1
|
Selvaggio G, Chaouiya C, Janody F. In Silico Logical Modelling to Uncover Cooperative Interactions in Cancer. Int J Mol Sci 2021; 22:ijms22094897. [PMID: 34063110 PMCID: PMC8125147 DOI: 10.3390/ijms22094897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
The multistep development of cancer involves the cooperation between multiple molecular lesions, as well as complex interactions between cancer cells and the surrounding tumour microenvironment. The search for these synergistic interactions using experimental models made tremendous contributions to our understanding of oncogenesis. Yet, these approaches remain labour-intensive and challenging. To tackle such a hurdle, an integrative, multidisciplinary effort is required. In this article, we highlight the use of logical computational models, combined with experimental validations, as an effective approach to identify cooperative mechanisms and therapeutic strategies in the context of cancer biology. In silico models overcome limitations of reductionist approaches by capturing tumour complexity and by generating powerful testable hypotheses. We review representative examples of logical models reported in the literature and their validation. We then provide further analyses of our logical model of Epithelium to Mesenchymal Transition (EMT), searching for additional cooperative interactions involving inputs from the tumour microenvironment and gain of function mutations in NOTCH.
Collapse
Affiliation(s)
- Gianluca Selvaggio
- Fondazione the Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Piazza Manifattura 1, 38068 Rovereto, Italy;
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Claudine Chaouiya
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
- CNRS, Centrale Marseille, I2M, Aix Marseille University, 13397 Marseille, France
- Correspondence: (C.C.); (F.J.)
| | - Florence Janody
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Correspondence: (C.C.); (F.J.)
| |
Collapse
|
2
|
McDougall LI, Powell RM, Ratajska M, Lynch-Sutherland CF, Hossain SM, Wiggins GAR, Harazin-Lechowska A, Cybulska-Stopa B, Motwani J, Macaulay EC, Reid G, Walker LC, Ryś J, Eccles MR. Differential Expression of BARD1 Isoforms in Melanoma. Genes (Basel) 2021; 12:320. [PMID: 33672422 PMCID: PMC7927127 DOI: 10.3390/genes12020320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/12/2021] [Accepted: 02/20/2021] [Indexed: 12/11/2022] Open
Abstract
Melanoma comprises <5% of cutaneous malignancies, yet it causes a significant proportion of skin cancer-related deaths worldwide. While new therapies for melanoma have been developed, not all patients respond well. Thus, further research is required to better predict patient outcomes. Using long-range nanopore sequencing, RT-qPCR, and RNA sequencing analyses, we examined the transcription of BARD1 splice isoforms in melanoma cell lines and patient tissue samples. Seventy-six BARD1 mRNA variants were identified in total, with several previously characterised isoforms (γ, φ, δ, ε, and η) contributing to a large proportion of the expressed transcripts. In addition, we identified four novel splice events, namely, Δ(E3_E9), ▼(i8), IVS10+131▼46, and IVS10▼176, occurring in various combinations in multiple transcripts. We found that short-read RNA-Seq analyses were limited in their ability to predict isoforms containing multiple non-contiguous splicing events, as compared to long-range nanopore sequencing. These studies suggest that further investigations into the functional significance of the identified BARD1 splice variants in melanoma are warranted.
Collapse
Affiliation(s)
- Lorissa I. McDougall
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - Ryan M. Powell
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - Magdalena Ratajska
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
- Department of Biology and Medical Genetics, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Chi F. Lynch-Sutherland
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - Sultana Mehbuba Hossain
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - George A. R. Wiggins
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (G.A.R.W.); (L.C.W.)
| | - Agnieszka Harazin-Lechowska
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 8011 Cracow, Poland; (A.H.-L.); (J.R.)
| | - Bożena Cybulska-Stopa
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 8011 Cracow, Poland;
| | - Jyoti Motwani
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - Erin C. Macaulay
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - Glen Reid
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
| | - Logan C. Walker
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (G.A.R.W.); (L.C.W.)
| | - Janusz Ryś
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 8011 Cracow, Poland; (A.H.-L.); (J.R.)
| | - Michael R. Eccles
- Department of Pathology, Otago Medical School, Dunedin Campus, University of Otago, Dunedin 9010, New Zealand; (L.I.M.); (R.M.P.); (M.R.); (C.F.L.-S.); (S.M.H.); (J.M.); (E.C.M.); (G.R.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1010, New Zealand
| |
Collapse
|
3
|
Abstract
The genetic alterations in cancer cells are tightly linked to signaling pathway dysregulation. Ras is a key molecule that controls several tumorigenesis-related processes, and mutations in RAS genes often lead to unbiased intensification of signaling networks that fuel cancer progression. In this article, we review recent studies that describe mutant Ras-regulated signaling routes and their cross-talk. In addition to the two main Ras-driven signaling pathways, i.e., the RAF/MEK/ERK and PI3K/AKT/mTOR pathways, we have also collected emerging data showing the importance of Ras in other signaling pathways, including the RAC/PAK, RalGDS/Ral, and PKC/PLC signaling pathways. Moreover, microRNA-regulated Ras-associated signaling pathways are also discussed to highlight the importance of Ras regulation in cancer. Finally, emerging data show that the signal alterations in specific cell types, such as cancer stem cells, could promote cancer development. Therefore, we also cover the up-to-date findings related to Ras-regulated signal transduction in cancer stem cells.
Collapse
Affiliation(s)
- Tamás Takács
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gyöngyi Kudlik
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Anita Kurilla
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Bálint Szeder
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - László Buday
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Department of Medical Chemistry, Semmelweis University Medical School, Budapest, Hungary
| | - Virag Vas
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary.
| |
Collapse
|
4
|
Chan XY, Singh A, Osman N, Piva TJ. Role Played by Signalling Pathways in Overcoming BRAF Inhibitor Resistance in Melanoma. Int J Mol Sci 2017; 18:ijms18071527. [PMID: 28708099 PMCID: PMC5536016 DOI: 10.3390/ijms18071527] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/06/2017] [Accepted: 07/12/2017] [Indexed: 12/15/2022] Open
Abstract
The discovery of the BRAFV600E mutation led to the development of vemurafenib (PLX4032), a selective BRAF inhibitor specific to the kinase, for the treatment of metastatic melanomas. However, initial success of the drug was dampened by the development of acquired resistance. Melanoma was shown to relapse in patients following treatment with vemurafenib which eventually led to patients' deaths. It has been proposed that mechanisms of resistance can be due to (1) reactivation of the mitogen-activated protein kinase (MAPK) signalling pathway via secondary mutations, amplification or activation of target kinase(s), (2) the bypass of oncogenic pathway via activation of alternative signalling pathways, (3) other uncharacterized mechanisms. Studies showed that receptor tyrosine kinases (RTK) such as PDGFRβ, IGF1R, EGFR and c-Met were overexpressed in melanoma cells. Along with increased secretion of growth factors such as HGF and TGF-α, this will trigger intracellular signalling cascades. This review discusses the role MAPK and Phosphatidylinositol-3-kinase-protein kinase B-mammalian target of rapamycin (PI3K-AKT-mTOR) pathways play in the mechanism of resistance of melanomas.
Collapse
Affiliation(s)
- Xian Yang Chan
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Alamdeep Singh
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Narin Osman
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
- Department of Immunology, Monash University, Melbourne 3004, Victoria, Australia.
- Department of Pharmacy, University of Queensland, Woolloongabba 4102, Queensland, Australia.
| | - Terrence J Piva
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| |
Collapse
|
5
|
Silva JM, Deuker MM, Baguley BC, McMahon M. PIK3CA-mutated melanoma cells rely on cooperative signaling through mTORC1/2 for sustained proliferation. Pigment Cell Melanoma Res 2017; 30:353-367. [PMID: 28233937 DOI: 10.1111/pcmr.12586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/20/2017] [Indexed: 01/01/2023]
Abstract
Malignant conversion of BRAF- or NRAS-mutated melanocytes into melanoma cells can be promoted by PI3'-lipid signaling. However, the mechanism by which PI3'-lipid signaling cooperates with mutationally activated BRAF or NRAS has not been adequately explored. Using human NRAS- or BRAF-mutated melanoma cells that co-express mutationally activated PIK3CA, we explored the contribution of PI3'-lipid signaling to cell proliferation. Despite mutational activation of PIK3CA, melanoma cells were more sensitive to the biochemical and antiproliferative effects of broader spectrum PI3K inhibitors than to an α-selective PI3K inhibitor. Combined pharmacological inhibition of MEK1/2 and PI3K signaling elicited more potent antiproliferative effects and greater inhibition of the cell division cycle compared to single-agent inhibition of either pathway alone. Analysis of signaling downstream of MEK1/2 or PI3K revealed that these pathways cooperate to regulate cell proliferation through mTORC1-mediated effects on ribosomal protein S6 and 4E-BP1 phosphorylation in an AKT-dependent manner. Although PI3K inhibition resulted in cytostatic effects on xenografted NRASQ61H /PIK3CAH1047R melanoma, combined inhibition of MEK1/2 plus PI3K elicited significant melanoma regression. This study provides insights as to how mutationally activated PIK3CA acts in concert with MEK1/2 signaling to cooperatively regulate mTORC1/2 to sustain PIK3CA-mutated melanoma proliferation.
Collapse
Affiliation(s)
- Jillian M Silva
- Helen Diller Family Comprehensive Cancer Center, Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Marian M Deuker
- Helen Diller Family Comprehensive Cancer Center, Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Martin McMahon
- Helen Diller Family Comprehensive Cancer Center, Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Hu S, Liu Y, Yang S, Ji K, Liu X, Zhang J, Fan R, Dong C. The effects of IGF1 on the melanogenesis in alpaca melanocytes in vitro. In Vitro Cell Dev Biol Anim 2016; 52:806-11. [PMID: 27173613 DOI: 10.1007/s11626-016-0052-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 04/25/2016] [Indexed: 11/29/2022]
Abstract
In order to investigate the effects of the insulin-like growth factor 1(IGF-1) on alpaca melanocyte in vitro, different dosees of IGF1 (0, 10, 20, 40 ng/ml) were added in the medium of alpaca melanocyte. The RTCA machine was used to monitor the proliferation, quantitative real-time PCR, and western blot to test the relative gene expression, ELISA to test cAMP production, and spectrum method to test the melanin production. The results showed that compared to the normal melanocyte, the proliferation of melanocytes was increased within 60 h following adding IGF1. It also showed that cAMP content produced by melanocytes was increased, microphthalmia-associtated transcription factor (MITF), tyrosinase (TYR) and tyrosinase-related protein 2 (TYRP2) expression was increased, and melanin production with most obvious change in 10 ng/ml supplementary group, when compared with the control group. The results suggested that IGF1 with the dose of 10 ng/ml had the important effects on the melanogenesis in alpaca melanocyte by the cAMP pathway.
Collapse
Affiliation(s)
- Shuaipeng Hu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| | - Shanshan Yang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| | - Kaiyuan Ji
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| | - Xuexian Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| | - Junzhen Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| | - Ruiwen Fan
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China.
| | - Changsheng Dong
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, Taigu, 030801, China
| |
Collapse
|
7
|
Schneider P, Schön M, Pletz N, Seitz CS, Liu N, Ziegelbauer K, Zachmann K, Emmert S, Schön MP. The novel PI3 kinase inhibitor, BAY 80-6946, impairs melanoma growth in vivo and in vitro. Exp Dermatol 2016; 23:579-84. [PMID: 24942196 DOI: 10.1111/exd.12470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 12/14/2022]
Abstract
Due to its almost universal resistance to chemotherapy, metastasized melanoma remains a major challenge in clinical oncology. Given that phosphatidyl inositol-3 kinase (PI3K) activation in melanoma cells is associated with poor prognosis, disease progression and resistance to chemotherapy, the PI3K-Akt signalling pathway is a promising therapeutic target for melanoma treatment. We analysed six human melanoma cell lines for their constitutive activation of Akt and then tested two representative lines, A375 and LOX, for their susceptibility to PI3K-inhibition by the highly specific small molecule inhibitor, BAY 80-6946. In addition, the effect of BAY 80-6946 on A375 and LOX melanoma cells was assessed in vivo in a xenotransplantation mouse model. We provide experimental evidence that specifically inhibiting the PI3K pathway and phosphorylation of Akt by this novel compound results in antitumoral activities including inhibition of proliferation, induction of apoptosis and cell cycle arrest in vitro and in vivo. However, the susceptibility did not show a clear-cut pattern and differed between the melanoma cell lines tested, resulting in in vivo growth inhibition of A375 but not LOX melanoma cells. Thus, in some cases BAY 80-6946 or related compounds may be a valuable addition to the therapeutic armamentarium.
Collapse
Affiliation(s)
- Philine Schneider
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Sweetlove M, Wrightson E, Kolekar S, Rewcastle GW, Baguley BC, Shepherd PR, Jamieson SMF. Inhibitors of pan-PI3K Signaling Synergize with BRAF or MEK Inhibitors to Prevent BRAF-Mutant Melanoma Cell Growth. Front Oncol 2015; 5:135. [PMID: 26137449 PMCID: PMC4468830 DOI: 10.3389/fonc.2015.00135] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/01/2015] [Indexed: 11/13/2022] Open
Abstract
BRAF and MEK inhibitors have improved outcomes for patients with BRAF-mutant melanoma, but their efficacy is limited by both intrinsic and acquired resistances. Activation of the PI3K pathway can mediate resistance to these agents, providing a strong rationale for combination therapy in melanoma. Here, a panel of nine low-passage human metastatic melanoma cell lines with BRAF mutations was tested in cell proliferation and protein expression assays for sensitivity to inhibitors of MEK (selumetinib) and BRAF (vemurafenib) as single agents and in combination with inhibitors of pan-PI3K (ZSTK474), pan-PI3K/mTOR (BEZ235), individual PI3K isoforms (p110α, A66; p110β, TGX-221; p110γ, AS-252424; p110δ, idelalisib), or mTORC1/2 (KU-0063794). Selumetinib and vemurafenib potently inhibited cell proliferation in all cell lines, especially in those that expressed low levels of phosphorylated AKT (pAKT). ZSTK474 and BEZ235 also inhibited cell proliferation in all cell lines and enhanced the antitumor activity of selumetinib and vemurafenib in the majority of lines by either interacting synergistically or additively to increase potency or by inducing cytotoxicity by significantly increasing the magnitude of cell growth inhibition. Furthermore, ZSTK474 or BEZ235 combined with selumetinib to produce robust inhibition of pERK, pAKT, and pS6 expression and synergistic inhibition of NZM20 tumor growth. The inhibitors of individual PI3K isoforms or mTORC1/2 were less effective at inhibiting cell proliferation either as single agents or in combination with selumetinib or vemurafenib, although KU-0063794 synergistically interacted with vemurafenib and increased the magnitude of cell growth inhibition with selumetinib or vemurafenib in certain cell lines. Overall, these results suggest that the sensitivity of BRAF-mutant melanoma cells to BRAF or MEK inhibitors is at least partly mediated by activation of the PI3K pathway and can be enhanced by combined inhibition of the BRAF/MEK and PI3K/mTOR signaling pathways.
Collapse
Affiliation(s)
- Melanie Sweetlove
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand
| | - Emma Wrightson
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand
| | - Sharada Kolekar
- Department of Molecular Medicine and Pathology, The University of Auckland , Auckland , New Zealand
| | - Gordon W Rewcastle
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| |
Collapse
|
9
|
Deuker MM, Marsh Durban V, Phillips WA, McMahon M. PI3'-kinase inhibition forestalls the onset of MEK1/2 inhibitor resistance in BRAF-mutated melanoma. Cancer Discov 2015; 5:143-53. [PMID: 25472943 PMCID: PMC4320669 DOI: 10.1158/2159-8290.cd-14-0856] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
UNLABELLED Phosphatidylinositide 3' (PI3')-lipid signaling cooperates with oncogenic BRAF(V600E) to promote melanomagenesis. Sustained PI3'-lipid production commonly occurs via silencing of the PI3'-lipid phosphatase PTEN or, less commonly, through mutational activation of PIK3CA, encoding the 110-kDa catalytic subunit of PI3'-kinase-α (PI3Kα). To define the PI3K catalytic isoform dependency of BRAF-mutated melanoma, we used pharmacologic, isoform-selective PI3K inhibitors in conjunction with melanoma-derived cell lines and genetically engineered mouse (GEM) models. Although BRAF(V600E)/PIK3CA(H1047R) melanomas were sensitive to the antiproliferative effects of selective PI3Kα blockade, inhibition of BRAF(V600E)/PTEN(Null) melanoma proliferation required combined blockade of PI3Kα, PI3Kδ, and PI3Kγ, and was insensitive to PI3Kβ blockade. In GEM models, isoform-selective PI3K inhibition elicited cytostatic effects, but significantly potentiated melanoma regression in response to BRAF(V600E) pathway-targeted inhibition. Interestingly, PI3K inhibition forestalled the onset of MEK inhibitor resistance in two independent GEM models of BRAF(V600E)-driven melanoma. These results suggest that combination therapy with PI3K inhibitors may be a useful strategy to extend the duration of clinical response of patients with BRAF-mutated melanoma to BRAF(V600E) pathway-targeted therapies. SIGNIFICANCE Although BRAF(V600E) pathway-targeted therapies elicit melanoma regression, the onset of drug resistance limits the durability of response. Here, we show that combined treatment with PI3K inhibitors significantly forestalled the onset of MEK1/2 inhibitor-resistant disease in BRAF-mutated GEM melanoma models. These results provide a conceptual framework for the combined deployment of BRAF(V600E) plus PI3K pathway-targeted inhibitors in the treatment of a subset of patients with BRAF-mutated melanoma.
Collapse
Affiliation(s)
- Marian M Deuker
- Helen Diller Family Comprehensive Cancer Center and Department of Cell and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - Victoria Marsh Durban
- Helen Diller Family Comprehensive Cancer Center and Department of Cell and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | | | - Martin McMahon
- Helen Diller Family Comprehensive Cancer Center and Department of Cell and Molecular Pharmacology, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
10
|
Leung EY, Kim JE, Askarian-Amiri M, Rewcastle GW, Finlay GJ, Baguley BC. Relationships between signaling pathway usage and sensitivity to a pathway inhibitor: examination of trametinib responses in cultured breast cancer lines. PLoS One 2014; 9:e105792. [PMID: 25170609 PMCID: PMC4149495 DOI: 10.1371/journal.pone.0105792] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 07/23/2014] [Indexed: 11/19/2022] Open
Abstract
Cellular signaling pathways involving mTOR, PI3K and ERK have dominated recent studies of breast cancer biology, and inhibitors of these pathways have formed a focus of numerous clinical trials. We have chosen trametinib, a drug targeting MEK in the ERK pathway, to address two questions. Firstly, does inhibition of a signaling pathway, as measured by protein phosphorylation, predict the antiproliferative activity of trametinib? Secondly, do inhibitors of the mTOR and PI3K pathways synergize with trametinib in their effects on cell proliferation? A panel of 30 human breast cancer cell lines was chosen to include lines that could be classified according to whether they were ER and PR positive, HER2 over-expressing, and “triple negative”. Everolimus (targeting mTOR), NVP-BEZ235 and GSK2126458 (both targeting PI3K/mTOR) were chosen for combination experiments. Inhibition of cell proliferation was measured by IC50 values and pathway utilization was measured by phosphorylation of signaling kinases. Overall, no correlation was found between trametinib IC50 values and inhibition of ERK signaling. Inhibition of ERK phosphorylation was observed at trametinib concentrations not affecting proliferation, and sensitivity of cell proliferation to trametinib was found in cell lines with low ERK phosphorylation. Evidence was found for synergy between trametinib and either everolimus, NVP-BEZ235 or GSK2126458, but this was cell line specific. The results have implications for the clinical application of PI3K/mTOR and MEK inhibitors.
Collapse
Affiliation(s)
- Euphemia Y. Leung
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- * E-mail: (EYL); (BCB)
| | - Ji Eun Kim
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Marjan Askarian-Amiri
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Gordon W. Rewcastle
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Graeme J. Finlay
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Bruce C. Baguley
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- * E-mail: (EYL); (BCB)
| |
Collapse
|
11
|
Selective cellular uptake and retention of SN 28049, a new DNA-binding topoisomerase II-directed antitumor agent. Cancer Chemother Pharmacol 2014; 74:25-35. [DOI: 10.1007/s00280-014-2469-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 04/12/2014] [Indexed: 10/25/2022]
|
12
|
D'mello SAN, Flanagan JU, Green TN, Leung EY, Askarian-Amiri ME, Joseph WR, McCrystal MR, Isaacs RJ, Shaw JHF, Furneaux CE, During MJ, Finlay GJ, Baguley BC, Kalev-Zylinska ML. Evidence That GRIN2A Mutations in Melanoma Correlate with Decreased Survival. Front Oncol 2014; 3:333. [PMID: 24455489 PMCID: PMC3888952 DOI: 10.3389/fonc.2013.00333] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 12/30/2013] [Indexed: 12/17/2022] Open
Abstract
Previous whole-exome sequencing has demonstrated that melanoma tumors harbor mutations in the GRIN2A gene. GRIN2A encodes the regulatory GluN2A subunit of the glutamate-gated N-methyl-d-aspartate receptor (NMDAR), involvement of which in melanoma remains undefined. Here, we sequenced coding exons of GRIN2A in 19 low-passage melanoma cell lines derived from patients with metastatic melanoma. Potential mutation impact was evaluated in silico, including within the GluN2A crystal structure, and clinical correlations were sought. We found that of 19 metastatic melanoma tumors, four (21%) carried five missense mutations in the evolutionarily conserved domains of GRIN2A; two were previously reported. Melanoma cells that carried these mutations were treatment-naïve. Sorting intolerant from tolerant analysis predicted that S349F, G762E, and P1132L would disrupt protein function. When modeled into the crystal structure of GluN2A, G762E was seen to potentially alter GluN1-GluN2A interactions and ligand binding, implying disruption to NMDAR functionality. Patients whose tumors carried non-synonymous GRIN2A mutations had faster disease progression and shorter overall survival (P < 0.05). This was in contrast to the BRAF V600E mutation, found in 58% of tumors but showing no correlation with clinical outcome (P = 0.963). Although numbers of patients in this study are small, and firm conclusions about the association between GRIN2A mutations and poor clinical outcome cannot be drawn, our results highlight the high prevalence of GRIN2A mutations in metastatic melanoma and suggest for the first time that mutated NMDARs impact melanoma progression.
Collapse
Affiliation(s)
- Stacey Ann N D'mello
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland , Auckland , New Zealand
| | - Jack U Flanagan
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland , Auckland , New Zealand
| | - Taryn N Green
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland , Auckland , New Zealand
| | - Euphemia Y Leung
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | | | - Wayne R Joseph
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Michael R McCrystal
- Department of Clinical Oncology, Auckland District Health Board , Auckland , New Zealand ; Canopy Cancer Care, Mercy Hospital , Auckland , New Zealand
| | - Richard J Isaacs
- Regional Cancer Treatment Service, Palmerston North Public Hospital , Palmerston North , New Zealand
| | | | | | - Matthew J During
- Department of Molecular Virology, Immunology and Medical Genetics, Neuroscience and Neurological Surgery, Ohio State University , Columbus, OH , USA ; Centre for Brain Research, University of Auckland , Auckland , New Zealand
| | - Graeme J Finlay
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Maggie L Kalev-Zylinska
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland , Auckland , New Zealand ; LabPlus Haematology, Auckland District Health Board , Auckland , New Zealand
| |
Collapse
|
13
|
Marsh Durban V, Deuker MM, Bosenberg MW, Phillips W, McMahon M. Differential AKT dependency displayed by mouse models of BRAFV600E-initiated melanoma. J Clin Invest 2013; 123:5104-18. [PMID: 24200692 PMCID: PMC3859393 DOI: 10.1172/jci69619] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 09/03/2013] [Indexed: 01/09/2023] Open
Abstract
Malignant melanoma is frequently driven by mutational activation of v-raf murine sarcoma viral oncogene homolog B1 (BRAF) accompanied by silencing of the phosphatase and tensin homology (PTEN) tumor suppressor. Despite the implied importance of PI3K signaling in PTENNull melanomas, mutational activation of the gene encoding the catalytic subunit of PI3Kα (PIK3CA), is rarely detected. Since PTEN has both PI3-lipid phosphatase-dependent and -independent tumor suppressor activities, we investigated the contribution of PI3K signaling to BRAFV600E-induced melanomagenesis using mouse models, cultured melanoma cells, and PI3K pathway-targeted inhibitors. These experiments revealed that mutationally activated PIK3CAH1047R cooperates with BRAFV600E for melanomagenesis in mice. Moreover, pharmacological inhibition of PI3Ks prevented growth of BRAFV600E/PTENNull melanomas in vivo and in tissue culture. Combined inhibition of BRAFV600E and PI3K had more potent effects on the regression of established BRAFV600E/PTENNull melanomas and cultured melanoma cells than individual blockade of either pathway. Surprisingly, growth of BRAFV600E/PIK3CAH1047R melanomas was dependent on the protein kinase AKT; however, AKT inhibition had no effect on growth of BRAFV600E/PTENNull melanomas. These data indicate that PTEN silencing contributes a PI3K-dependent, but AKT-independent, function in melanomagenesis. Our findings enhance our knowledge of how BRAFV600E and PI3K signaling cooperate in melanomagenesis and provide preclinical validation for combined pathway-targeted inhibition of PI3K and BRAFV600E in the therapeutic management of BRAFV600E/PTENNull melanomas.
Collapse
Affiliation(s)
- Victoria Marsh Durban
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Marian M. Deuker
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Marcus W. Bosenberg
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Wayne Phillips
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Martin McMahon
- Helen Diller Family Comprehensive Cancer Center, Department of Cell and Molecular Pharmacology, UCSF, San Francisco, California, USA.
Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA.
Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Lukka PB, Chen YY, Finlay GJ, Joseph WR, Richardson E, Paxton JW, Baguley BC. Tumour tissue selectivity in the uptake and retention of SN 28049, a new topoisomerase II-directed anticancer agent. Cancer Chemother Pharmacol 2013; 72:1013-22. [PMID: 24036845 DOI: 10.1007/s00280-013-2280-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/27/2013] [Indexed: 01/03/2023]
Abstract
PURPOSE A variety of anticancer drugs, including doxorubicin and mitoxantrone, have structures in which a DNA-intercalating chromophore is linked to a positively charged side chain. These drugs generally inhibit tumour growth and survival by poisoning the enzyme DNA topoisomerase II. SN 28049, a benzonaphthyridine derivative with these properties, has curative activity against the Colon 38 tumour in mice. Previous pharmacokinetic studies have demonstrated tumour-selective retention with approximately 20-fold higher area under the concentration-time curve (AUC) for tumour tissue as compared to normal tissues. We have investigated here whether such retention is tumour specific. METHODS Plasma and tissue pharmacokinetics were assessed in the murine Lewis lung (LL3) tumour in C57 BL/6 mice and in xenografts of the NZM4, NZM10 and NZM52 human melanoma lines in Balb/c Rag-1 immunodeficient mice. The in vitro cellular localisation of SN 28049 in murine and human cell lines was studied by confocal fluorescence microscopy. RESULTS A 260-fold variation, from 8.9 μM h (NZM4) to 2,334 μM h (Colon 38), was found among the different tumours. Only small variations were observed in the corresponding plasma AUC (2.9-5 μM h). Moreover, in vivo activity, as measured by tumour growth delay, varied from 1 day (NZM4) to curative (Colon 38), consistent with the tumour pharmacokinetic data. In cultured cell lines, SN 28049 was found in cytoplasmic bodies, suggesting that drug sequestration could contribute to tumour pharmacokinetics. CONCLUSION SN 28049 shows dramatic differences in both tumour AUC and antitumour activity against different tumours. These differences point to the presence of a tumour-specific uptake and retention mechanism.
Collapse
Affiliation(s)
- Pradeep B Lukka
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | | | | | | | | | | | | |
Collapse
|
15
|
Stepanenko AA, Vassetzky YS, Kavsan VM. Antagonistic functional duality of cancer genes. Gene 2013; 529:199-207. [PMID: 23933273 DOI: 10.1016/j.gene.2013.07.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/08/2013] [Accepted: 07/09/2013] [Indexed: 12/21/2022]
Abstract
Cancer evolution is a stochastic process both at the genome and gene levels. Most of tumors contain multiple genetic subclones, evolving in either succession or in parallel, either in a linear or branching manner, with heterogeneous genome and gene alterations, extensively rewired signaling networks, and addicted to multiple oncogenes easily switching with each other during cancer progression and medical intervention. Hundreds of discovered cancer genes are classified according to whether they function in a dominant (oncogenes) or recessive (tumor suppressor genes) manner in a cancer cell. However, there are many cancer "gene-chameleons", which behave distinctly in opposite way in the different experimental settings showing antagonistic duality. In contrast to the widely accepted view that mutant NADP(+)-dependent isocitrate dehydrogenases 1/2 (IDH1/2) and associated metabolite 2-hydroxyglutarate (R)-enantiomer are intrinsically "the drivers" of tumourigenesis, mutant IDH1/2 inhibited, promoted or had no effect on cell proliferation, growth and tumorigenicity in diverse experiments. Similar behavior was evidenced for dozens of cancer genes. Gene function is dependent on genetic network, which is defined by the genome context. The overall changes in karyotype can result in alterations of the role and function of the same genes and pathways. The diverse cell lines and tumor samples have been used in experiments for proving gene tumor promoting/suppressive activity. They all display heterogeneous individual karyotypes and disturbed signaling networks. Consequently, the effect and function of gene under investigation can be opposite and versatile in cells with different genomes that may explain antagonistic duality of cancer genes and the cell type- or the cellular genetic/context-dependent response to the same protein. Antagonistic duality of cancer genes might contribute to failure of chemotherapy. Instructive examples of unexpected activity of cancer genes and "paradoxical" effects of different anticancer drugs depending on the cellular genetic context/signaling network are discussed.
Collapse
Affiliation(s)
- A A Stepanenko
- State Key Laboratory of Molecular and Cellular Biology, Institute of Molecular Biology and Genetics, 150 Zabolotnogo Street, Kyiv 03680, Ukraine.
| | | | | |
Collapse
|
16
|
Kim JE, Leung E, Baguley BC, Finlay GJ. Heterogeneity of expression of epithelial-mesenchymal transition markers in melanocytes and melanoma cell lines. Front Genet 2013; 4:97. [PMID: 23755070 PMCID: PMC3668138 DOI: 10.3389/fgene.2013.00097] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 05/14/2013] [Indexed: 11/13/2022] Open
Abstract
The epithelial–mesenchymal transition (EMT) describes a reversible switch from an epithelial-like to a mesenchymal-like phenotype. It is essential for the development of the normal epithelium and also contributes to the invasive properties of carcinomas. At the molecular level, the EMT transition is characterized by a series of coordinated changes including downregulation of the junctional protein E-cadherin (CDH1), up-regulation of transcriptional repressors of E-cadherin such as Snail (SNAI1) and Slug (SNAI2), and up-regulation of N-cadherin. We wished to determine whether cultured normal melanocytes and melanoma cell lines, which are derived from the neural crest, showed signs of a similarly coordinated phenotypic switch. We investigated normal melanocytes and 25 cell lines derived from New Zealand patients with metastatic melanoma. Most lines had been previously genotyped for common mutations such as BRAF, NRAS, PIK3CA (phosphatidylinositol-3-kinase), TP53 (p53), and CDKN2A (p16). Expression of E-cadherin, N-cadherin, microphthalmia-associated transcription factor (MITF), Snail, Slug, Axl, p53, and Hdm2 was compared by western blotting. Normal melanocytes expressed each of these proteins except for Snail, while normal melanocytes and almost every melanoma line expressed Slug. Expression of individual markers among different melanoma lines varied from high to low or undetectable. Quantitation of western blots showed that expression of MITF-M, the melanocyte-specific isoform of MITF, was positively related to that of E-cadherin but inversely related to that of N-cadherin and Axl. There was also no apparent relationship between expression of any particular marker and the presence of BRAF, NRAS, PIK3CA, TP53, or CDKN2A mutations. The results suggest that melanomas do not show the classical epithelial and mesenchymal phenotypes but rather display either high E-cadherin/high MITF-M expression on one hand, or high N-cadherin/high Axl expression on the other. These may correspond to differentiated and invasive phenotypes in vivo.
Collapse
Affiliation(s)
- Ji Eun Kim
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland Auckland, New Zealand
| | | | | | | |
Collapse
|
17
|
Kim JE, Finlay GJ, Baguley BC. The role of the hippo pathway in melanocytes and melanoma. Front Oncol 2013; 3:123. [PMID: 23720711 PMCID: PMC3655322 DOI: 10.3389/fonc.2013.00123] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 05/03/2013] [Indexed: 12/13/2022] Open
Abstract
The Hippo signaling pathway comprises a series of cytoplasmic tumor suppressor proteins including Merlin and the Lats1/2 and MST1/2 kinases, and is thought to play a critical role in determining the sizes of organs and tissues. The Hippo pathway is regulated upstream by extracellular mechanosensory signaling arising from cell shape and polarity, as well as by a variety of extracellular signaling molecules. When active, the pathway maintains the transcriptional activators Yes-associated protein (YAP) and TAZ in phosphorylated forms in the cytoplasm, preventing cell proliferation. When the Hippo pathway is inactivated, YAP and TAZ are translocated to the nucleus and induce the expression of a variety of proteins concerned with entry into the cell division cycle, such as cyclin D1 and Fox M1, as well as the inhibition of apoptosis. The failure of the Hippo pathway has been implicated in the development of many different types of cancer but there is limited information available as to its involvement in melanoma. We hypothesize here firstly that the Hippo pathway is involved in maintaining density of cutaneous melanocytes on the basement membrane at the junction of the epidermis and the dermis, and secondly, that its function is disturbed in melanoma. We have analyzed a series of 23 low passage human melanoma lines as well as cultured normal melanoma, and find that melanocytes, as well as all melanoma cell lines examined express TAZ. Melanocytes and most melanoma lines also express YAP. E-cadherin, an upstream regulator of the Hippo pathway, and Axl, a receptor tyrosine kinase regulated by the Hippo pathway, are expressed in melanocytes and in several melanoma cell lines. These observations, together with published evidence for the presence of Merlin, Lats1/2, and MST1/2 in melanocytes and melanoma cells, support the hypothesis that the Hippo pathway is an important component of melanocyte and melanoma behavior.
Collapse
Affiliation(s)
- Ji Eun Kim
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland Auckland, New Zealand
| | | | | |
Collapse
|
18
|
Stones CJ, Kim JE, Joseph WR, Leung E, Marshall ES, Finlay GJ, Shelling AN, Baguley BC. Comparison of responses of human melanoma cell lines to MEK and BRAF inhibitors. Front Genet 2013; 4:66. [PMID: 23658559 PMCID: PMC3647113 DOI: 10.3389/fgene.2013.00066] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/09/2013] [Indexed: 11/13/2022] Open
Abstract
The NRAS and BRAF genes are frequently mutated in melanoma, suggesting that the NRAS-BRAF-MEK-ERK signaling pathway is an important target for therapy. Two classes of drugs, one targeting activated BRAF and one targeting MEK, are currently undergoing clinical evaluation. We have analysed the NRAS and BRAF mutational status of a series of 44 early passage lines developed from New Zealand patients with metastatic melanoma. 41% of the lines analysed had BRAF mutations, 23% had NRAS mutations, and 36% had neither. We then determined IC50 values (drug concentrations for 50% growth inhibition) for CI-1040, a commonly used inhibitor of MEK kinase; trametinib, a clinical agent targeting MEK kinase; and vemurafenib, an inhibitor of mutant BRAF kinase. Cell lines with activating BRAF mutations were significantly more sensitive to vemurafenib than lines with NRAS mutations or lines lacking either mutation (p < 0.001). IC50 values for CI-1040 and trametinib were strongly correlated (r = 0.98) with trametinib showing ~100-fold greater potency. Cell lines sensitive to vemurafenib were also sensitive to CI-1040 and trametinib, but there was no relationship between IC50 values and NRAS mutation status. A small number of lines lacking a BRAF mutation were sensitive to CI-1040 but resistant to vemurafenib. We used western blotting to investigate the effect on ERK phosphorylation of CI-1040 in four lines, of vemurafenib in two lines and of trametinib in two lines. The results support the view that MEK inhibitors might be combined with BRAF inhibitors in the treatment of melanomas with activated BRAF. The high sensitivity to trametinib of some lines with wildtype BRAF status also suggests that MEK inhibitors could have a therapeutic effect against some melanomas as single agents.
Collapse
Affiliation(s)
- Clare J Stones
- Department of Obstetrics and Gynaecology, The University of Auckland Auckland, New Zealand ; Auckland Cancer Society Research Centre, The University of Auckland Auckland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|