1
|
Deng M, Ren B, Zhao J, Guo X, Yang Y, Shi H, Bian X, Wu M, Xu C, Tao M, Liang R, Li Q. Temozolomide-Promoted MGMT Transcription Contributes to Chemoresistance by Activating the ERK Signalling Pathway in Malignant Melanoma. J Cell Mol Med 2025; 29:e70380. [PMID: 39873425 PMCID: PMC11773391 DOI: 10.1111/jcmm.70380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Tumour cells possess a multitude of chemoresistance mechanisms, which could plausibly contribute to the ineffectiveness of chemotherapy. O6-methylguanine-DNA methyltransferase (MGMT) is an important effector protein associated with Temozolomide (TMZ) resistance in various tumours. To some extent, the expression level of MGMT determines the sensitivity of cells to TMZ, but the mechanism of its expression regulation has not been fully elucidated. Cultured malignant melanoma cell lines A375 and Sk-MEL28 were employed. A luciferase assay was used to detect the transcriptional activity of the MGMT promoter. Western blotting was used to compare the expression levels of phosphorylated ERK1/2 (P-ERK1/2) after TMZ treatment. Immunofluorescent staining was used to detect TMZ-induced DNA damage protein levels. The sensitivity of melanoma cells to TMZ was detected by MTT assay and animal experiments. The expression of MGMT mRNA was tested by Quantitative real-time PCR (RT-qPCR). Flow cytometry was used to measure the apoptosis of TMZ-treated cells. TMZ enhanced the transcription of MGMT through activating the ERK pathway. ERK inhibitors U0126 and vemurafenib (vMF) inhibited the TMZ induced transcription of MGMT. The expression of MGMT and p-ERK1/2 was closely related in human MM tissues. vMF increased the sensitivity of melanoma (MM) to TMZ in vitro and in vivo through downregulating MGMT and promoting the TMZ induced DNA damage in MM. TMZ-promoted MGMT transcription contributed to instinctive chemoresistance by activating the ERK signalling pathway in malignant melanoma. Our study indicates that the use of the ERK inhibitor in combination with TMZ could potentially enhance the effectiveness of clinical treatment for malignant melanoma.
Collapse
Affiliation(s)
- Meiyi Deng
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouJiangsuChina
- Suzhou Sano Precision Medicine LtdSuzhouJiangsuChina
| | - Bingjie Ren
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of OncologyNanyang Second General HospitalNanyangHenanChina
| | - Jing Zhao
- Department of Radiation OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xia Guo
- Department of PathologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yuanyuan Yang
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Huiling Shi
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouJiangsuChina
| | - Xuyu Bian
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Mengyao Wu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Caihua Xu
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Min Tao
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouJiangsuChina
- Suzhou Sano Precision Medicine LtdSuzhouJiangsuChina
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Rongrui Liang
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouJiangsuChina
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Qiang Li
- Department of ChemotherapyJiangxi Cancer HospitalNanchangJiangxiChina
| |
Collapse
|
2
|
Pham JP, Joshua AM, da Silva IP, Dummer R, Goldinger SM. Chemotherapy in Cutaneous Melanoma: Is There Still a Role? Curr Oncol Rep 2023; 25:609-621. [PMID: 36988735 PMCID: PMC10164011 DOI: 10.1007/s11912-023-01385-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 03/30/2023]
Abstract
Abstract
Purpose of Review
In the preceding decade, the management of metastatic cutaneous melanoma has been revolutionised with the development of highly effective therapies including immune checkpoint inhibitors (specifically CTLA-4 and PD-1 inhibitors) and targeted therapies (BRAF and MEK inhibitors). The role of chemotherapy in the contemporary management of melanoma is undefined.
Recent Findings
Extended analyses highlight substantially improved 5-year survival rates of approximately 50% in patients with metastatic melanoma treated with first-line therapies. However, most patients will progress on these first-line treatments. Sequencing of chemotherapy following failure of targeted and immunotherapies is associated with low objective response rates and short progression-free survival, and thus, meaningful benefits to patients are minimal.
Summary
Chemotherapy has limited utility in the contemporary management of cutaneous melanoma (with a few exceptions, discussed herein) and should not be the standard treatment sequence following failure of first-line therapies. Instead, enrolment onto clinical trials should be standard-of-care in these patients.
Collapse
Affiliation(s)
- James P Pham
- Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst, NSW, Australia
| | - Anthony M Joshua
- Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst, NSW, Australia
- Melanoma Institute Australia, The University of Sydney, Wollstonecraft, NSW, Australia
| | - Ines P da Silva
- Melanoma Institute Australia, The University of Sydney, Wollstonecraft, NSW, Australia
- Medical Oncology, Blacktown Hospital, Blacktown, NSW, Australia
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Simone M Goldinger
- Department of Dermatology, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Polymethoxyflavones from Gardenia oudiepe (Rubiaceae) induce cytoskeleton disruption-mediated apoptosis and sensitize BRAF-mutated melanoma cells to chemotherapy. Chem Biol Interact 2020; 325:109109. [PMID: 32376239 DOI: 10.1016/j.cbi.2020.109109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/18/2020] [Accepted: 04/13/2020] [Indexed: 11/21/2022]
Abstract
A series of 10 natural and semisynthetic flavonoids (1 to 10) were obtained from Gardenia oudiepe (Rubiaceae), an endemic plant from New Caledonia. Most of them were polymethoxylated flavones (PMFs) of rare occurrence. After a cell viability screening test, PMFs 2 and 3 showed significant cytotoxic activity against A2058 human melanoma cells (IC50 = 3.92 and 8.18 μM, respectively) and were selected for in-depth pharmacological assays. Both compounds inhibited cell migration and induced apoptosis and cell cycle arrest after 72h of treatment. Immunofluorescence assays indicated that these outcomes were possibly related to the induction of cytoskeleton disruption associated to actin and tubulin depolymerization. These data were confirmed by molecular docking studies, which showed a good interaction between PMFs 2 and 3 and tubulin, particularly at the colchicine binding site. As A2058 are considered as chemoresistant to conventional chemotherapy, compounds 2 and 3 (½IC50) were associated to clinically-used antimelanoma drugs (vemurafenib and dacarbazine) and combined therapies efficacy was assessed by the MTT assay. PMFs 2 restored the sensitivity of A2058 cells to dacarbazine treatment (IC50 = 49.38 μM vs. >100 μM). Taken together, these data suggest that PMFs from G. oudiepe could be potential leaders for the design of new antimelanoma drugs.
Collapse
|
4
|
de Oliveira-Júnior RG, Nicolau E, Bonnet A, Prunier G, Beaugeard L, Joguet N, Thiéry V, Picot L. Carotenoids from Rhodomonas salina Induce Apoptosis and Sensitize A2058 Melanoma Cells to Chemotherapy. REVISTA BRASILEIRA DE FARMACOGNOSIA 2020. [DOI: 10.1007/s43450-020-00036-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
5
|
Abstract
Pharmacogenomics, a key part of precision medicine, has the potential to tailor drug therapy with the high safety margins and optimized response. Before the Human Genome Project in 2003, articles on pharmacogenomics were limited. The last 15 years have seen significant growth in the field, with gene variant discoveries that play essential roles in individuals' drug response. Dermatology has started to take advantage of this information. This overview shows how pharmacogenomic databases have expanded in the treatment of skin diseases and provides a list of current dermatologic drugs with their FDA-approved biomarkers and clinical implementation.
Collapse
Affiliation(s)
- Le Hanh Dung Do
- a Pham Ngoc Thach University of Medicine , Ho Chi Minh city , Vietnam
| | - Howard Maibach
- b Department of Dermatology, School of Medicine , University of California , San Francisco , CA , USA
| |
Collapse
|
6
|
Shirkavand A, Farivar S, Mohajerani E, Ataie-Fashtami L, Ghazimoradi MH. Non-invasive Reflectance Spectroscopy for Normal and Cancerous Skin Cells Refractive Index Determination: An In Vitro Study. Lasers Surg Med 2019; 51:742-750. [PMID: 31094015 DOI: 10.1002/lsm.23095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE Optical reflectance spectroscopy is a non-invasive technique for optical characterization of biological samples. Any alteration in a cell from normal or carcinogenic causes will change its refractive index. The aim of this study is to develop a computerized program for extraction of a refractive index of normal and cancerous skin cell lines, including melanoma, fibroblast, and adipose cells, using visible near-infrared reflectance spectra and the Kramers-Kronig (K-K) relations. MATERIALS AND METHOD A fiber optic reflectance spectrometer in visible near-infrared wavelength was used for spectrum acquisition in an in vitro study. Human skin cell lines for melanoma (A375), fibroblast, and adipose sample were cultured for optical spectroscopy. Following data acquisition, an analytical MATLAB code was developed to run the K-K relations. The program was validated for three biological samples using an Abbe refractometer. RESULTS The validation error (below 5%) and determination of changes in the refractive index of melanoma, normal fibroblasts, and adipose skin cells was carried out at wavelengths of 450-950 nm. The refractive index of melanoma was 1.59270 ± 0.0550 at 450 nm, the minimum amount of 1.27790 ± 0.0550 to 1.321 ± 0.0550 at 620 nm, and rose sharply to 1.44321 ± 0.0550 at 935 nm. The respective results for fibroblast and adipose tissue cells were 1.33282 ± 0.0134 and 1.28345 ± 0.0163 at 450 nm with an increasing trend to 1.30494 ± 0.0135 and 1.26716 ± 0.0163 at 935 nm. CONCLUSION Refractive index characteristics show potential for cancer screening and diagnosis. The results show that optical spectroscopy is a promising, non-invasive tool for assessment of the refractive index of living biological cells in in vitro settings. Tracking changes in the refractive index allows screening of normal and abnormal cells for probable alterations in a non-invasive label-free method. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Afshan Shirkavand
- POMP Lab, Photonics Department, Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, 1983969411, Iran
| | - Shirin Farivar
- Genetics, Stem Cells, Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 1983963113, Iran
| | - Ezeddin Mohajerani
- POMP Lab, Photonics Department, Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, 1983969411, Iran
| | - Leila Ataie-Fashtami
- Department of Regenerative Medicine, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Mohammad H Ghazimoradi
- Genetics, Stem Cells, Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 1983963113, Iran
| |
Collapse
|
7
|
Mokhamatam RB, Sahoo BK, Manna SK. Suppression of microphthalmia-associated transcription factor, but not NF-kappa B sensitizes melanoma specific cell death. Apoptosis 2018; 21:928-40. [PMID: 27325430 DOI: 10.1007/s10495-016-1260-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Mutation in B-Raf leads to gain of function in melanoma and causes aggressive behavior for proliferation. Most of the therapeutics are ineffective in this scenario. However, regulation of this aggressive behavior by targeting the key molecules would be viable strategy to develop novel and effective therapeutics. In this report we provide evidences that the resveratrol is potent to regulate melanoma cell growth than other inducers of apoptosis. Resveratrol inhibits pronounced cell proliferation in melanoma than other tumor cell types. Cell cycle analysis using flow cytometry shows that the treatment with resveratrol results in S phase arrest. Resveratrol inhibits microphthalmia-associated transcription factor (MITF) and its dependent genes without interfering the MITF DNA binding in vitro. Resveratrol-mediated cell death is protected in MITF overexpressed cells and it is aggravated in MITF knocked down cells. These suggest the resveratrol-mediated decrease in MITF is the possible cause of melanoma cell death. Though resveratrol-mediated downregulation of NF-κB is responsible for cell apoptosis, but the downregulation of MITF is the main reason for melanoma-specific cell death. Thus, resveratrol can be effective chemotherapeutic agent against rapid proliferative melanoma cells.
Collapse
Affiliation(s)
- Raveendra B Mokhamatam
- Laboratory of Immunology, Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad, Telangana, 500 001, India.,Graduate studies, Manipal University, Manipal, Karnataka, 576104, India
| | - Binay K Sahoo
- Laboratory of Immunology, Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad, Telangana, 500 001, India
| | - Sunil K Manna
- Laboratory of Immunology, Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad, Telangana, 500 001, India.
| |
Collapse
|
8
|
Gupta A, Gomes F, Lorigan P. The role for chemotherapy in the modern management of melanoma. Melanoma Manag 2017; 4:125-136. [PMID: 30190915 PMCID: PMC6094602 DOI: 10.2217/mmt-2017-0003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/04/2017] [Indexed: 01/04/2023] Open
Abstract
The treatment of malignant melanoma has changed beyond recognition in the last 7 years. Where previously single agent dacarbazine was often the only treatment used for advanced disease, now there are potentially multiple lines of treatment, based on immunotherapy and targeted treatment options, either as monotherapy or in combination. In this brave new world the question arises, does chemotherapy still have any relevance in the modern management of melanoma? In this review, we summarize the various chemotherapeutic options that have been trialled in melanoma to date, and discuss the role chemotherapy may still play in treating melanoma, potentially in combination with more novel agents, or in certain subtypes of melanoma.
Collapse
Affiliation(s)
- Avinash Gupta
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Fabio Gomes
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Paul Lorigan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
- Division of Molecular & Clinical Cancer Sciences, University of Manchester, Manchester, M13 9PL, UK
| |
Collapse
|
9
|
Haass NK, Gabrielli B. Cell cycle-tailored targeting of metastatic melanoma: Challenges and opportunities. Exp Dermatol 2017; 26:649-655. [PMID: 28109167 DOI: 10.1111/exd.13303] [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: 01/18/2017] [Indexed: 12/21/2022]
Abstract
The advent of targeted therapies of metastatic melanoma, such as MAPK pathway inhibitors and immune checkpoint antagonists, has turned dermato-oncology from the "bad guy" to the "poster child" in oncology. Current targeted therapies are effective, although here is a clear need to develop combination therapies to delay the onset of resistance. Many antimelanoma drugs impact on the cell cycle but are also dependent on certain cell cycle phases resulting in cell cycle phase-specific drug insensitivity. Here, we raise the question: Have combination trials been abandoned prematurely as ineffective possibly only because drug scheduling was not optimized? Firstly, if both drugs of a combination hit targets in the same melanoma cell, cell cycle-mediated drug insensitivity should be taken into account when planning combination therapies, timing of dosing schedules and choice of drug therapies in solid tumors. Secondly, if the combination is designed to target different tumor cell subpopulations of a heterogeneous tumor, one drug effective in a particular subpopulation should not negatively impact on the other drug targeting another subpopulation. In addition to the role of cell cycle stage and progression on standard chemotherapeutics and targeted drugs, we discuss the utilization of cell cycle checkpoint control defects to enhance chemotherapeutic responses or as targets themselves. We propose that cell cycle-tailored targeting of metastatic melanoma could further improve therapy outcomes and that our real-time cell cycle imaging 3D melanoma spheroid model could be utilized as a tool to measure and design drug scheduling approaches.
Collapse
Affiliation(s)
- Nikolas K Haass
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia.,The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| | - Brian Gabrielli
- Mater Medical Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
10
|
Computed determination of the in vitro optimal chemocombinations of sphaeropsidin A with chemotherapeutic agents to combat melanomas. Cancer Chemother Pharmacol 2017; 79:971-983. [DOI: 10.1007/s00280-017-3293-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/24/2017] [Indexed: 12/19/2022]
|
11
|
Redmer T, Walz I, Klinger B, Khouja S, Welte Y, Schäfer R, Regenbrecht C. The role of the cancer stem cell marker CD271 in DNA damage response and drug resistance of melanoma cells. Oncogenesis 2017; 6:e291. [PMID: 28112719 PMCID: PMC5294251 DOI: 10.1038/oncsis.2016.88] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/31/2016] [Accepted: 11/25/2016] [Indexed: 12/14/2022] Open
Abstract
Several lines of evidence have suggested that stemness and acquired resistance to targeted inhibitors or chemotherapeutics are mechanistically linked. Here we observed high cell surface and total levels of nerve growth factor receptor/CD271, a marker of melanoma-initiating cells, in sub-populations of chemoresistant cell lines. CD271 expression was increased in drug-sensitive cells but not resistant cells in response to DNA-damaging chemotherapeutics etoposide, fotemustine and cisplatin. Comparative analysis of melanoma cells engineered to stably express CD271 or a targeting short hairpin RNA by expression profiling provided numerous genes regulated in a CD271-dependent manner. In-depth analysis of CD271-responsive genes uncovered the association of CD271 with regulation of DNA repair components. In addition, gene set enrichment analysis revealed enrichment of CD271-responsive genes in drug-resistant cells, among them DNA repair components. Moreover, our comparative screen identified the fibroblast growth factor 13 (FGF13) as a target of CD271, highly expressed in chemoresistant cells. Further we show that levels of CD271 determine drug response. Knock-down of CD271 in fotemustine-resistant cells decreased expression of FGF13 and at least partly restored sensitivity to fotemustine. Together, we demonstrate that expression of CD271 is responsible for genes associated with DNA repair and drug response. Further, we identified 110 CD271-responsive genes predominantly expressed in melanoma metastases, among them were NEK2, TOP2A and RAD51AP1 as potential drivers of melanoma metastasis. In addition, we provide mechanistic insight in the regulation of CD271 in response to drugs. We found that CD271 is potentially regulated by p53 and in turn is needed for a proper p53-dependent response to DNA-damaging drugs. In summary, we provide for the first time insight in a CD271-associated signaling network connecting CD271 with DNA repair, drug response and metastasis.
Collapse
Affiliation(s)
- T Redmer
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - I Walz
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - B Klinger
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - S Khouja
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Y Welte
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - R Schäfer
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - C Regenbrecht
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,CPO-Cellular Phenomics and Oncology Berlin-Buch GmbH, Berlin, Germany
| |
Collapse
|
12
|
Mitochondrial ATF2 translocation contributes to apoptosis induction and BRAF inhibitor resistance in melanoma through the interaction of Bim with VDAC1. Oncotarget 2016; 6:36338-53. [PMID: 26462148 PMCID: PMC4742181 DOI: 10.18632/oncotarget.5537] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/29/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The mitochondrial accumulation of ATF2 is involved in tumor suppressor activities via cytochrome c release in melanoma cells. However, the signaling pathways that connect mitochondrial ATF2 accumulation and cytochrome c release are not well documented. METHODS Several melanoma cell lines, B16F10, K1735M2, A375 and A375-R1, were treated with paclitaxel and vemurafenib to test the function of mitochondrial ATF2 and its connection to Bim and voltage-dependent anion channel 1 (VDAC1). Immunoprecipitation analysis was performed to investigate the functional interaction between the involved proteins. VDAC1 oligomerization was evaluated using an EGS-based crosslinking assay. RESULTS The expression and migration of ATF2 to the mitochondria accounted for paclitaxel stimuli and acquired resistance to BRAF inhibitors. Mitochondrial ATF2 facilitated Bim stabilization through the inhibition of its degradation by the proteasome, thereby promoting cytochrome c release and inducing apoptosis in B16F10 and A375 cells. Studies using B16F10 and A375 cells genetically modified for ATF2 indicated that mitochondrial ATF2 was able to dissociate Bim from the Mcl-1/Bim complex to trigger VDAC1 oligomerization. Immunoprecipitation analysis revealed that Bim interacts with VDAC1, and this interaction was remarkably enhanced during apoptosis. CONCLUSION These results reveal that mitochondrial ATF2 is associated with the induction of apoptosis and BRAF inhibitor resistance through Bim activation, which might suggest potential novel therapies for the targeted induction of apoptosis in melanoma therapy.
Collapse
|
13
|
Combined vemurafenib and fotemustine in patients with BRAF V600 melanoma progressing on vemurafenib. Oncotarget 2016; 9:12408-12417. [PMID: 29552321 PMCID: PMC5844757 DOI: 10.18632/oncotarget.10589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background BRAF inhibitor vemurafenib achieves high response rate and an improvement in survival in patients with BRAF-mutated metastatic melanoma. However, median progression-free survival is only 6.9 months in the phase 3 study. Retrospective analyses suggest that treatment with BRAF inhibitors beyond initial progression might be associated with improved overall survival. We aimed to prospectively investigate the activity of prolonged treatment with vemurafenib and the addition of fotemustine in patients with systemic progression on prior single-agent BRAF inhibitor. Patients and Methods In this two-centres, single-arm Phase 2 trial, we enrolled patients with systemic progressive disease during single-agent vemurafenib treatment. Participants received vemurafenib 960 mg twice daily or dose administered at time of disease progression with vemurafenib previous treatment and fotemustine 100 mg/m2 intravenously every three weeks. The primary endpoint was PFS. Results Thirty-one patients were enrolled in the study; 16 patients had brain metastases at baseline. Median PFS was 3.9 months and 19 patients (61.3%) achieved disease control (1 CR, 4 PR, 14 SD). For patients achieving disease control, median duration of treatment was 6 months. Median OS was 5.8 months from enrolment and 15.4 months from start of previous vemurafenib. Five patients (16.1%) had a G3-4 AE, the most common being thrombocytopenia, which occurred in 3 patients. This trial is registered with ClinicalTrials.gov number NCT01983124. Conclusion The combination of vemurafenib plus fotemustine has clinical activity and an acceptable safety profile in BRAF-refractory patients.
Collapse
|
14
|
Jeong JH, Hong YS, Park Y, Kim J, Kim JE, Kim KP, Kim SY, Park JH, Kim JH, Park IJ, Lim SB, Yu CS, Kim JC, Kim TW. Phase 1 Study of Preoperative Chemoradiation Therapy With Temozolomide and Capecitabine in Patients With Locally Advanced Rectal Cancer. Int J Radiat Oncol Biol Phys 2016; 96:289-295. [PMID: 27473815 DOI: 10.1016/j.ijrobp.2016.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/05/2016] [Accepted: 05/10/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE Preoperative chemoradiation therapy (CRT) with capecitabine is a standard treatment strategy in patients with locally advanced rectal cancer (LARC). Temozolomide improves the survival of patients with glioblastoma with hypermethylated O(6)-methylguanine DNA methyltransferase (MGMT); MGMT hypermethylation is one of the colorectal carcinogenesis pathways. We aimed to determine the dose-limiting toxicity (DLT) and recommended dose (RD) of temolozomide in combination with capecitabine-based preoperative CRT for LARC. METHODS AND MATERIALS Radiation therapy was delivered with 45 Gy/25 daily fractions with coned-down boost of 5.4 Gy/3 fractions. Concurrent chemotherapy comprised fixed and escalated doses of capecitabine and temozolomide, respectively. The MGMT hypermethylation was evaluated in pretreatment tumor samples. This trial is registered with ClinicalTrials.gov with the number NCT01781403. RESULTS Twenty-two patients with LARC of cT3-4N0 or cTanyN1-2 were accrued. Dose level 3 was chosen as the RD because DLT was noticeably absent in 10 patients treated up to dose level 3. An additional 12 patients were recruited in this group. Grade III adverse events were noted, and pathologic complete response (pCR) was observed in 7 patients (31.8%); MGMT hypermethylation was detected in 16. The pCR rate was 37.5% and 16.7% in the hypermethylated and unmethylated MGMT groups, respectively (P=.616). CONCLUSIONS There was a tendency toward higher pCR rates in patients with hypermethylated MGMT. Future randomized studies are therefore warranted.
Collapse
Affiliation(s)
- Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong Sang Hong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yangsoon Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jihun Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Eun Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sun Young Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Hong Park
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Hoon Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In Ja Park
- Department of Colorectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seok-Byung Lim
- Department of Colorectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chang Sik Yu
- Department of Colorectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin Cheon Kim
- Department of Colorectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Krumm A, Barckhausen C, Kücük P, Tomaszowski KH, Loquai C, Fahrer J, Krämer OH, Kaina B, Roos WP. Enhanced Histone Deacetylase Activity in Malignant Melanoma Provokes RAD51 and FANCD2-Triggered Drug Resistance. Cancer Res 2016; 76:3067-77. [PMID: 26980768 DOI: 10.1158/0008-5472.can-15-2680] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/29/2016] [Indexed: 11/16/2022]
Abstract
DNA-damaging anticancer drugs remain a part of metastatic melanoma therapy. Epigenetic reprogramming caused by increased histone deacetylase (HDAC) activity arising during tumor formation may contribute to resistance of melanomas to the alkylating drugs temozolomide, dacarbazine, and fotemustine. Here, we report on the impact of class I HDACs on the response of malignant melanoma cells treated with alkylating agents. The data show that malignant melanomas in situ contain a high level of HDAC1/2 and malignant melanoma cells overexpress HDAC1/2/3 compared with noncancer cells. Furthermore, pharmacologic inhibition of class I HDACs sensitizes malignant melanoma cells to apoptosis following exposure to alkylating agents, while not affecting primary melanocytes. Inhibition of HDAC1/2/3 caused sensitization of melanoma cells to temozolomide in vitro and in melanoma xenografts in vivo HDAC1/2/3 inhibition resulted in suppression of DNA double-strand break (DSB) repair by homologous recombination because of downregulation of RAD51 and FANCD2. This sensitized cells to the cytotoxic DNA lesion O(6)-methylguanine and caused a synthetic lethal interaction with the PARP-1 inhibitor olaparib. Furthermore, knockdown experiments identified HDAC2 as being responsible for the regulation of RAD51. The influence of class I HDACs on DSB repair by homologous recombination and the possible clinical implication on malignant melanoma therapy with temozolomide and other alkylating drugs suggests a combination approach where class I HDAC inhibitors such as valproic acid or MS-275 (entinostat) appear to counteract HDAC- and RAD51/FANCD2-mediated melanoma cell resistance. Cancer Res; 76(10); 3067-77. ©2016 AACR.
Collapse
Affiliation(s)
- Andrea Krumm
- Institute of Toxicology, Medical Center of the University Mainz, Mainz, Germany
| | | | - Pelin Kücük
- Institute of Toxicology, Medical Center of the University Mainz, Mainz, Germany
| | | | - Carmen Loquai
- Department of Dermatology, Medical Center of the University Mainz, Mainz, Germany
| | - Jörg Fahrer
- Institute of Toxicology, Medical Center of the University Mainz, Mainz, Germany
| | | | - Bernd Kaina
- Institute of Toxicology, Medical Center of the University Mainz, Mainz, Germany
| | - Wynand Paul Roos
- Institute of Toxicology, Medical Center of the University Mainz, Mainz, Germany.
| |
Collapse
|
16
|
Delmas A, Cherier J, Pohorecka M, Medale-Giamarchi C, Meyer N, Casanova A, Sordet O, Lamant L, Savina A, Pradines A, Favre G. The c-Jun/RHOB/AKT pathway confers resistance of BRAF-mutant melanoma cells to MAPK inhibitors. Oncotarget 2016; 6:15250-64. [PMID: 26098773 PMCID: PMC4558149 DOI: 10.18632/oncotarget.3888] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/25/2015] [Indexed: 11/25/2022] Open
Abstract
The response of BRAF-mutant melanoma patients to BRAF inhibitors is dramatically impaired by secondary resistances and rapid relapse. So far, the molecular mechanisms driving these resistances are not completely understood. Here, we show that, in BRAF-mutant melanoma cells, inhibition of BRAF or its target MEK induces RHOB expression by a mechanism that depends on the transcription factor c-Jun. In those cells, RHOB deficiency causes hypersensitivity to BRAF and MEK inhibitors-induced apoptosis. Supporting these results, loss of RHOB expression in metastatic melanoma tissues is associated with an increased progression-free survival of BRAF-mutant patients treated with vemurafenib. Following BRAF inhibition, RHOB activates AKT whose inhibition causes hypersensitivity of BRAF-mutant melanoma cells to BRAF inhibitors. In mice, AKT inhibition synergizes with vemurafenib to block tumor growth of BRAF-mutant metastatic melanoma. Our findings reveal that BRAF inhibition activates a c-Jun/RHOB/AKT pathway that promotes tumor cell survival and further support a role of this pathway in the resistance of melanoma to vemurafenib. Our data also highlight the importance of using RHOB tumor levels as a biomarker to predict vemurafenib patient's response and to select those that would benefit of the combination with AKT inhibitors.
Collapse
Affiliation(s)
- Audrey Delmas
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Julia Cherier
- Inserm, UMR 1037-CRCT, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Magdalena Pohorecka
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Claire Medale-Giamarchi
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Nicolas Meyer
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Department of Dermatology, Toulouse, France
| | - Anne Casanova
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Olivier Sordet
- Inserm, UMR 1037-CRCT, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Laurence Lamant
- Inserm, UMR 1037-CRCT, Toulouse, France.,Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Department of Pathology, Toulouse, France
| | - Ariel Savina
- Scientific Partnerships, Roche SAS, Boulogne Billancourt, France
| | - Anne Pradines
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Gilles Favre
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| |
Collapse
|
17
|
Beaumont KA, Hill DS, Daignault SM, Lui GYL, Sharp DM, Gabrielli B, Weninger W, Haass NK. Cell Cycle Phase-Specific Drug Resistance as an Escape Mechanism of Melanoma Cells. J Invest Dermatol 2016; 136:1479-1489. [PMID: 26970356 DOI: 10.1016/j.jid.2016.02.805] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/06/2016] [Accepted: 02/25/2016] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment is characterized by cancer cell subpopulations with heterogeneous cell cycle profiles. For example, hypoxic tumor zones contain clusters of cancer cells that arrest in G1 phase. It is conceivable that neoplastic cells exhibit differential drug sensitivity based on their residence in specific cell cycle phases. In this study, we used two-dimensional and organotypic melanoma culture models in combination with fluorescent cell cycle indicators to investigate the effects of cell cycle phases on clinically used drugs. We demonstrate that G1-arrested melanoma cells, irrespective of the underlying cause mediating G1 arrest, are resistant to apoptosis induced by the proteasome inhibitor bortezomib or the alkylating agent temozolomide. In contrast, G1-arrested cells were more sensitive to mitogen-activated protein kinase pathway inhibitor-induced cell death. Of clinical relevance, pretreatment of melanoma cells with a mitogen-activated protein kinase pathway inhibitor, which induced G1 arrest, resulted in resistance to temozolomide or bortezomib. On the other hand, pretreatment with temozolomide, which induced G2 arrest, did not result in resistance to mitogen-activated protein kinase pathway inhibitors. In summary, we established a model to study the effects of the cell cycle on drug sensitivity. Cell cycle phase-specific drug resistance is an escape mechanism of melanoma cells that has implications on the choice and timing of drug combination therapies.
Collapse
Affiliation(s)
- Kimberley A Beaumont
- The Centenary Institute, Newtown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - David S Hill
- The Centenary Institute, Newtown, NSW, Australia; Dermatological Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Sheena M Daignault
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Goldie Y L Lui
- The Centenary Institute, Newtown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Danae M Sharp
- The Centenary Institute, Newtown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Brian Gabrielli
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Wolfgang Weninger
- The Centenary Institute, Newtown, NSW, Australia; Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Nikolas K Haass
- The Centenary Institute, Newtown, NSW, Australia; The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia; Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|