51
|
Leonetti A, Facchinetti F, Rossi G, Minari R, Conti A, Friboulet L, Tiseo M, Planchard D. BRAF in non-small cell lung cancer (NSCLC): Pickaxing another brick in the wall. Cancer Treat Rev 2018; 66:82-94. [PMID: 29729495 DOI: 10.1016/j.ctrv.2018.04.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/28/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
Molecular characterization of non-small cell lung cancer (NSCLC) marked an historical turning point for the treatment of lung tumors harboring kinase alterations suitable for specific targeted drugs inhibition, translating into major clinical improvements. Besides EGFR, ALK and ROS1, BRAF represents a novel therapeutic target for the treatment of advanced NSCLC. BRAF mutations, found in 1.5-3.5% of NSCLC, are responsible of the constitutive activation of mitogen activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway. Clinical trials evaluating the efficacy of the BRAF inhibitor dabrafenib in combination with the downstream MEK inhibitor trametinib in metastatic BRAFV600E-mutated NSCLC guaranteed FDA and EMA rapid approval of the combination regimen in this clinical setting. In line with the striking results observed in metastatic melanoma harboring the same molecular alteration, BRAF and MEK inhibition should be considered a new standard of care in this molecular subtype of NSCLC. In the present review, we propose an overview of the available evidence about BRAF in NSCLC mutations (V600E and non-V600E), from biological significance to emerging clinical implications of BRAF mutations detection. Focusing on the current strategies to act against the mutated kinase, we moreover approach additional strategies to overcome treatment resistance.
Collapse
Affiliation(s)
| | | | - Giulio Rossi
- Pathology Unit, Santa Maria delle Croci Hospital, Ravenna, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Luc Friboulet
- INSERM, U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - David Planchard
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
52
|
Raaijmakers MIG, Widmer DS, Narechania A, Eichhoff O, Freiberger SN, Wenzina J, Cheng PF, Mihic-Probst D, Desalle R, Dummer R, Levesque MP. Co-existence of BRAF and NRAS driver mutations in the same melanoma cells results in heterogeneity of targeted therapy resistance. Oncotarget 2018; 7:77163-77174. [PMID: 27791198 PMCID: PMC5363577 DOI: 10.18632/oncotarget.12848] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/13/2016] [Indexed: 12/30/2022] Open
Abstract
Acquired chemotherapeutic resistance of cancer cells can result from a Darwinistic evolution process in which heterogeneity plays an important role. In order to understand the impact of genetic heterogeneity on acquired resistance and second line therapy selection in metastatic melanoma, we sequenced the exomes of 27 lesions which were collected from 3 metastatic melanoma patients treated with targeted or non-targeted inhibitors. Furthermore, we tested the impact of a second NRAS mutation in 7 BRAF inhibitor resistant early passage cell cultures on the selection of second line therapies.We observed a rapid monophyletic evolution of melanoma subpopulations in response to targeted therapy that was not observed in non-targeted therapy. We observed the acquisition of NRAS mutations in the BRAF mutated patient treated with a BRAF inhibitor in 1 of 5 of his post-resistant samples. In an additional cohort of 5 BRAF-inhibitor treated patients we detected 7 NRAS mutations in 18 post-resistant samples. No NRAS mutations were detected in pre-resistant samples. By sequencing 65 single cell clones we prove that NRAS mutations co-occur with BRAF mutations in single cells. The double mutated cells revealed a heterogeneous response to MEK, ERK, PI3K, AKT and multi RTK - inhibitors.We conclude that BRAF and NRAS co-mutations are not mutually exclusive. However, the sole finding of double mutated cells in a resistant tumor is not sufficient to determine follow-up therapy. In order to target the large pool of heterogeneous cells in a patient, we think combinational therapy targeting different pathways will be necessary.
Collapse
Affiliation(s)
| | - Daniel S Widmer
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland
| | | | - Ossia Eichhoff
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland
| | - Sandra N Freiberger
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland.,Department of Dermatology, Skin and Endothelium Research Division, Medical University of Vienna, Austria
| | - Judith Wenzina
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland.,Department of Dermatology, Skin and Endothelium Research Division, Medical University of Vienna, Austria
| | - Phil F Cheng
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland
| | - Daniela Mihic-Probst
- Department of Pathology, University of Zurich, University Hospital Zürich, Switzerland
| | - Rob Desalle
- American Museum of Natural History, New York, New York, USA
| | - Reinhard Dummer
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University of Zurich, University Hospital Zürich, Switzerland
| |
Collapse
|
53
|
Moschos SJ, Sullivan RJ, Hwu WJ, Ramanathan RK, Adjei AA, Fong PC, Shapira-Frommer R, Tawbi HA, Rubino J, Rush TS, Zhang D, Miselis NR, Samatar AA, Chun P, Rubin EH, Schiller J, Long BJ, Dayananth P, Carr D, Kirschmeier P, Bishop WR, Deng Y, Cooper A, Shipps GW, Moreno BH, Robert L, Ribas A, Flaherty KT. Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors. JCI Insight 2018; 3:92352. [PMID: 29467321 DOI: 10.1172/jci.insight.92352] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 12/28/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Constitutive activation of ERK1/2 occurs in various cancers, and its reactivation is a well-described resistance mechanism to MAPK inhibitors. ERK inhibitors may overcome the limitations of MAPK inhibitor blockade. The dual mechanism inhibitor SCH772984 has shown promising preclinical activity across various BRAFV600/RAS-mutant cancer cell lines and human cancer xenografts. METHODS We have developed an orally bioavailable ERK inhibitor, MK-8353; conducted preclinical studies to demonstrate activity, pharmacodynamic endpoints, dosing, and schedule; completed a study in healthy volunteers (P07652); and subsequently performed a phase I clinical trial in patients with advanced solid tumors (MK-8353-001). In the P07652 study, MK-8353 was administered as a single dose in 10- to 400-mg dose cohorts, whereas in the MK-8353-001 study, MK-8353 was administered in 100- to 800-mg dose cohorts orally twice daily. Safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity were analyzed. RESULTS MK-8353 exhibited comparable potency with SCH772984 across various preclinical cancer models. Forty-eight patients were enrolled in the P07652 study, and twenty-six patients were enrolled in the MK-8353-001 study. Adverse events included diarrhea (44%), fatigue (40%), nausea (32%), and rash (28%). Dose-limiting toxicity was observed in the 400-mg and 800-mg dose cohorts. Sufficient exposure to MK-8353 was noted that correlated with biological activity in preclinical data. Three of fifteen patients evaluable for treatment response in the MK-8353-001 study had partial response, all with BRAFV600-mutant melanomas. CONCLUSION MK-8353 was well tolerated up to 400 mg twice daily and exhibited antitumor activity in patients with BRAFV600-mutant melanoma. However, antitumor activity was not particularly correlated with pharmacodynamic parameters. TRIAL REGISTRATION ClinicalTrials.gov NCT01358331. FUNDING Merck Sharp & Dohme Corp., a subsidiary of Merck & Co. Inc., and NIH (P01 CA168585 and R35 CA197633).
Collapse
Affiliation(s)
- Stergios J Moschos
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ramesh K Ramanathan
- Translational Genomics Research Institute, Phoenix, Arizona, USA; Virginia G. Piper Cancer Center, Scottsdale, Arizona, USA
| | - Alex A Adjei
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Peter C Fong
- The University of Auckland and Auckland City Hospital, Auckland, New Zealand
| | | | - Hussein A Tawbi
- University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | | | | | - Da Zhang
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | | | | | | | | | | | | | | | - Donna Carr
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | | | | | - Yongqi Deng
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Alan Cooper
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | | | - Blanca Homet Moreno
- Jonsson Comprehensive Cancer Center at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Lidia Robert
- Jonsson Comprehensive Cancer Center at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
54
|
Tolcher AW, Peng W, Calvo E. Rational Approaches for Combination Therapy Strategies Targeting the MAP Kinase Pathway in Solid Tumors. Mol Cancer Ther 2018; 17:3-16. [DOI: 10.1158/1535-7163.mct-17-0349] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/03/2017] [Accepted: 10/13/2017] [Indexed: 11/16/2022]
|
55
|
Fisher ML, Grun D, Adhikary G, Xu W, Eckert RL. Inhibition of YAP function overcomes BRAF inhibitor resistance in melanoma cancer stem cells. Oncotarget 2017; 8:110257-110272. [PMID: 29299145 PMCID: PMC5746380 DOI: 10.18632/oncotarget.22628] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/25/2017] [Indexed: 12/11/2022] Open
Abstract
Treating BRAF inhibitor-resistant melanoma is an important therapeutic goal. Thus, it is important to identify and target mechanisms of resistance to improve therapy. The YAP1 and TAZ proteins of the Hippo signaling pathway are important drivers of cancer cell survival, and are BRAF inhibitor resistant factors in melanoma. We examine the role of YAP1/TAZ in melanoma cancer stem cells (MCS cells). We demonstrate that YAP1, TAZ and TEAD (TEA domain transcription factor) levels are elevated in BRAF inhibitor resistant MCS cells and enhance cell survival, spheroid formation, matrigel invasion and tumor formation. Moreover, increased YAP1, TAZ and TEAD are associated with sustained ERK1/2 activity that is not suppressed by BRAF inhibitor. Xenograft studies show that treating BRAF inhibitor-resistant tumors with verteporfin, an agent that interferes with YAP1 function, reduces YAP1/TAZ level, restores BRAF inhibitor suppression of ERK1/2 signaling and reduces tumor growth. Verteporfin is highly effective as concentrations of verteporfin that do not impact tumor formation restore BRAF inhibitor suppression of tumor formation, suggesting that co-treatment with agents that inhibit YAP1 and BRAF(V600E) may be a viable therapy for cancer stem cell-derived BRAF inhibitor-resistant melanoma.
Collapse
Affiliation(s)
- Matthew L. Fisher
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
| | - Daniel Grun
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
| | - Gautam Adhikary
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
| | - Wen Xu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
| | - Richard L. Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
- Department of Reproductive Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
- The Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
| |
Collapse
|
56
|
Echevarría-Vargas IM, Villanueva J. COMBATING NRAS MUTANT MELANOMA: FROM BENCH TO BEDSIDE. Melanoma Manag 2017; 4:183-186. [PMID: 29785260 DOI: 10.2217/mmt-2017-0023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
| | - Jessie Villanueva
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA.,Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| |
Collapse
|
57
|
Xue Y, Martelotto L, Baslan T, Vides A, Solomon M, Mai TT, Chaudhary N, Riely GJ, Li BT, Scott K, Cechhi F, Stierner U, Chadalavada K, de Stanchina E, Schwartz S, Hembrough T, Nanjangud G, Berger MF, Nilsson J, Lowe SW, Reis-Filho JS, Rosen N, Lito P. An approach to suppress the evolution of resistance in BRAF V600E-mutant cancer. Nat Med 2017; 23:929-937. [PMID: 28714990 PMCID: PMC5696266 DOI: 10.1038/nm.4369] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
Abstract
The principles governing evolution of tumors exposed to targeted therapy are poorly understood. Here we modeled the selection and propagation of BRAF amplification (BRAFamp) in patient-derived tumor xenografts (PDX) treated with a direct ERK inhibitor, alone or in combination with other pathway inhibitors. Single cell sequencing and multiplex-fluorescence in situ hybridization mapped the emergence of extra-chromosomal amplification in parallel evolutionary tracts, arising in the same tumor shortly after treatment. The evolutionary selection of BRAFamp is determined by the fitness threshold, the barrier subclonal populations need to overcome to regain fitness in the presence of therapy. This differed for ERK signaling inhibitors, suggesting that sequential monotherapy is ineffective and selects for a progressively higher BRAF copy number. Concurrent targeting of RAF, MEK and ERK, however, imposes a sufficiently high fitness threshold to prevent the propagation of subclones with high-level amplification. Administered on an intermittent schedule, this treatment inhibited tumor growth in 11/11-lung cancer and melanoma PDX without apparent toxicity in mice. Thus, gene amplification can be acquired and expanded through parallel evolution, enabling tumors to adapt while maintaining their intratumoral heterogeneity. Treatments that impose the highest fitness threshold will likely prevent the evolution of resistance-causing alterations and merit testing in patients.
Collapse
Affiliation(s)
- Yaohua Xue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA.,Weill Cornell-Rockefeller-Sloan Kettering Tri-institutional MD-PhD Program, New York, New York, USA
| | - Luciano Martelotto
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alberto Vides
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Martha Solomon
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Trang Thi Mai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Neelam Chaudhary
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Greg J Riely
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Bob T Li
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Ulrika Stierner
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Kalyani Chadalavada
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Gouri Nanjangud
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Jonas Nilsson
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Piro Lito
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell Medical College, Cornell University, New York, New York, USA
| |
Collapse
|
58
|
Liu P, Zhang Z, Wang Q, Guo R, Mei W. Lithium Chloride Facilitates Autophagy Following Spinal Cord Injury via ERK-dependent Pathway. Neurotox Res 2017; 32:535-543. [PMID: 28593525 DOI: 10.1007/s12640-017-9758-1] [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: 02/06/2017] [Revised: 05/18/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is one major cause of death and results in long-term disability even in the most productive periods of human lives with few efficacious drugs. Autophagy is a potential therapeutic target for SCI. In the present study, we examined the role of lithium in functional recovery in the rat model of SCI and explored the related mechanism. Locomotion tests were employed to assess the functional recovery after SCI, Western blotting and RT-PCT to determine the level of p-ERK and LC3-II as well as p62, immunofluorescence imaging to localize LC3 and p62. Here, we found that both the expression of LC3-II and p62 were increased after SCI. However, lithium chloride enhanced the level of LC3-II while abrogated the abundance of p62. Furthermore, lithium treatment facilitated ERK activation in vivo, and inhibition of MEK/ERK signaling pathway suppressed lithium-evoked autophagy flux. Taken together, our results illustrated that lithium facilitated functional recovery by enhancing autophagy flux.
Collapse
Affiliation(s)
- Peilin Liu
- Department of Spine Surgery, Zhengzhou Orthopaedic Hospital, Zhengzhou, China
| | - Zijuan Zhang
- Experimental Teaching Center, School of Basic Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qingde Wang
- Department of Spine Surgery, Zhengzhou Orthopaedic Hospital, Zhengzhou, China
| | - Rundong Guo
- Department of Spine Surgery, Zhengzhou Orthopaedic Hospital, Zhengzhou, China
| | - Wei Mei
- Department of Spine Surgery, Zhengzhou Orthopaedic Hospital, Zhengzhou, China.
| |
Collapse
|
59
|
Chatterjee S, Huang EHB, Christie I, Burns TF. Reactivation of the p90RSK-CDC25C Pathway Leads to Bypass of the Ganetespib-Induced G 2-M Arrest and Mediates Acquired Resistance to Ganetespib in KRAS-Mutant NSCLC. Mol Cancer Ther 2017; 16:1658-1668. [PMID: 28566436 DOI: 10.1158/1535-7163.mct-17-0114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/08/2017] [Accepted: 05/12/2017] [Indexed: 11/16/2022]
Abstract
A subset of non-small cell lung cancers (NSCLC) are dependent upon oncogenic driver mutations, including the most frequently observed driver mutant KRAS, which is associated with a poor prognosis. As direct RAS targeting in the clinic has been unsuccessful to date, use of Hsp90 inhibitors appeared to be a promising therapy for KRAS-mutant NSCLC; however, limited clinical efficacy was observed due to rapid resistance. Furthermore, the combination of the Hsp90 inhibitor (Hsp90i), ganetespib, and docetaxel was tested in a phase III clinical trial and failed to demonstrate benefit. Here, we investigated the mechanism(s) of resistance to ganetespib and explored why the combination with docetaxel failed in the clinic. We have not only identified a critical role for the bypass of the G2-M cell-cycle checkpoint as a mechanism of ganetespib resistance (GR) but have also found that GR leads to cross-resistance to docetaxel. Reactivation of p90RSK and its downstream target, CDC25C, was critical for GR and mediated the bypass of a G2-M arrest. Overexpression of either p90RSK or CDC25C lead to bypass of G2-M arrest and induced ganetespib resistance in vitro and in vivo Moreover, resistance was dependent on p90RSK/CDC25C signaling, as synthetic lethality to ERK1/2, p90RSK, or CDC25C inhibitors was observed. Importantly, the combination of ganetespib and p90RSK or CDC25C inhibitors was highly efficacious in parental cells. These studies provide a way forward for Hsp90 inhibitors through the development of novel rationally designed Hsp90 inhibitor combinations that may prevent or overcome resistance to Hsp90i. Mol Cancer Ther; 16(8); 1658-68. ©2017 AACR.
Collapse
Affiliation(s)
- Suman Chatterjee
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Eric H-B Huang
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Ian Christie
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Timothy F Burns
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
60
|
Deng M, Qin Y, Chen X, Li D, Wang Q, Zheng H, Gu L, Deng C, Xue Y, Zhu D, Wang Q, Wang J. Combination of celecoxib and PD184161 exerts synergistic inhibitory effects on gallbladder cancer cell proliferation. Oncol Lett 2017; 13:3850-3858. [PMID: 28521485 PMCID: PMC5431146 DOI: 10.3892/ol.2017.5914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/24/2016] [Indexed: 12/16/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) and extracellular signal-regulated kinase 1/2 (ERK1/2) may serve as potential targets in various types of cancer; however, the roles of these proteins in gallbladder carcinoma (GBC) have not been reported previously. In the present study, the expression levels of COX-2 and phospho (p)-ERK1/2 in GBC were examined and the biological activities of celecoxib and PD184161 (specific inhibitors of COX-2 and p-ERK1/2, respectively) on the proliferation, cell cycle and apoptosis of the GBC-SD and NOZ human GBC cell lines were evaluated by a series of in vitro and in vivo studies. COX-2 and p-ERK1/2 protein expression levels were found to be significantly elevated in GBC tissues as well as in GBC-SD and NOZ cells. Treatments with celecoxib and PD184161 significantly inhibited GBC-SD and NOZ cell growth in a concentration-dependent manner, and their combination produced a synergistic inhibitory effect. In addition, celecoxib and PD184161 significantly inhibited tumor growth in xenograft nude mice. Celecoxib treatment led to G1 arrest via the upregulation of p21 and p27 expression in GBC-SD and NOZ cells, whereas PD184161 did not affect cell cycle distribution. The combination of celecoxib and PD184161 was able to promote cell apoptosis by triggering a collapse of mitochondrial membrane potential and activating caspase-3-mediated apoptosis. In conclusion, COX-2 and p-ERK1/2 protein may serve as potential targets for GBC chemotherapy, and the combination of celecoxib and PD184161 could significantly inhibit GBC cell growth, induce cell G1 arrest and trigger cell apoptosis of GBC cells.
Collapse
Affiliation(s)
- Min Deng
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Yiyu Qin
- Clinical Medical College, Research Centre of Biomedical Technology, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224005, P.R. China
| | - Xiaodong Chen
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Dapeng Li
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Qiangwu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Hailun Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Lin Gu
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Chaojing Deng
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Yongju Xue
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Danyu Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
- Correspondence to: Dr Qizhi Wang or Dr Jianchao Wang, Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui 233004, P.R. China, E-mail: , E-mail:
| | - Jianchao Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
- Correspondence to: Dr Qizhi Wang or Dr Jianchao Wang, Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui 233004, P.R. China, E-mail: , E-mail:
| |
Collapse
|
61
|
Ahronian LG, Corcoran RB. Strategies for monitoring and combating resistance to combination kinase inhibitors for cancer therapy. Genome Med 2017; 9:37. [PMID: 28431544 PMCID: PMC5399860 DOI: 10.1186/s13073-017-0431-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Targeted therapies such as kinase inhibitors and monoclonal antibodies have dramatically altered cancer care in recent decades. Although these targeted therapies have improved patient outcomes in several cancer types, resistance ultimately develops to these agents. One potential strategy proposed to overcome acquired resistance involves taking repeat tumor biopsies at the time of disease progression, to identify the specific molecular mechanism driving resistance in an individual patient and to select a new agent or combination of agents capable of surmounting that specific resistance mechanism. However, recent studies sampling multiple metastatic lesions upon acquired resistance, or employing “liquid biopsy” analyses of circulating tumor DNA, have revealed that multiple, heterogeneous resistance mechanisms can emerge in distinct tumor subclones in the same patient. This heterogeneity represents a major clinical challenge for devising therapeutic strategies to overcome resistance. In many cancers, multiple drug resistance mechanisms often converge to reactivate the original pathway targeted by the drug. This convergent evolution creates an opportunity to target a common signaling node to overcome resistance. Furthermore, integration of liquid biopsy approaches into clinical practice may allow real-time monitoring of emerging resistance alterations, allowing intervention prior to standard detection of radiographic progression. In this review, we discuss recent advances in understanding tumor heterogeneity and resistance to targeted therapies, focusing on combination kinase inhibitors, and we discuss approaches to address these issues in the clinic.
Collapse
Affiliation(s)
- Leanne G Ahronian
- Massachusetts General Hospital Cancer Center, Boston, MA, 02129, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ryan B Corcoran
- Massachusetts General Hospital Cancer Center, Boston, MA, 02129, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
62
|
Zhao W, Yan J, Gao L, Zhao J, Zhao C, Gao C, Luo X, Zhu X. Cdk5 is required for the neuroprotective effect of transforming growth factor-β1 against cerebral ischemia-reperfusion. Biochem Biophys Res Commun 2017; 485:775-781. [DOI: 10.1016/j.bbrc.2017.02.130] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/26/2017] [Indexed: 12/01/2022]
|
63
|
Liu S, Gao G, Yan D, Chen X, Yao X, Guo S, Li G, Zhao Y. Effects of miR-145-5p through NRAS on the cell proliferation, apoptosis, migration, and invasion in melanoma by inhibiting MAPK and PI3K/AKT pathways. Cancer Med 2017; 6:819-833. [PMID: 28332309 PMCID: PMC5387172 DOI: 10.1002/cam4.1030] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/09/2017] [Accepted: 01/16/2017] [Indexed: 12/21/2022] Open
Abstract
We aimed to detect the effects of miR-145-5p on the cell proliferation, apoptosis, migration, and invasion in NRAS-mutant, BRAF-mutant, and wild-type melanoma cells, in order to figure out the potential mechanisms and provide a novel therapeutic target of melanoma. RT-qPCR and western blot were used to detect the expression of miR-145-5p and NRAS in melanoma tumor tissues and cells, respectively. Luciferase assay was performed to determine whether miR-145-5p directly targeted NRAS. After transfecting miR-145-5p mimics, miR-145-5p inhibitors, NRAS cDNA and NRAS siRNA into CHL-1, VMM917 and SK-mel-28 cells, functional assays were used to detect the proliferation, apoptosis, invasion and migration, including MTT, flow cytometry, Transwell and wound healing assays. In addition, xenograft models in nude mice were also conducted to verify the role of miR-145-5p in vivo. MiR-145-5p was able to suppress proliferation, invasion, and migration of VMM917 and CHL-1 cells and induce apoptosis by inhibiting MAPK and PI3K/AKT pathways. However, aberrant expression of miR-145-5p and NRAS has little impact on the viability and metastasis of BRAF-mutant melanoma. The higher expression of miR-145-5p in xenograft models repressed the VMM917-induced and CHL-1-induced tumor growth observably and has little effect on SK-mel-28-induced tumor growth which was consistent with the results in vitro. Through targeting NRAS, miR-145-5p could suppress cell proliferation, invasion, and migration and induce apoptosis of CHL-1 and VMM917 melanoma cells by inhibiting MAPK and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Sha Liu
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.,Department of Burn and Plastic Surgery, The 253rd Hospital of PLA, Hohhot, Inner Mongolia, 010051, China
| | - Guozhen Gao
- Department of Burn and Plastic Surgery, The 253rd Hospital of PLA, Hohhot, Inner Mongolia, 010051, China
| | - Dexiong Yan
- Department of Burn and Plastic Surgery, The 253rd Hospital of PLA, Hohhot, Inner Mongolia, 010051, China
| | - Xiangjun Chen
- Department of Burn and Plastic Surgery, The 253rd Hospital of PLA, Hohhot, Inner Mongolia, 010051, China
| | - Xingwei Yao
- Department of Burn and Plastic Surgery, The 253rd Hospital of PLA, Hohhot, Inner Mongolia, 010051, China
| | - Shuzhong Guo
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guirong Li
- Department of Otolaryngology-Head and Neck Surgery, The Fourth People's Hospital of Shaanxi Province, Xi'an, 710043, China
| | - Yu Zhao
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
64
|
Chatterjee S, Huang EHB, Christie I, Kurland BF, Burns TF. Acquired Resistance to the Hsp90 Inhibitor, Ganetespib, in KRAS-Mutant NSCLC Is Mediated via Reactivation of the ERK-p90RSK-mTOR Signaling Network. Mol Cancer Ther 2017; 16:793-804. [PMID: 28167505 DOI: 10.1158/1535-7163.mct-16-0677] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/01/2023]
Abstract
Approximately 25% of non-small cell lung cancer (NSCLC) patients have KRAS mutations, and no effective therapeutic strategy exists for these patients. The use of Hsp90 inhibitors in KRAS-mutant NSCLC appeared to be a promising approach, as these inhibitors target many KRAS downstream effectors; however, limited clinical efficacy has been observed due to resistance. Here, we examined the mechanism(s) of acquired resistance to the Hsp90 inhibitor, ganetespib, and identified novel and rationally devised Hsp90 inhibitor combinations, which may prevent and overcome resistance to Hsp90 inhibitors. We derived KRAS-mutant NSCLC ganetespib-resistant cell lines to identify the resistance mechanism(s) and identified hyperactivation of RAF/MEK/ERK/RSK and PI3K/AKT/mTOR pathways as key resistance mechanisms. Furthermore, we found that ganetespib-resistant cells are "addicted" to these pathways, as ganetespib resistance leads to synthetic lethality to a dual PI3K/mTOR, a PI3K, or an ERK inhibitor. Interestingly, the levels and activity of a key activator of the mTOR pathway and an ERK downstream target, p90 ribosomal S6 kinase (RSK), were also increased in the ganetespib-resistant cells. Genetic or pharmacologic inhibition of p90RSK in ganetespib-resistant cells restored sensitivity to ganetespib, whereas p90RSK overexpression induced ganetespib resistance in naïve cells, validating p90RSK as a mediator of resistance and a novel therapeutic target. Our studies offer a way forward for Hsp90 inhibitors through the rational design of Hsp90 inhibitor combinations that may prevent and/or overcome resistance to Hsp90 inhibitors, providing an effective therapeutic strategy for KRAS-mutant NSCLC. Mol Cancer Ther; 16(5); 793-804. ©2017 AACR.
Collapse
Affiliation(s)
- Suman Chatterjee
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Eric H-B Huang
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Ian Christie
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Brenda F Kurland
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Timothy F Burns
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
65
|
Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, Grasso CS, Hugo W, Sandoval S, Torrejon DY, Palaskas N, Rodriguez GA, Parisi G, Azhdam A, Chmielowski B, Cherry G, Seja E, Berent-Maoz B, Shintaku IP, Le DT, Pardoll DM, Diaz LA, Tumeh PC, Graeber TG, Lo RS, Comin-Anduix B, Ribas A. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov 2017; 7:188-201. [PMID: 27903500 PMCID: PMC5296316 DOI: 10.1158/2159-8290.cd-16-1223] [Citation(s) in RCA: 985] [Impact Index Per Article: 123.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 01/05/2023]
Abstract
Loss-of-function mutations in JAK1/2 can lead to acquired resistance to anti-programmed death protein 1 (PD-1) therapy. We reasoned that they may also be involved in primary resistance to anti-PD-1 therapy. JAK1/2-inactivating mutations were noted in tumor biopsies of 1 of 23 patients with melanoma and in 1 of 16 patients with mismatch repair-deficient colon cancer treated with PD-1 blockade. Both cases had a high mutational load but did not respond to anti-PD-1 therapy. Two out of 48 human melanoma cell lines had JAK1/2 mutations, which led to a lack of PD-L1 expression upon interferon gamma exposure mediated by an inability to signal through the interferon gamma receptor pathway. JAK1/2 loss-of-function alterations in The Cancer Genome Atlas confer adverse outcomes in patients. We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to primary resistance to PD-1 blockade therapy. SIGNIFICANCE A key functional result from somatic JAK1/2 mutations in a cancer cell is the inability to respond to interferon gamma by expressing PD-L1 and many other interferon-stimulated genes. These mutations result in a genetic mechanism for the absence of reactive PD-L1 expression, and patients harboring such tumors would be unlikely to respond to PD-1 blockade therapy. Cancer Discov; 7(2); 188-201. ©2016 AACR.See related commentary by Marabelle et al., p. 128This article is highlighted in the In This Issue feature, p. 115.
Collapse
Affiliation(s)
| | - Jesse M Zaretsky
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Angel Garcia-Diaz
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Anusha Kalbasi
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Willy Hugo
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Salemiz Sandoval
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Davis Y Torrejon
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Nicolaos Palaskas
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Giulia Parisi
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Ariel Azhdam
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Bartosz Chmielowski
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Grace Cherry
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Elizabeth Seja
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Beata Berent-Maoz
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - I Peter Shintaku
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Dung T Le
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Drew M Pardoll
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Luis A Diaz
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Paul C Tumeh
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Thomas G Graeber
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Roger S Lo
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Begoña Comin-Anduix
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Antoni Ribas
- University of California, Los Angeles (UCLA), Los Angeles, California.
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| |
Collapse
|
66
|
The NF1 gene in tumor syndromes and melanoma. J Transl Med 2017; 97:146-157. [PMID: 28067895 PMCID: PMC5413358 DOI: 10.1038/labinvest.2016.142] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 11/10/2016] [Accepted: 11/29/2016] [Indexed: 02/06/2023] Open
Abstract
Activation of the RAS/MAPK pathway is critical in melanoma. Melanoma can be grouped into four molecular subtypes based on their main genetic driver: BRAF-mutant, NRAS-mutant, NF1-mutant, and triple wild-type tumors. The NF1 protein, neurofibromin 1, negatively regulates RAS proteins through GTPase activity. Germline mutations in NF1 cause neurofibromatosis type I, a common genetic tumor syndrome caused by dysregulation of the RAS/MAPK pathway, ie, RASopathy. Melanomas with NF1 mutations typically occur on chronically sun-exposed skin or in older individuals, show a high mutation burden, and are wild-type for BRAF and NRAS. Additionally, NF1 mutations characterize certain clinicopathologic melanoma subtypes, specifically desmoplastic melanoma. This review discusses the current knowledge of the NF1 gene and neurofibromin 1 in neurofibromatosis type I and in melanoma.
Collapse
|
67
|
Abstract
Loss-of-function mutations in JAK1/2 can lead to acquired resistance to anti-programmed death protein 1 (PD-1) therapy. We reasoned that they may also be involved in primary resistance to anti-PD-1 therapy. JAK1/2-inactivating mutations were noted in tumor biopsies of 1 of 23 patients with melanoma and in 1 of 16 patients with mismatch repair-deficient colon cancer treated with PD-1 blockade. Both cases had a high mutational load but did not respond to anti-PD-1 therapy. Two out of 48 human melanoma cell lines had JAK1/2 mutations, which led to a lack of PD-L1 expression upon interferon gamma exposure mediated by an inability to signal through the interferon gamma receptor pathway. JAK1/2 loss-of-function alterations in The Cancer Genome Atlas confer adverse outcomes in patients. We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to primary resistance to PD-1 blockade therapy. SIGNIFICANCE A key functional result from somatic JAK1/2 mutations in a cancer cell is the inability to respond to interferon gamma by expressing PD-L1 and many other interferon-stimulated genes. These mutations result in a genetic mechanism for the absence of reactive PD-L1 expression, and patients harboring such tumors would be unlikely to respond to PD-1 blockade therapy. Cancer Discov; 7(2); 188-201. ©2016 AACR.See related commentary by Marabelle et al., p. 128This article is highlighted in the In This Issue feature, p. 115.
Collapse
|
68
|
Baby B, Antony P, Al Halabi W, Al Homedi Z, Vijayan R. Structural insights into the polypharmacological activity of quercetin on serine/threonine kinases. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3109-3123. [PMID: 27729770 PMCID: PMC5045902 DOI: 10.2147/dddt.s118423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Polypharmacology, the discovery or design of drug molecules that can simultaneously interact with multiple targets, is gaining interest in contemporary drug discovery. Serine/threonine kinases are attractive targets for therapeutic intervention in oncology due to their role in cellular phosphorylation and altered expression in cancer. Quercetin, a naturally occurring flavonoid, inhibits multiple cancer cell lines and is used as an anticancer drug in Phase I clinical trial. Quercetin glycosides have also received some attention due to their high bioavailability and activity against various diseases including cancer. However, these have been studied to a lesser extent. In this study, the structural basis of the multitarget inhibitory activity of quercetin and isoquercitrin, a glycoside derivative, on serine/threonine kinases using molecular modeling was explored. Structural analysis showed that both quercetin and isoquercitrin exhibited good binding energies and interacted with aspartate in the highly conserved Asp–Phe–Gly motif. The results indicate that isoquercitrin could be a more potent inhibitor of several members of the serine/threonine kinase family. In summary, the current structural evaluation highlights the multitarget inhibitory property of quercetin and its potential to be a chemical platform for oncological polypharmacology.
Collapse
Affiliation(s)
- Bincy Baby
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Priya Antony
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Walaa Al Halabi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Zahrah Al Homedi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
69
|
Gillis NK, McLeod HL. The pharmacogenomics of drug resistance to protein kinase inhibitors. Drug Resist Updat 2016; 28:28-42. [PMID: 27620953 PMCID: PMC5022787 DOI: 10.1016/j.drup.2016.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023]
Abstract
Dysregulation of growth factor cell signaling is a major driver of most human cancers. This has led to development of numerous drugs targeting protein kinases, with demonstrated efficacy in the treatment of a wide spectrum of cancers. Despite their high initial response rates and survival benefits, the majority of patients eventually develop resistance to these targeted therapies. This review article discusses examples of established mechanisms of drug resistance to anticancer therapies, including drug target mutations or gene amplifications, emergence of alternate signaling pathways, and pharmacokinetic variation. This reveals a role for pharmacogenomic analysis to identify and monitor for resistance, with possible therapeutic strategies to combat chemoresistance.
Collapse
Affiliation(s)
- Nancy K Gillis
- Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC, United States; H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States
| | - Howard L McLeod
- H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States; Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
70
|
Lim J, Kelley EH, Methot JL, Zhou H, Petrocchi A, Chen H, Hill SE, Hinton MC, Hruza A, Jung JO, Maclean JKF, Mansueto M, Naumov GN, Philippar U, Raut S, Spacciapoli P, Sun D, Siliphaivanh P. Discovery of 1-(1H-Pyrazolo[4,3-c]pyridin-6-yl)urea Inhibitors of Extracellular Signal-Regulated Kinase (ERK) for the Treatment of Cancers. J Med Chem 2016; 59:6501-11. [PMID: 27329786 DOI: 10.1021/acs.jmedchem.6b00708] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The ERK/MAPK pathway plays a central role in the regulation of critical cellular processes and is activated in more than 30% of human cancers. Specific BRAF and MEK inhibitors have shown clinical efficacy in patients for the treatment of BRAF-mutant melanoma. However, the majority of responses are transient, and resistance is often associated with pathway reactivation of the ERK signal pathway. Acquired resistance to these agents has led to greater interest in ERK, a downstream target of the MAPK pathway. De novo design efforts of a novel scaffold derived from SCH772984 by employing hydrogen bond interactions specific for ERK in the binding pocket identified 1-(1H-pyrazolo[4,3-c]pyridin-6-yl)ureas as a viable lead series. Sequential SAR studies led to the identification of highly potent and selective ERK inhibitors with low molecular weight and high LE. Compound 21 exhibited potent target engagement and strong tumor regression in the BRAF(V600E) xenograft model.
Collapse
Affiliation(s)
- Jongwon Lim
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Elizabeth H Kelley
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Joey L Methot
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Hua Zhou
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Alessia Petrocchi
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Hongmin Chen
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Susan E Hill
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Marlene C Hinton
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Alan Hruza
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Joon O Jung
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - John K F Maclean
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - My Mansueto
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - George N Naumov
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Ulrike Philippar
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Shruti Raut
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Peter Spacciapoli
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Dongyu Sun
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Phieng Siliphaivanh
- Departments of †Chemistry, ‡Oncology, §In Vitro Pharmacology, ∥In Vivo Pharmacology, ⊥Chemistry Modeling and Informatics, #Pharmacokinetics, Pharmacodynamics and Drug Metabolism, and ∇Structural Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
71
|
Johnson DB, Pollack MH, Sosman JA. Emerging targeted therapies for melanoma. Expert Opin Emerg Drugs 2016; 21:195-207. [DOI: 10.1080/14728214.2016.1184644] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
72
|
Zhu G, Yi X, Haferkamp S, Hesbacher S, Li C, Goebeler M, Gao T, Houben R, Schrama D. Combination with γ-secretase inhibitor prolongs treatment efficacy of BRAF inhibitor in BRAF-mutated melanoma cells. Cancer Lett 2016; 376:43-52. [PMID: 27000992 DOI: 10.1016/j.canlet.2016.03.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 12/19/2022]
Abstract
Oncogenic triggering of the MAPK pathway in melanocytes results in senescence, and senescence escape is considered as one critical step for melanocytic transformation. In melanoma, induction of a senescent-like state by BRAF-inhibitors (BRAFi) in a fraction of treated cells - instead of killing - contributes to the repression of tumor growth, but may also provide a source for relapse. Here, we demonstrate that NOTCH activation in melanocytes is not only growth-promoting but it also protects these cells against oncogene-induced senescence. In turn, treatment of melanoma cells with an inhibitor of the NOTCH-activating enzyme γ-secretase led to induction of a senescent-like status in a fraction of the cells but overall achieved only a moderate inhibition of melanoma cell growth. However, combination of γ-secretase inhibitor (GSI) with BRAFi markedly increased the treatment efficacy particularly in long-term culture. Moreover, even melanoma cells starting to regrow after continuous BRAFi treatment - the major problem of BRAFi therapy in patients - can still be affected by the combination treatment. Thus, combining GSI with BRAFi increases the therapeutic efficacy by, at least partially, prolonging the senescent-like state of treated cells.
Collapse
Affiliation(s)
- Guannan Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | - Sonja Hesbacher
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Matthias Goebeler
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Roland Houben
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - David Schrama
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
73
|
Jenkins RW, Sullivan RJ. NRAS mutant melanoma: an overview for the clinician for melanoma management. Melanoma Manag 2016; 3:47-59. [PMID: 30190872 PMCID: PMC6097550 DOI: 10.2217/mmt.15.40] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/06/2015] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer and the incidence continues to rise in the United States and worldwide. Activating mutations in RAS oncogenes are found in roughly a third of all human cancers. Mutations in NRAS occur in approximately a fifth of cutaneous melanomas and are associated with aggressive clinical behavior. Cells harboring oncogenic NRAS mutations exhibit activation of multiple signaling cascades, including PI3K/Akt, MEK-ERK and RAL, which collectively stimulate cancer growth. While strategies to target N-Ras itself have proven ineffective, targeting one or more N-Ras effector pathways has shown promise in preclinical models. Despite promising preclinical data, current therapies for NRAS mutant melanoma remain limited. Immune checkpoint inhibitors and targeted therapies for BRAF mutant melanoma are transforming the treatment of metastatic melanoma, but the ideal treatment for NRAS mutant melanoma remains unknown. Improved understanding of NRAS mutant melanoma and relevant N-Ras effector signaling modules will be essential to develop new treatment strategies.
Collapse
Affiliation(s)
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
74
|
|
75
|
Jha S, Morris EJ, Hruza A, Mansueto MS, Schroeder GK, Arbanas J, McMasters D, Restaino CR, Dayananth P, Black S, Elsen NL, Mannarino A, Cooper A, Fawell S, Zawel L, Jayaraman L, Samatar AA. Dissecting Therapeutic Resistance to ERK Inhibition. Mol Cancer Ther 2016; 15:548-59. [PMID: 26832798 DOI: 10.1158/1535-7163.mct-15-0172] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 12/23/2015] [Indexed: 11/16/2022]
Abstract
The MAPK pathway is frequently activated in many human cancers, particularly melanomas. A single-nucleotide mutation in BRAF resulting in the substitution of glutamic acid for valine (V(600E)) causes constitutive activation of the downstream MAPK pathway. Selective BRAF and MEK inhibitor therapies have demonstrated remarkable antitumor responses in BRAF(V600) (E)-mutant melanoma patients. However, initial tumor shrinkage is transient and the vast majority of patients develop resistance. We previously reported that SCH772984, an ERK 1/2 inhibitor, effectively suppressed MAPK pathway signaling and cell proliferation in BRAF, MEK, and concurrent BRAF/MEK inhibitor-resistant tumor models. ERK inhibitors are currently being evaluated in clinical trials and, in anticipation of the likelihood of clinical resistance, we sought to prospectively model acquired resistance to SCH772984. Our data show that long-term exposure of cells to SCH772984 leads to acquired resistance, attributable to a mutation of glycine to aspartic acid (G(186D)) in the DFG motif of ERK1. Structural and biophysical studies demonstrated specific defects in SCH772984 binding to mutant ERK. Taken together, these studies describe the interaction of SCH772984 with ERK and identify a novel mechanism of ERK inhibitor resistance through mutation of a single residue within the DFG motif. Mol Cancer Ther; 15(4); 548-59. ©2016 AACR.
Collapse
Affiliation(s)
- Sharda Jha
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Erick J Morris
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Alan Hruza
- Early Development and Discovery Sciences, Merck Research Laboratories, Kenilworth, New Jersey
| | - My Sam Mansueto
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Gottfried K Schroeder
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Jaren Arbanas
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Daniel McMasters
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Clifford R Restaino
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Priya Dayananth
- Early Development and Discovery Sciences, Merck Research Laboratories, Kenilworth, New Jersey
| | - Stuart Black
- Early Development and Discovery Sciences, Merck Research Laboratories, Kenilworth, New Jersey
| | - Nathaniel L Elsen
- Early Development and Discovery Sciences, Merck Research Laboratories, Kenilworth, New Jersey
| | - Anthony Mannarino
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Alan Cooper
- Early Development and Discovery Sciences, Merck Research Laboratories, Kenilworth, New Jersey
| | - Stephen Fawell
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Leigh Zawel
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts
| | - Lata Jayaraman
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts.
| | - Ahmed A Samatar
- Early Development and Discovery Sciences, Merck Research Laboratories, Boston, Massachusetts.
| |
Collapse
|
76
|
Akabane H, Sullivan RJ. The Future of Molecular Analysis in Melanoma: Diagnostics to Direct Molecularly Targeted Therapy. Am J Clin Dermatol 2016; 17:1-10. [PMID: 26518880 DOI: 10.1007/s40257-015-0159-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Melanoma is a malignancy of pigment-producing cells that is driven by a variety of genetic mutations and aberrations. In most cases, this leads to upregulation of the mitogen-activated protein kinase (MAPK) pathway through activating mutations of upstream mediators of the pathway including BRAF and NRAS. With the advent of effective MAPK pathway inhibitors, including the US FDA-approved BRAF inhibitors vemurafenib and dabrafenib and MEK inhibitor trametinib, molecular analysis has become an integral part of the care of patients with metastatic melanoma. In this article, the key molecular targets and strategies to inhibit these targets therapeutically are presented, and the techniques of identifying these targets, in both tissue and blood, are discussed.
Collapse
Affiliation(s)
- Hugo Akabane
- Department of Medicine, Metrowest Medical Center, Framingham, MA, USA
| | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
77
|
Liu B, Fu L, Zhang C, Zhang L, Zhang Y, Ouyang L, He G, Huang J. Computational design, chemical synthesis, and biological evaluation of a novel ERK inhibitor (BL-EI001) with apoptosis-inducing mechanisms in breast cancer. Oncotarget 2016; 6:6762-75. [PMID: 25742792 PMCID: PMC4466648 DOI: 10.18632/oncotarget.3105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 01/08/2015] [Indexed: 02/05/2023] Open
Abstract
Extracellular signal-regulated kinase1/2 (ERK1/2) plays a crucial role in the resistance of apoptosis in carcinogenesis; however, its targeted small-molecule inhibitors still remain to be discovered. Thus, in this study, we computationally and experimentally screened a series of small-molecule inhibitors targeting ERK toward different types of human breast cancer cells. Subsequently, we synthesized some candidate ERK inhibitors, identified a novel ERK inhibitor (BL-EI001) with anti-proliferative activities, and analyzed the BL-EI001/ERK complex. Moreover, we found that BL-EI001 induced breast cancer cell apoptosis via mitochondrial pathway but independent on Ras/Raf/MEK pathway. In addition, we carried out proteomics analyses for exploring some possible BL-EI001-induced apoptotic pathways, and further found that BL-EI001-induced apoptosis affected ERK phosphorylation in breast cancer. Further, we found that BL-EI001 bear anti-tumor activities without remarkable toxicities, and also induced mitochondrial apoptosis by targeting ERK in vivo. Taken together, these results demonstrate that in silico design and experimental discovery of a synthesized small-molecule ERK inhibitor (BL-EI001) as a potential novel apoptosis-inducing drug in the treatment of breast cancer.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Leilei Fu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cui Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lan Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yonghui Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.,Collaborative Innovation Center for Biotherapy, Department of Pharmacology & Pharmaceutical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gu He
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
78
|
Abstract
The past several years can be considered a renaissance era in the treatment of metastatic melanoma. Following a 30-year stretch in which oncologists barely put a dent in a very grim overall survival (OS) rate for these patients, things have rapidly changed course with the recent approval of three new melanoma drugs by the FDA. Both oncogene-targeted therapy and immune checkpoint blockade approaches have shown remarkable efficacy in a subset of melanoma patients and have clearly been game-changers in terms of clinical impact. However, most patients still succumb to their disease, and thus, there remains an urgent need to improve upon current therapies. Fortunately, innovations in molecular medicine have led to many silent gains that have greatly increased our understanding of the nature of cancer biology as well as the complex interactions between tumors and the immune system. They have also allowed for the first time a detailed understanding of an individual patient's cancer at the genomic and proteomic level. This information is now starting to be employed at all stages of cancer treatment, including diagnosis, choice of drug therapy, treatment monitoring, and analysis of resistance mechanisms upon recurrence. This new era of personalized medicine will foreseeably lead to paradigm shifts in immunotherapeutic treatment approaches such as individualized cancer vaccines and adoptive transfer of genetically modified T cells. Advances in xenograft technology will also allow for the testing of drug combinations using in vivo models, a truly necessary development as the number of new drugs needing to be tested is predicted to skyrocket in the coming years. This chapter will provide an overview of recent technological developments in cancer research, and how they are expected to impact future diagnosis, monitoring, and development of novel treatments for metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | - Patrick Hwu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Gregory Lizée
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
79
|
Abstract
Vemurafenib and dabrafenib, two potent tyrosine kinase inhibitors (TKIs) of the BRAF(V600E) kinase, are highly effective in the treatment of a BRAF (V600) -mutant metastatic melanoma. These are selective type I inhibitors (functional against the active conformation of the kinase) of the RAF kinases, which are key players in the mitogen-activated protein kinase (MAPK) pathway. BRAF (V600) mutations are present in approximately 7 % of all cancers, including high frequencies of mutations reported in 50 % of advanced melanomas and 100 % of hairy cell leukemias. As with most targeted therapies, resistance to BRAF inhibitors is an issue, and mechanisms of resistance are varied. Combining BRAF inhibitors with MEK inhibitors such as trametinib delays the development of resistance. Rationally combining targeted therapies to address the mechanism of resistance or combining BRAF inhibitors with other effective therapies such as immunotherapy may result in further improvement in outcomes for patients.
Collapse
|
80
|
González-Cao M, Rodón J, Karachaliou N, Sánchez J, Santarpia M, Viteri S, Pilotto S, Teixidó C, Riso A, Rosell R. Other targeted drugs in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:266. [PMID: 26605312 DOI: 10.3978/j.issn.2305-5839.2015.08.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Targeted therapy drugs are developed against specific molecular alterations on cancer cells. Because they are "targeted" to the tumor, these therapies are more effective and better tolerated than conventional therapies such as chemotherapy. In the last decade, great advances have been made in understanding of melanoma biology and identification of molecular mechanisms involved in malignant transformation of cells. The identification of oncogenic mutated kinases involved in this process provides an opportunity for development of new target therapies. The dependence of melanoma on BRAF-mutant kinase has provided an opportunity for development of mutation-specific inhibitors with high activity and excellent tolerance that are now being used in clinical practice. This marked a new era in the treatment of metastatic melanoma and much research is now ongoing to identify other "druggable" kinases and transduction signaling networking. It is expected that in the near future the spectrum of target drugs for melanoma treatment will increase. Herein, we review the most relevant potential novel drugs for melanoma treatment based on preclinical data and the results of early clinical trials.
Collapse
Affiliation(s)
- María González-Cao
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Jordi Rodón
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Niki Karachaliou
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Jesús Sánchez
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Mariacarmela Santarpia
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Santiago Viteri
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Sara Pilotto
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Cristina Teixidó
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Aldo Riso
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| | - Rafael Rosell
- 1 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 2 Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain ; 3 Immunology Department, CNICV, Madrid, Spain ; 4 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy ; 5 Department of Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy ; 6 Pangaea Biotech S.L, Barcelona, Spain ; 7 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain ; 8 Fundación Molecular Oncology Research, Barcelona, Spain
| |
Collapse
|
81
|
Abstract
The three RAS genes comprise the most frequently mutated oncogene family in cancer. With significant and compelling evidence that continued function of mutant RAS is required for tumor maintenance, it is widely accepted that effective anti-RAS therapy will have a significant impact on cancer growth and patient survival. However, despite more than three decades of intense research and pharmaceutical industry efforts, a clinically effective anti-RAS drug has yet to be developed. With the recent renewed interest in targeting RAS, exciting and promising progress has been made. In this review, we discuss the prospects and challenges of drugging oncogenic RAS. In particular we focus on new inhibitors of RAS effector signaling and the ERK mitogen-activated protein kinase cascade.
Collapse
|
82
|
Nörz D, Grottke A, Bach J, Herzberger C, Hofmann BT, Nashan B, Jücker M, Ewald F. Discontinuing MEK inhibitors in tumor cells with an acquired resistance increases migration and invasion. Cell Signal 2015; 27:2191-200. [PMID: 26210887 DOI: 10.1016/j.cellsig.2015.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 07/19/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Development of small molecular inhibitors against BRAF and MEK has been a breakthrough in the treatment of malignant melanoma. However, the long-term effect is foiled in virtually all patients by the emergence of resistant tumor cell populations. Therefore, mechanisms resulting in the acquired resistance against BRAF and MEK inhibitors have gained much attention and several strategies have been proposed to overcome tumor resistance, including interval treatment or withdrawal of these compounds after disease progression. METHODS Using a panel of cell lines with an acquired resistance against MEK inhibitors, we have evaluated the sensitivity of these cells against compounds targeting AKT/mTOR signaling, as well as novel ERK1/2 inhibitors. Furthermore, the effects of withdrawal of MEK inhibitor on migration in resistant cell lines were analyzed. RESULTS We demonstrate that withdrawal of BRAF or MEK inhibitors in tumor cells with an acquired resistance results in reactivation of ERK1/2 signaling and upregulation of EMT-inducing transcription factors, leading to a highly migratory and invasive phenotype of cancer cells. Furthermore, we show that migration in these cells is independent from AKT/mTOR signaling. However, combined targeting of AKT/mTOR using MK-2206 and AZD8055 efficiently inhibits proliferation in all resistant tumor cell lines analyzed. CONCLUSIONS We propose that combined targeting of MEK/AKT/mTOR or treatment with a novel ERK1/2 inhibitor downstream of BRAF/MEK suppresses proliferation as well as migration and invasion in resistant tumor cells. We provide a rationale against the discontinuation of BRAF or MEK inhibitors in patients with an acquired resistance, and provide a rationale for combined targeting of AKT/mTOR and MEK/ERK1/2, or direct targeting of ERK1/2 as an effective treatment strategy.
Collapse
Affiliation(s)
- Dominik Nörz
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany.
| | - Astrid Grottke
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany.
| | - Johanna Bach
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany.
| | - Christiane Herzberger
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany.
| | - Bianca T Hofmann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Germany.
| | - Bjorn Nashan
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse52, 20246 Hamburg, Germany.
| | - Manfred Jücker
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany.
| | - Florian Ewald
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse52, 20246 Hamburg, Germany.
| |
Collapse
|
83
|
Tetrahydropyrrolo-diazepenones as inhibitors of ERK2 kinase. Bioorg Med Chem Lett 2015; 25:3788-92. [PMID: 26259804 DOI: 10.1016/j.bmcl.2015.07.091] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 11/22/2022]
Abstract
A series of structure based drug design hypotheses and focused screening efforts led to the identification of tetrahydropyrrolo-diazepenones with striking potency against ERK2 kinase. The role of fluorination in mitigating microsomal clearance was systematically explored. Ultimately, it was found that fluorination of a cyclopentanol substructure provided significant improvement in both potency and human metabolic stability.
Collapse
|
84
|
Wong DJL, Robert L, Atefi MS, Lassen A, Avarappatt G, Cerniglia M, Avramis E, Tsoi J, Foulad D, Graeber TG, Comin-Anduix B, Samatar A, Lo RS, Ribas A. Erratum to: Antitumor activity of the ERK inhibitor SCH722984 against BRAF mutant, NRAS mutant and wild-type melanoma. Mol Cancer 2015; 14:128. [PMID: 26134498 PMCID: PMC4488977 DOI: 10.1186/s12943-015-0393-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Deborah J L Wong
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA
| | - Lidia Robert
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA.,Universitat Autonoma de Barcelona, UAB, Barcelona, Spain
| | - Mohammad S Atefi
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA
| | - Amanda Lassen
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA
| | - Geetha Avarappatt
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA.,University of Applied Sciences, Vienna, Austria
| | - Michael Cerniglia
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA
| | - Earl Avramis
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA
| | - Jennifer Tsoi
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - David Foulad
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Begonya Comin-Anduix
- Department of Surgery, Division of Surgical-Oncology, UCLA, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1782, USA
| | - Ahmed Samatar
- Discovery Oncology Merck Research Laboratories, Merck Research Laboratories, Boston, Massachusetts, USA
| | - Roger S Lo
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1782, USA.,Department of Medicine, Division of Dermatology, UCLA, Los Angeles, California, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), 11-934 Factor Building, Los Angeles, CA, USA. .,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA. .,Department of Surgery, Division of Surgical-Oncology, UCLA, Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Center at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1782, USA.
| |
Collapse
|
85
|
Bagdanoff JT, Jain R, Han W, Poon D, Lee PS, Bellamacina C, Lindvall M. Ligand efficient tetrahydro-pyrazolopyridines as inhibitors of ERK2 kinase. Bioorg Med Chem Lett 2015; 25:3626-9. [PMID: 26144345 DOI: 10.1016/j.bmcl.2015.06.063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/14/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Abstract
A series of structure based drug design hypotheses and focused screening efforts drove improvements in the potency and lipophilic efficiency of tetrahydro-pyrazolopyridine based ERK2 inhibitors. Elaboration of a fragment chemical lead established a new lipophilic aryl-Tyr interaction resulting in a substantial potency improvement. Subsequent cleavage of the lipophilic moiety led to reconfiguration of the ligand bound binding cleft. The reconfiguration established a polar contact between a newly liberated N-H and a vicinal Asp, resulting in further improvements in lipophilic efficiency and in vitro clearance.
Collapse
Affiliation(s)
- Jeffrey T Bagdanoff
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 250 Massachusetts Ave., Cambridge, MA 02139, USA.
| | - Rama Jain
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 4560 Horton St., Building 4, Emeryville, CA 94608, USA
| | - Wooseok Han
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 4560 Horton St., Building 4, Emeryville, CA 94608, USA
| | - Daniel Poon
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 4560 Horton St., Building 4, Emeryville, CA 94608, USA
| | - Patrick S Lee
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 4560 Horton St., Building 4, Emeryville, CA 94608, USA
| | - Cornelia Bellamacina
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 4560 Horton St., Building 4, Emeryville, CA 94608, USA
| | - Mika Lindvall
- Global Discovery Chemistry/Oncology and Exploratory Chemistry, Novartis Institutes for BioMedical Research, 4560 Horton St., Building 4, Emeryville, CA 94608, USA
| |
Collapse
|
86
|
Wen L, Cheng F, Zhou Y, Yin C. MiR-26a enhances the sensitivity of gastric cancer cells to cisplatin by targeting NRAS and E2F2. Saudi J Gastroenterol 2015; 21:313-9. [PMID: 26458859 PMCID: PMC4632257 DOI: 10.4103/1319-3767.166206] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIMS MiR-26a has been identified as a tumor suppressor in various tumors, but the relationship between miR-26a and the sensitivity of gastric cancer to chemotherapies has not been established. The present study was performed to investigate the effect of miR-26a on drug sensitivity in gastric cancer (GC). MATERIALS AND METHODS The expression level of miRNA-26a in cisplatin-resistant SGC-7901/DDP cells and parent SGC-7901 cells was evaluated by qRT-PCR. The effect of miR-26a on sensitivity of GC cells to cisplatin was assayed using MTS method. The effect of miR-26a on cisplatin-induced apoptosis were determined by Annexin V/propidium iodide (PI) double staining method and flow cytometry. The targets of miR-26a were identified using a luciferase activity assay and miR-26a-mediated target genes expression analysis. Furthermore, the role of the targets neuroblastoma RAS viral (v-ras) oncogene homolog (NRAS) and E2F2 on sensitivity of chemotherapy in GC by MTS and apoptotic cell analysis was assessed. RESULTS We found that miR-26a was downregulated in cisplatin-resistant SGC-7901/DDP cells compared with SGC-7901 cells. Using both gain- and loss-of-function analyses, we further revealed that miR-26a could improve the sensitivity of GC cells to cisplatin. Furthermore, miR-26a has target sites in the 3'-UTR of NRAS and E2F2 by luciferase reporter assay and reduces the expression levels of NRAS and E2F2. In addition, knockdown of NRAS or E2F2 sensitize GC cells to cisplatin. CONCLUSION Our results suggest that miR-26a can improve the sensitivity of GC cells to cisplatin-based chemotherapies through targeting NRAS and E2F2, and provide the first evidence of the potential utility of miR-26a as a sensitizer in chemotherapy for GC.
Collapse
Affiliation(s)
- Lan Wen
- Department of Gastroenterology, Affiliated Zhuzhou People's Hospital, Changsha Medical University, Hunan Province, China
| | - Fangzhi Cheng
- Department of Shaoxing Central Hospital, Shaoxing, Zhejiang Province, Hunan Province, China
| | - Yanyan Zhou
- Department of Intensive Care Unit, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Address for correspondence: Dr. Yanyan Zhou, Intensive Care Unit, the Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha 410011, Hunan Province, China. E-mail:
| | - Chunhua Yin
- Department of Intensive Care Unit, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
87
|
Sullivan R, LoRusso P, Boerner S, Dummer R. Achievements and challenges of molecular targeted therapy in melanoma. Am Soc Clin Oncol Educ Book 2015:177-186. [PMID: 25993155 DOI: 10.14694/edbook_am.2015.35.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The treatment of melanoma has been revolutionized over the past decade with the development of effective molecular and immune targeted therapies. The great majority of patients with melanoma have mutations in oncogenes that predominantly drive signaling through the mitogen activated protein kinase (MAPK) pathway. Analytic tools have been developed that can effectively stratify patients into molecular subsets based on the identification of mutations in oncogenes and/or tumor suppressor genes that drive the MAPK pathway. At the same time, potent and selective inhibitors of mediators of the MAPK pathway such as RAF, MEK, and ERK have become available. The most dramatic example is the development of single-agent inhibitors of BRAF (vemurafenib, dabrafenib, encorafenib) and MEK (trametinib, cobimetinib, binimetinib) for patients with metastatic BRAFV600-mutant melanoma, a subset that represents 40% to 50% of patients with metastatic melanoma. More recently, the elucidation of mechanisms underlying resistance to single-agent BRAF inhibitor therapy led to a second generation of trials that demonstrated the superiority of BRAF inhibitor/MEK inhibitor combinations (dabrafenib/trametinib; vemurafenib/cobimetinib) compared to single-agent BRAF inhibitors. Moving beyond BRAFV600 targeting, a number of other molecular subsets--such as mutations in MEK, NRAS, and non-V600 BRAF and loss of function of the tumor suppressor neurofibromatosis 1 (NF1)--are predicted to respond to MAPK pathway targeting by single-agent pan-RAF, MEK, or ERK inhibitors. As these strategies are being tested in clinical trials, preclinical and early clinical trial data are now emerging about which combinatorial approaches might be best for these patients.
Collapse
Affiliation(s)
- Ryan Sullivan
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Patricia LoRusso
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Scott Boerner
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- From the Massachusetts General Hospital Cancer Center, Boston, MA; Yale Cancer Center, New Haven, CT; Yale University, New Haven, CT; University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
88
|
Evolutionary triage governs fitness in driver and passenger mutations and suggests targeting never mutations. Nat Commun 2014; 5:5499. [PMID: 25407411 PMCID: PMC4260773 DOI: 10.1038/ncomms6499] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/06/2014] [Indexed: 02/06/2023] Open
Abstract
Genetic and epigenetic changes in cancer cells are typically divided into “drivers” and “passengers”. Drug development strategies target driver mutations, but inter- and intra-tumoral heterogeneity usually results in emergence of resistance. Here we model intratumoral evolution in the context of a fecundity/survivorship trade-off. Simulations demonstrate the fitness value, of any genetic change is not fixed but dependent on evolutionary triage governed by initial cell properties, current selection forces, and prior genotypic/phenotypic trajectories. We demonstrate spatial variations in molecular properties of tumor cells are the result of changes in environmental selection forces such as blood flow. Simulated therapies targeting fitness-increasing (driver) mutations usually decrease the tumor burden but almost inevitably fail due to population heterogeneity. An alternative strategy targets gene mutations that are never observed. Because up or down regulation of these genes unconditionally reduces cellular fitness, they are eliminated by evolutionary triage but can be exploited for targeted therapy.
Collapse
|