bronson
- Rodent Histopathology Core, Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Miaofen G Hu
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Rutao Cui
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Philip W Hinds
- Graduate Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.,Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA.,Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
33
|
PICKARSKI MAUREEN, GLEASON ALEXA, BEDNAR BOHUMIL, DUONG LET. Orally active αvβ3 integrin inhibitor MK-0429 reduces melanoma metastasis. Oncol Rep 2015; 33:2737-45. [PMID: 25872534 PMCID: PMC4431436 DOI: 10.3892/or.2015.3910] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/20/2015] [Indexed: 11/16/2022] Open
Abstract
Melanoma remains one of the most aggressive types of cancer with a historically low survival rate. The αvβ3 integrin is involved in the progression of malignant melanoma. In the present study, the efficacy of MK-0429, a selective inhibitor of the αvβ3 integrin, was evaluated for its potential in the prevention of melanoma metastasis. Female B6D2F1 mice injected via the tail vein with murine B16F10 melanoma developed lung metastases within ~10 days. In the first experiment, the prevention of lung metastasis was assessed in the model treated with either vehicle, MK-0429 at 100 and 300 mg/kg orally twice daily or cyclophosphamide at 300 mg/kg, i.p. once daily. Study endpoints included determination of the study time period to achieve metastasis in lungs in this model, evaluation of the health effects on the study animals, the total number of lung colonies identified and lung tumor area. Unlike cyclophosphamide, the MK-0429 treatment did not lead to a significant weight reduction in mice. MK-0429 at 100 and 300 mg/kg reduced the number of metastatic tumor colonies by 64 and 57%, respectively, and the high dose also reduced the tumor area by 60% as compared to the vehicle. The second experiment employed B16F10 luciferase-expressing cells to examine the de novo progression of melanoma metastasis over 15 days with bioluminescent imaging of mice treated with MK-0429 at 300 mg/kg as compared to the vehicle. Tumor burden progressively advanced in the lungs of the B16F10-treated animals. However, MK-0429 reduced the progression of ventral and dorsal lung metastases by 22 and 38%, respectively, as compared to the vehicle, by study completion. Quantification of ex vivo tumor burden showed a 30-40% reduction in lung colonies by MK-0429. The two studies collectively demonstrated that MK-0429 was safe and efficacious in significantly decreasing melanoma metastasis in the lungs. The results emphasized the potential of MK-0429 as a novel, therapeutic agent for the prevention of metastatic melanoma.
Collapse
Affiliation(s)
| | - ALEXA GLEASON
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - BOHUMIL BEDNAR
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | | |
Collapse
|
34
|
Golebiewska A, Fritah S, Girotti MR. OECI-EACR precision medicine for cancer: Conference report 1-4 March 2015, Luxembourg. Ecancermedicalscience 2015; 9:519. [PMID: 25932043 PMCID: PMC4404038 DOI: 10.3332/ecancer.2015.519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Indexed: 11/25/2022] Open
Abstract
The ‘Precision Medicine for Cancer’ was the first meeting of a new series of conferences organised biannually by the European Association for Cancer Research (EACR) and the Organisation for European Cancer Institutes (OECI). The main objective of the meeting was to focus on novel topics in precision medicine by allowing strong interactions between participants and to access the speakers easily. As the first implementations of personalised medicine are appreciated in the clinic, the aim of the meeting was to further educate both researchers and clinicians and learn more from the novel approaches in the field. Similarly, the interaction between two organisations—the research-oriented EACR and the clinic-oriented OECI—was of a great value for the meeting. This OECI-EACR 2015 report will highlight the major findings of this outstanding meeting.
Collapse
Affiliation(s)
- Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (L.I.H), Luxembourg, L-1526 Luxembourg
| | - Sabrina Fritah
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (L.I.H), Luxembourg, L-1526 Luxembourg
| | - Maria Romina Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, M20 4BX, UK
| |
Collapse
|
35
|
Abstract
The RAS genes are critical oncogenic drivers activated by point mutation in some 20% of human malignancies. However, no pharmacologic approaches to targeting RAS proteins directly have yet succeeded, leading to suggestions that these proteins may be "undruggable." This has led to two alternative indirect approaches to targeting RAS function in cancer. One has been to target RAS signaling pathways downstream at tractable enzymes such as kinases, particularly in combination. The other, which is the focus of this review, has been to seek targets that are essential in cells bearing an activated RAS oncogene, but not those without. This synthetic lethal approach, while rooted in ideas from invertebrate genetics, has been inspired most strongly by the successful use of PARP inhibitors, such as olaparib, in the clinic to treat BRCA defective cancers. Several large-scale screens have been carried out using RNA interference-mediated expression silencing to find genes that are uniquely essential to RAS-mutant but not wild-type cells. These screens have been notable for the low degree of overlap between their results, with the possible exception of proteasome components, and have yet to lead to successful new clinical approaches to the treatment of RAS-mutant cancers. Possible reasons for these disappointing results are discussed here, along with a reevaluation of the approaches taken. On the basis of experience to date, RAS synthetic lethality has so far fallen some way short of its original promise and remains unproven as an approach to finding effective new ways of tackling RAS-mutant cancers. Clin Cancer Res; 21(8); 1802-9. ©2015 AACR. See all articles in this CCR Focus section, "Targeting RAS-Driven Cancers."
Collapse
Affiliation(s)
- Julian Downward
- Signal Transduction Laboratory, Francis Crick Institute, London, United Kingdom. Lung Cancer Group, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
36
|
Attaoua C, Vincent LA, Abdel Jaoued A, Hadj-Kaddour K, Baï Q, De Vos J, Vian L, Cuq P. Differential involvement of glutathione S-transferase mu 1 and multidrug resistance protein 1 in melanoma acquired resistance to vinca alkaloids. Fundam Clin Pharmacol 2014; 29:62-71. [DOI: 10.1111/fcp.12093] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/15/2014] [Accepted: 09/29/2014] [Indexed: 01/23/2023]
Affiliation(s)
- Chaker Attaoua
- Laboratoire de Toxicologie du Médicament; Institut des Biomolécules Max Mousseron (UMR5247); UFR des Sciences Pharmaceutiques et Biologiques; Université Montpellier I; 15 avenue Charles Flahault, BP14491 Montpellier 34093 France
| | - Laure-Anaïs Vincent
- Laboratoire de Toxicologie du Médicament; Institut des Biomolécules Max Mousseron (UMR5247); UFR des Sciences Pharmaceutiques et Biologiques; Université Montpellier I; 15 avenue Charles Flahault, BP14491 Montpellier 34093 France
| | - Aida Abdel Jaoued
- Laboratoire de Toxicologie du Médicament; Institut des Biomolécules Max Mousseron (UMR5247); UFR des Sciences Pharmaceutiques et Biologiques; Université Montpellier I; 15 avenue Charles Flahault, BP14491 Montpellier 34093 France
| | - Kamel Hadj-Kaddour
- Laboratoire de Toxicologie du Médicament; Institut des Biomolécules Max Mousseron (UMR5247); UFR des Sciences Pharmaceutiques et Biologiques; Université Montpellier I; 15 avenue Charles Flahault, BP14491 Montpellier 34093 France
| | - Qiang Baï
- Institut de Recherche en Biothérapie; Hôpital Saint-Eloi; 80 avenue Augustin Fliche Montpellier 34295 France
| | - John De Vos
- Institut de Recherche en Biothérapie; Hôpital Saint-Eloi; 80 avenue Augustin Fliche Montpellier 34295 France
- CHU Montpellier; Unit for Cell Therapy; Hôpital Saint-Eloi; INSERM; U1040 Montpellier F34000 France
| | - Laurence Vian
- Laboratoire de Toxicologie du Médicament; Institut des Biomolécules Max Mousseron (UMR5247); UFR des Sciences Pharmaceutiques et Biologiques; Université Montpellier I; 15 avenue Charles Flahault, BP14491 Montpellier 34093 France
| | - Pierre Cuq
- Laboratoire de Toxicologie du Médicament; Institut des Biomolécules Max Mousseron (UMR5247); UFR des Sciences Pharmaceutiques et Biologiques; Université Montpellier I; 15 avenue Charles Flahault, BP14491 Montpellier 34093 France
| |
Collapse
|
37
|
Girotti MR, Saturno G, Lorigan P, Marais R. No longer an untreatable disease: how targeted and immunotherapies have changed the management of melanoma patients. Mol Oncol 2014; 8:1140-58. [PMID: 25178978 PMCID: PMC5528622 DOI: 10.1016/j.molonc.2014.07.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 12/23/2022] Open
Abstract
The discovery that BRAF is a driver oncogene in cancer, and complementary improvements in our understanding of the immune system have resulted in new targeted and immune-therapies for metastatic melanoma. Targeted therapies achieve impressive clinical results in carefully selected patients but the development of resistance seems inevitable in most cases. Conversely, immune-checkpoints inhibitors can achieve long-term remission and cures, but in a smaller proportion of patients, and biomarkers to predict which patients will respond are not available. Nevertheless, melanoma has led the evolution of cancer treatment from relatively nonspecific cytotoxic agents to highly selective therapies and here we review the lessons from this paradigm shift in treatment and the opportunities for further improvements in outcomes for melanoma patients.
Collapse
Affiliation(s)
- Maria Romina Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, UK
| | - Grazia Saturno
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, UK
| | - Paul Lorigan
- University of Manchester, Christie NHS Foundation Trust, Manchester, UK
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, UK.
| |
Collapse
|
38
|
Peeper DS. Cancer drug resistance: old concept, novel solutions required. Mol Oncol 2014; 8:1064-6. [PMID: 25174652 DOI: 10.1016/j.molonc.2014.07.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 10/24/2022] Open
Affiliation(s)
- Daniel S Peeper
- Netherlands Cancer Institute, Division of Molecular Oncology, Amsterdam, The Netherlands.
| |
Collapse
|