151
|
Abstract
Activating mutation of KRAS plays a significant role in the pathogenesis of common human malignancies and molecular testing of KRAS mutation has emerged as an essential biomarker in the current practice of clinical oncology. The presence of KRAS mutation is generally associated with clinical aggressiveness of the cancer and reduced survival of the patient. Therapeutically, KRAS mutation testing has maximum utility in stratifying metastatic colorectal carcinoma and lung cancer patients for treatment with targeted therapy. Diagnostically, KRAS mutation testing is useful in the workup of pancreaticobiliary and thyroid cancers, particularly using cytological specimens. In the era of precision medicine, the role of KRAS mutation testing is poised to expand, likely in a setting of combinatorial therapeutic strategy and requiring additional mutation testing of its upstream and/or downstream effectors.
Collapse
Affiliation(s)
- Sudhir Perincheri
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street, New Haven, CT 06520-8023, USA
| | | |
Collapse
|
152
|
Ou SHI. Republished: lung cancer in never-smokers. Does smoking history matter in the era of molecular diagnostics and targeted therapy? Postgrad Med J 2014; 90:228-35. [PMID: 24643262 DOI: 10.1136/postgradmedj-2012-201296rep] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lung cancer in never-smokers was recognised as a distinct clinical entity around the mid-2000s because these patients tended to be Asian women and diagnosed at a younger age with a preponderance of adenocarcinoma and better survival outcome despite a more advanced stage of presentation. It was soon discovered that lung cancer in never-smokers had a higher prevalence of activating EGFR mutations and we tend to classify lung cancer by smoking status for screening purpose. With the discoveries of many actionable driver mutations such as activating EGFR mutations and ALK rearrangement in adenocarcinoma of the lung we have switched to classifying non-small cell lung cancer into different individual molecular subgroups based on the presence of a dominant driver mutation. Although many actionable driver mutations are found in never-smokers with adenocarcinoma, this review will summarise that a substantial proportion of patients with these actionable driver mutations had a previous smoking history. Alternatively among the driver mutations that are associated with smoking history, a fair amount of these patients were never-smokers. Thus smoking status should not be used as a screen strategy for identifying driver mutations in clinical practice. Finally smoking history may have predictive and/or prognostic significance within individual molecular subgroups and identifying the difference according to smoking history may help optimise future targeted therapy.
Collapse
|
153
|
Affiliation(s)
- Donal P McLornan
- From King's College Hospital NHS Foundation Trust, London (D.P.M., G.J.M.); and Moffitt Cancer Center, Tampa, FL (A.L.)
| | | | | |
Collapse
|
154
|
Felip E, Concha Á, de Castro J, Gómez-Román J, Garrido P, Ramírez J, Isla D, Sanz J, Paz-Ares L, López-Ríos F. Biomarker testing in advanced non-small-cell lung cancer: a National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology. Clin Transl Oncol 2014; 17:103-12. [PMID: 25351175 DOI: 10.1007/s12094-014-1248-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/13/2014] [Indexed: 01/13/2023]
Abstract
In 2011, the Spanish Society of Medical Oncology and the Spanish Society of Pathology started a joint project to establish recommendations on biomarker testing in patients with advanced non-small-cell lung cancer based on the current evidence. Most of these recommendations are still valid, but new evidence requires some aspects to be updated. Specifically, the recommendation about which biomarkers to test in which patients is being amended, and the best way to manage tumour samples and minimum requirements for biomarker test material are defined. Suitable techniques for testing for epidermal growth factor receptor mutations and anaplastic lymphoma kinase rearrangement are also reviewed, and a consensus is reached on which situations warrant re-biopsy.
Collapse
Affiliation(s)
- E Felip
- Department of Medical Oncology, Oncology Service, Hospital Universitari Vall d'Hebron, P. Vall d'Hebron, 119-129, 08035, Barcelona, Spain,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Zugazagoitia J, Enguita AB, Nuñez JA, Iglesias L, Ponce S. The new IASLC/ATS/ERS lung adenocarcinoma classification from a clinical perspective: current concepts and future prospects. J Thorac Dis 2014; 6:S526-36. [PMID: 25349703 DOI: 10.3978/j.issn.2072-1439.2014.01.27] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 01/30/2014] [Indexed: 12/27/2022]
Abstract
The new the International Association for the Study of Lung Cancer (IASLC)/the American Thoracic Society (ATS)/the European Respiratory Society (ERS) pathologic classification of lung cancer has markedly changed the pathologic diagnosis of lung adenocarcinoma. This classification deals with many aspects that directly affect clinical practice, and opens new gateways for future research. By means of a multidisciplinary approach, it differs significantly from the former 2004 the World Health Organization (WHO) classification, which was mainly written by pathologist. The present review, in line with the consensus article, is divided in two components: the diagnosis and classification of lung adenocarcinoma in resection specimens and the diagnosis of lung cancer in small biopsies and cytology. Resection specimens are currently classified according to the predominant histologic pattern after comprehensive subtyping in 5% increments. This approach has led to the addition of new pathologic subtypes [adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA) and micropapillary predominant adenocarcinoma)] and to the discontinuation of some heterogeneous entities included in the former 2004 WHO classification (mixed subtype adenocarcinoma and bronchioloalveolar carcinoma). Overall, these changes have resulted in a better stratification of lung adenocarcinoma tumors in more homogeneous morphologic, clinical and biological subgroups. Pathologic subtyping has demonstrated prognostic utility in resected stage I-III patients, and recent data support their predictive role for the benefit of adjuvant chemotherapy. Moreover, comprehensive pathologic subtyping may potentially affect TNM staging and surgical management or early-stage tumors. On the other hand, for the first time, the novel pathologic classification provides standardized terminology and diagnostic criteria of small biopsies and cytology. Criteria are proposed not only for adenocarcinoma but also for other histologies, but special emphasis was put on the distinction between adenocarcinoma and squamous-cell carcinoma due to its major clinical implications. This review outlines the main issues of the new lung adenocarcinoma classification from a clinical perspective. We describe the different pathologic subtypes in resection specimens, with their most relevant clinical implications. Further on, we address the new terminology and diagnostic criteria for lung adenocarcinomas in small specimens, oriented to their importance for the management and treatment of metastatic lung cancer patients. Finally, we discuss some unanswered questions and relevant issues for the near future.
Collapse
Affiliation(s)
- Jon Zugazagoitia
- 1 Medical Oncology Department, 2 Pathology Department, Hospital 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Ana Belen Enguita
- 1 Medical Oncology Department, 2 Pathology Department, Hospital 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Juan Antonio Nuñez
- 1 Medical Oncology Department, 2 Pathology Department, Hospital 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Lara Iglesias
- 1 Medical Oncology Department, 2 Pathology Department, Hospital 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Santiago Ponce
- 1 Medical Oncology Department, 2 Pathology Department, Hospital 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| |
Collapse
|
156
|
Stinchcombe TE. Novel agents in development for advanced non-small cell lung cancer. Ther Adv Med Oncol 2014; 6:240-53. [PMID: 25342991 DOI: 10.1177/1758834014532510] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The identification of EGFR mutations and ALK rearrangements in nonsmall cell lung cancer (NSCLC) has led to the rapid development of targeted therapies and significant changes in the treatment paradigm. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) and crizotinib are now standard therapies for patients with the appropriate molecular alteration. Current investigations are determining the mechanisms of resistance to targeted therapies and developing novel agents to combat resistance. For patients with KRAS mutant NSCLC, a phase III trial of the MEK inhibitor, selumetinib, has been initiated. For patients without a defined mutation or a mutation without a known targeted therapy, immunotherapy, ganetespib, nintedanib and MET inhibitors in combination with EGFR TKIs are in development. Preliminary results of phase III trials raise doubts about the future development of dacomitinib as a second-line agent.
Collapse
Affiliation(s)
- Thomas E Stinchcombe
- University of North Carolina at Chapel Hill, 170 Manning Drive, POB 3rd Floor, Chapel Hill, NC 27599-7305, USA
| |
Collapse
|
157
|
Rotella V, Fornaro L, Vasile E, Tibaldi C, Boldrini L, Chella A, D'Incecco A, Cirigliano G, Chioni A, Lupi C, Sensi E, Ginocchi L, Giovannelli S, Pennucci MC, Fontanini G, Baldini E. EGFR and K-Ras mutations in women with lung adenocarcinoma: implications for treatment strategy definition. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:77. [PMID: 25300933 PMCID: PMC4198726 DOI: 10.1186/s13046-014-0077-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 09/08/2014] [Indexed: 11/13/2022]
Abstract
Background We aimed at investigating the outcomes of female patients with stage IIIB-IV adenocarcinoma of the lung according to EGFR and K-Ras mutational status. Methods One hundred and three consecutive female patients genotyped at a single Italian Institution were analyzed. Patients were planned to receive first-line platinum-based chemotherapy (CT) and a salvage treatment with anti-EGFR tyrosine-kinase inhibitors (TKIs) was proposed irrespective of tumor mutational status. EGFR (exons 18–21) and K-Ras (exon 2, codons 12–13) mutations were evaluated by real-time PCR and pyrosequencing. The association of mutational status with clinical variables and treatment benefit was investigated by chi-square test and log-rank test. Results EGFR and K-Ras mutations were found in 31 (30%) and 13 (15%) cases, respectively. Sixty-six patients received platinum CT: no correlation was observed between EGFR or K-Ras mutational status and response rate (RR) (p > 0.05). However, patients treated with first-line CT harboring EGFR activating mutations experienced a significantly reduced progression-free survival (PFS) in comparison with wild-type ones (4.4 vs. 6.4 months, respectively; HR 0.597, 95% CI 0.287-0.975; p = 0.048). Thirty-nine patients received salvage treatment with erlotinib: EGFR activating mutations were significantly correlated with RR (60% vs. 12.5%; p = 0.004) and PFS (11.4 vs. 4.5 months; HR 0.491, 95% CI 0.216-0.936; p = 0.044). Responses to erlotinib were not reported among women with K-Ras mutant tumors, while 50% of those with wild-type K-Ras achieved an objective remission (p = 0.296). Median PFS (3.5 vs. 8.8 months; HR 0.284, 95% CI 0.015-0.510; p = 0.010) and OS (3.9 vs. 19.8 months; HR 0.158, 95% CI 0.001-0.075; p < 0.001) were significantly shorter among K-Ras mutant patients treated with TKI. Conclusions In our population of Caucasian women with advanced lung adenocarcinoma we observed that the presence of EGFR activating mutations correlates with a significant reduction in the benefit from first-line platinum-based CT, emphasizing the importance of an upfront use of anti-EGFR TKIs in this patient subset. K-Ras mutations seem to correlate with a detrimental effect from anti-EGFR TKI, but this finding deserves further investigation.
Collapse
|
158
|
Schink JC, Trosman JR, Weldon CB, Siziopikou KP, Tsongalis GJ, Rademaker AW, Patel JD, Benson AB, Perez EA, Gradishar WJ. Biomarker testing for breast, lung, and gastroesophageal cancers at NCI designated cancer centers. J Natl Cancer Inst 2014; 106:dju256. [PMID: 25217578 PMCID: PMC4176043 DOI: 10.1093/jnci/dju256] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/03/2014] [Accepted: 07/17/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Molecular biomarkers, a cornerstone of precision oncology, are critical in breast, gastroesophageal, and non-small cell lung cancer management (BC, GEC, NSCLC). Testing practices are intensely debated, impacting diagnostic quality and affecting pathologists, oncologists and patients. However, little is known about testing approaches used in practice. Our study described biomarker practices in BC, GEC, and NSCLC at the leading US cancer centers. METHODS We conducted a survey of the National Cancer Institute (NCI) designated centers on BC, GEC, and NSCLC biomarker testing. We used simple frequencies to describe practices, two-sided Fisher's exact test and two-sided McNemar's test for cross-cancer comparison. All statistical tests were two-sided. RESULTS For BC human epidermal growth factor receptor 2 (HER2), 39% of centers combine guidelines by using in situ hybridization (ISH) and immunohistochemistry (IHC) concurrently, and 21% reflex-test beyond guideline-recommended IHC2+. For GEC HER2, 44% use ISH and IHC concurrently, and 28% reflex-test beyond IHC2+. In NSCLC, the use of IHC is limited to 4% for epidermal growth factor receptor (EGFR) and 7% for anaplastic lymphoma kinase (ALK). 43.5% test NSCLC biomarkers on oncologist order; 34.5% run all biomarkers upfront, and 22% use a sequential protocol. NSCLC external testing is statistically significantly higher than BC (P < .0001) and GEC (P < .0001). NSCLC internally developed tests are statistically significantly more common than BC (P < .0001) and GEC (P < .0001). CONCLUSIONS At the NCI cancer centers, biomarker testing practices vary, but exceeding guidelines is a common practice for established biomarkers and emerging practice for newer biomarkers. Use of internally developed tests declines as biomarkers mature. Implementation of multibiomarker protocols is lagging. Our study represents a step toward developing a biomarker testing practice landscape.
Collapse
Affiliation(s)
- Julian C Schink
- * Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Julia R Trosman
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Christine B Weldon
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Kalliopi P Siziopikou
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Gregory J Tsongalis
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Alfred W Rademaker
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Jyoti D Patel
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Al B Benson
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - Edith A Perez
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI
| | - William J Gradishar
- Northwestern University Feinberg School of Medicine, Chicago, IL (JCS, JRT, CBW, KPS, AWR, JDP, ABB, WJG); Center for Business Models in Healthcare, Chicago, IL (JRT, CBW); UCSF Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California, San Francisco, CA (JRT); Department of Pathology, Dartmouth Hitchcock Medical Center and the Audrey and Theodor Geisel School of Medicine, Dartmouth College, Lebanon, NH (GJT); Mayo Clinic Cancer Center, Mayo Clinic, Jacksonville, FL (EAP).* Current affiliation: Spectrum Health Medical Group, Grand Rapids, MI.
| |
Collapse
|
159
|
KRAS-G12C Mutation Is Associated with Poor Outcome in Surgically Resected Lung Adenocarcinoma. J Thorac Oncol 2014; 9:1513-22. [DOI: 10.1097/jto.0000000000000305] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
160
|
Stinchcombe TE, Johnson GL. MEK inhibition in non-small cell lung cancer. Lung Cancer 2014; 86:121-5. [PMID: 25257766 DOI: 10.1016/j.lungcan.2014.09.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 09/07/2014] [Indexed: 01/07/2023]
Abstract
KRAS mutations are the most common mutations in non-small cell lung cancer (NSCLC) with adenocarcinoma histology. KRAS mutations result in the activation of the RAF-MEK-ERK pathway, and agents that target RAF-MEK-ERK pathways have been investigated in KRAS mutant NSCLC. The two agents furthest in development are selumetinib and trametinib. Trametinib has greater binding for the MEK1/2 allosteric site, and generally has superior pharmacokinetics. A randomized phase II trial of docetaxel with and without selumetinib revealed that the combination resulted numerically superior overall survival, and a statistically significant improvement in progression-free survival and objective response rate. However, a concerning rate of hospital admission, grade 3 or 4 neutropenia, and febrile neutropenia was observed with the combination. Trials have investigated MEK inhibitors as single agents and in combination with erlotinib, and the data do not support the further development. The activity of MEK inhibitors appears to be similar in patients with KRAS mutant and wild-type NSCLC suggesting KRAS mutation status is not a reliable biomarker for efficacy. It is possible that mutations of genes in addition to KRAS mutations impact the activity of MEK inhibitors, or specific subsets of KRAS mutations may be resistant or susceptible to MEK inhibition. Other potential explanations are gene amplifications, alternative RNA splicing of genes resulting in activation of their protein products, and deregulation of noncoding RNAs and consequent altered protein expression.
Collapse
Affiliation(s)
- Thomas E Stinchcombe
- Division of Hematology and Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| | - Gary L Johnson
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
161
|
Martín-Bernabé A, Cortés R, Lehmann SG, Seve M, Cascante M, Bourgoin-Voillard S. Quantitative proteomic approach to understand metabolic adaptation in non-small cell lung cancer. J Proteome Res 2014; 13:4695-704. [PMID: 25029028 DOI: 10.1021/pr500327v] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
KRAS mutations in non-small cell lung cancer (NSCLC) are a predictor of resistance to EGFR-targeted therapies. Because approaches to target RAS signaling have been unsuccessful, targeting lung cancer metabolism might help to develop a new strategy that could overcome drug resistance in such cancer. In this study, we applied a large screening quantitative proteomic analysis to evidence key enzymes involved in metabolic adaptations in lung cancer. We carried out the proteomic analysis of two KRAS-mutated NSCLC cell lines (A549 and NCI-H460) and a non tumoral bronchial cell line (BEAS-2B) using an iTRAQ (isobaric tags for relative and absolute quantitation) approach combined with two-dimensional fractionation (OFFGEL/RP nanoLC) and MALDI-TOF/TOF mass spectrometry analysis. Protein targets identified by our iTRAQ approach were validated by Western blotting analysis. Among 1038 proteins identified and 834 proteins quantified, 49 and 82 proteins were respectively found differently expressed in A549 and NCI-H460 cells compared to the BEAS-2B non tumoral cell line. Regarding the metabolic pathways, enzymes involved in glycolysis (GAPDH/PKM2/LDH-A/LDH-B) and pentose phosphate pathway (PPP) (G6PD/TKT/6PGD) were up-regulated. The up-regulation of enzyme expression in PPP is correlated to their enzyme activity and will be further investigated to confirm those enzymes as promising metabolic targets for the development of new therapeutic treatments or biomarker assay for NSCLC.
Collapse
Affiliation(s)
- Alfonso Martín-Bernabé
- Department of Biochemistry and Molecular Biology, IBUB, Faculty of Biology, Universitat de Barcelona and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , 08007 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
162
|
Korpanty GJ, Graham DM, Vincent MD, Leighl NB. Biomarkers That Currently Affect Clinical Practice in Lung Cancer: EGFR, ALK, MET, ROS-1, and KRAS. Front Oncol 2014; 4:204. [PMID: 25157335 PMCID: PMC4127527 DOI: 10.3389/fonc.2014.00204] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/16/2014] [Indexed: 12/31/2022] Open
Abstract
Lung cancer remains the most lethal malignancy in the world. Despite improvements in surgical treatment, systemic therapy, and radiotherapy, the 5-year survival rate for all patients diagnosed with lung cancer remains between 15 and 20%. Newer therapeutic strategies rely on specific molecular alterations, or biomarkers, that provide opportunities for a personalized approach to specific patient populations. Classification of lung cancer is becoming increasingly focused on these biomarkers, which renders the term "non-small cell lung" cancer less clinically useful. Non-small cell lung cancer is now recognized as a complex malignancy and its molecular and genomic diversity allows for patient-centered treatment options. Here, we review advances in targeted treatment of lung adenocarcinoma with respect to five clinically relevant biomarkers - EGFR, ALK, MET, ROS-1, and KRAS.
Collapse
Affiliation(s)
- Grzegorz J. Korpanty
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Donna M. Graham
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Mark D. Vincent
- London Regional Cancer Program, Department of Medical Oncology, London Health Sciences Centre, London, ON, Canada
| | - Natasha B. Leighl
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
163
|
Li LH, Wu P, Lee JY, Li PR, Hsieh WY, Ho CC, Ho CL, Chen WJ, Wang CC, Yen MY, Yang SM, Chen HW. Hinokitiol induces DNA damage and autophagy followed by cell cycle arrest and senescence in gefitinib-resistant lung adenocarcinoma cells. PLoS One 2014; 9:e104203. [PMID: 25105411 PMCID: PMC4126702 DOI: 10.1371/journal.pone.0104203] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 07/07/2014] [Indexed: 12/28/2022] Open
Abstract
Despite good initial responses, drug resistance and disease recurrence remain major issues for lung adenocarcinoma patients with epidermal growth factor receptor (EGFR) mutations taking EGFR-tyrosine kinase inhibitors (TKI). To discover new strategies to overcome this issue, we investigated 40 essential oils from plants indigenous to Taiwan as alternative treatments for a wide range of illnesses. Here, we found that hinokitiol, a natural monoterpenoid from the heartwood of Calocedrus formosana, exhibited potent anticancer effects. In this study, we demonstrated that hinokitiol inhibited the proliferation and colony formation ability of lung adenocarcinoma cells as well as the EGFR-TKI-resistant lines PC9-IR and H1975. Transcriptomic analysis and pathway prediction algorithms indicated that the main implicated pathways included DNA damage, autophagy, and cell cycle. Further investigations confirmed that in lung cancer cells, hinokitiol inhibited cell proliferation by inducing the p53-independent DNA damage response, autophagy (not apoptosis), S-phase cell cycle arrest, and senescence. Furthermore, hinokitiol inhibited the growth of xenograft tumors in association with DNA damage and autophagy but exhibited fewer effects on lung stromal fibroblasts. In summary, we demonstrated novel mechanisms by which hinokitiol, an essential oil extract, acted as a promising anticancer agent to overcome EGFR-TKI resistance in lung cancer cells via inducing DNA damage, autophagy, cell cycle arrest, and senescence in vitro and in vivo.
Collapse
Affiliation(s)
- Lan-Hui Li
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory, Kunming Branch, Taipei City Hospital, Taipei, Taiwan
| | - Ping Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jen-Yi Lee
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Rong Li
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Yu Hsieh
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chao-Chi Ho
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University Medical College, Taipei, Taiwan
| | - Chen-Lung Ho
- Division of Wood Cellulose, Taiwan Forestry Research Institute, Taipei, Taiwan
| | - Wan-Jiun Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chun Wang
- Division of Infectious Diseases, Kunming Branch, Taipei City Hospital, Taipei, Taiwan
| | - Muh-Yong Yen
- Division of Infectious Diseases, Kunming Branch, Taipei City Hospital, Taipei, Taiwan
- Department of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shun-Min Yang
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
164
|
Weng TY, Yen MC, Huang CT, Hung JJ, Chen YL, Chen WC, Wang CY, Chang JY, Lai MD. DNA vaccine elicits an efficient antitumor response by targeting the mutant Kras in a transgenic mouse lung cancer model. Gene Ther 2014; 21:888-96. [PMID: 25077772 DOI: 10.1038/gt.2014.67] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/09/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022]
Abstract
Mutant Kras (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog) is observed in more than 20% of non-small-cell lung cancers; however, no effective Kras target therapy is available at present. The Kras DNA vaccine may represent as a novel immunotherapeutic agent in lung cancer. In this study, we investigated the antitumor efficacy of the Kras DNA vaccine in a genetically engineered inducible mouse lung tumor model driven by Kras(G12D). Lung tumors were induced by doxycycline, and the therapeutic effects of Kras DNA vaccine were evaluated with delivery of Kras(G12D) plasmids. Mutant Kras(G12D) DNA vaccine significantly decreased the tumor nodules. A dominant-negative mutant Kras(G12D)N17, devoid of oncogenic activity, achieved similar therapeutic effects. The T-helper 1 immune response was enhanced in mice treated with Kras DNA vaccine. Splenocytes from mice receiving Kras DNA vaccine presented an antigen-specific response by treatment with peptides of Kras but not Hras or OVA. The number of tumor-infiltrating CD8(+) T cells increased after Kras vaccination. In contrast, Kras DNA vaccine was not effective in the lung tumor in transgenic mice, which was induced by mutant L858R epidermal growth factor receptor. Overall, these results indicate that Kras DNA vaccine produces an effective antitumor response in transgenic mice, and may be useful in treating lung cancer-carrying Ras mutation.
Collapse
Affiliation(s)
- T-Y Weng
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - M-C Yen
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [3] Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan, ROC
| | - C-T Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - J-J Hung
- Institute of Bioinformatics and Biosignal Transduction, College of Life Science, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Y-L Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan, ROC
| | - W-C Chen
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - C-Y Wang
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - J-Y Chang
- National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan, ROC
| | - M-D Lai
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [3] Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan, ROC
| |
Collapse
|
165
|
Sarosi V, Losonczy G, Francovszky E, Tolnay E, Torok S, Galffy G, Hegedus B, Dome B, Ostoros G. Effectiveness of erlotinib treatment in advanced KRAS mutation-negative lung adenocarcinoma patients: Results of a multicenter observational cohort study (MOTIVATE). Lung Cancer 2014; 86:54-8. [PMID: 25129367 DOI: 10.1016/j.lungcan.2014.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Erlotinib is an epidermal growth factor receptor tyrosine-kinase inhibitor (EGFR-TKI), used for the treatment of non-small cell lung cancer. As the clinical significance of KRAS mutational status has not yet been clearly determined in this setting, our aim was to investigate the efficacy of erlotinib in advanced KRAS mutation-negative lung adenocarcinoma patients. MATERIALS AND METHODS MOTIVATE is an open-label, multicenter, observational trial with Tarceva(®) (erlotinib) monotherapy. Enrolled patients with advanced (stage IIIB/IV) KRAS wild type (WT) lung adenocarcinoma refractory to one or two courses of prior chemotherapy were treated with erlotinib at 150mg/day. The primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS) and best tumor response rate (RR). RESULTS AND CONCLUSION In total, 327 patients were included. Median PFS and OS were 3.3 and 14.4 months, respectively. Three patients (1.2%) had complete response, 51 patients (20.2%) had partial response and 123 patients (48.8%) had SD. Significantly longer median PFS and OS were observed in Eastern Oncology Cooperative Group Performance Status (ECOG PS) 0-1 patients, as compared to ECOG PS 2-3 patients. The longest median OS (20.5 months) was found in patients with ECOG PS 0-1 who received erlotinib as a second-line therapy. There was no difference in median OS in cohorts stratified to disease stage and smoking status. Female patients had both longer median PFS and OS. Disease control rate was 70.2%. Our results suggest that erlotinib represents a valid treatment option for patients with KRAS WT lung adenocarcinoma and, moreover, that KRAS mutation analysis could help to identify clinically relevant subgroups of NSCLC patients that may benefit from EGFR-TKI therapy.
Collapse
Affiliation(s)
- Veronika Sarosi
- Department of Respiratory Medicine, I. Internal Medicine, Medical University, Ifjúság út 13, 7624 Pécs, Hungary
| | - Gyorgy Losonczy
- Department of Pulmonology, Semmelweis University, Diósárok utca 1/C, 1125 Budapest, Hungary
| | - Eva Francovszky
- National Koranyi Institute of TB and Pulmonology, Pihenő út 1, 1121 Budapest, Hungary
| | - Edina Tolnay
- Klinik für Onkologie und Palliativmedizin, Helios-Klinikum-Aue, Gartenstr. 6, 08280 Aue Sachsen, Germany
| | - Szilvia Torok
- National Koranyi Institute of TB and Pulmonology, Pihenő út 1, 1121 Budapest, Hungary
| | - Gabriella Galffy
- Department of Pulmonology, Semmelweis University, Diósárok utca 1/C, 1125 Budapest, Hungary
| | - Balazs Hegedus
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; MTA-SE Tumor Progression Research Group, Hungarian Academy of Sciences, Üllői út 93, 1091 Budapest, Hungary
| | - Balazs Dome
- National Koranyi Institute of TB and Pulmonology, Pihenő út 1, 1121 Budapest, Hungary; Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Ráth György u. 7-9, 1122 Budapest, Hungary.
| | - Gyula Ostoros
- Department of Pulmonology, Semmelweis University, Diósárok utca 1/C, 1125 Budapest, Hungary.
| |
Collapse
|
166
|
Mak KS, Gainor JF, Niemierko A, Oh KS, Willers H, Choi NC, Loeffler JS, Sequist LV, Shaw AT, Shih HA. Significance of targeted therapy and genetic alterations in EGFR, ALK, or KRAS on survival in patients with non-small cell lung cancer treated with radiotherapy for brain metastases. Neuro Oncol 2014; 17:296-302. [PMID: 25053852 DOI: 10.1093/neuonc/nou146] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND We determined the impact of genetic alterations in EGFR, ALK, or KRAS on survival after radiotherapy for brain metastases in non-small cell lung cancer (NSCLC). METHODS Of 172 genotyped NSCLC patients treated with radiotherapy for brain metastases in 2005-2012, 54 had cancers with EGFR mutations, 12 had ALK rearrangements, 38 had KRAS mutations, and 68 were wild-type (WT). Overall survival (OS) was determined. RESULTS Median follow-up was 8.6 months. Median OS was 13.6 months for patients with EGFR mutations and 26.3 months for patients with ALK rearrangements, in contrast to 5.7 months for KRAS-mutant patients and 5.5 months for WT patients (P = .001). On multivariate analysis, adjusting for receipt of targeted therapy after cranial radiotherapy, ALK rearrangements were associated with improved OS (HR, 0.31; 95% CI, 0.13-0.74; P = .008). EGFR mutations were not significantly associated with improved OS on multivariate analysis (HR, 0.71; 95% CI, 0.37-1.38; P = .3). KRAS mutations were also not associated with improved OS (HR, 0.93; 95% CI, 0.59-1.47; P = .8). Receipt of targeted therapy after cranial radiotherapy was independently associated with improved OS (HR, 0.30; 95% CI, 0.17-0.54; P < .001). Receipt of chemotherapy after cranial radiotherapy, number of brain metastases, extracranial metastases, age, and performance status were also associated with OS. CONCLUSIONS NSCLC patients with genetic alterations in ALK have improved survival outcomes after radiotherapy for brain metastases compared with EGFR, KRAS, or WT. Subsequent receipt of targeted therapy was associated with additional improvement in OS.
Collapse
Affiliation(s)
- Kimberley S Mak
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Justin F Gainor
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Andrzej Niemierko
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Kevin S Oh
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Henning Willers
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Noah C Choi
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Jay S Loeffler
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Lecia V Sequist
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Alice T Shaw
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| | - Helen A Shih
- Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, Massachusetts (K.S.M.); Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.F.G.); Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts (A.N., K.S.O., H.W., N.C.C., J.S.L., H.A.S.); Massachusetts General Hospital Cancer Center, Boston, Massachusetts (L.V.S., A.T.S.)
| |
Collapse
|
167
|
Subtype-specific KRAS mutations in advanced lung adenocarcinoma: A retrospective study of patients treated with platinum-based chemotherapy. Eur J Cancer 2014; 50:1819-1828. [DOI: 10.1016/j.ejca.2014.04.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/18/2013] [Accepted: 02/21/2014] [Indexed: 11/23/2022]
|
168
|
Vaeteewoottacharn K, Seubwai W, Bhudhisawasdi V, Okada S, Wongkham S. Potential targeted therapy for liver fluke associated cholangiocarcinoma. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2014; 21:362-370. [PMID: 24408866 DOI: 10.1002/jhbp.65] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biliary tree cancer or cholangiocarcinoma (CCA) is an unusual subtype of liver cancer with exceptionally poor prognosis. Lack of specific symptoms and availability of early diagnostic markers account for late diagnosis of CCA. Surgical treatment is a gold standard choice but few patients are candidates and local recurrence after surgery is high. Benefit of systemic chemotherapy is limited; hence, better treatment options are required. The differences in etiology, anatomical positions and pathology make it difficult to generalize all CCA subtypes for a single treatment regimen. Herein, we review the uniqueness of molecular profiling identified by multiple approaches, for example, serial analysis of gene expression, exome sequencing, transcriptomics/proteomics profiles, protein kinase profile, etc., that provide the opportunity for treatment of liver fluke-associated CCA. Anti-inflammatory, immunomodulator/immunosuppressor, epidermal growth factor receptor or platelet-derived growth factor receptor inhibitors, multi-targeted tyrosine kinase inhibitor, IL6 antagonist, nuclear factor-κB inhibitor, histone modulator, proteasome inhibitor as well as specific inhibitors suggested from various study approaches, such as MetAP2 inhibitor, 1,25(OH)2 D3 and cyclosporine A are suggested in this review for the treatments of this specific CCA subtype. This might provide an alternative treatment option for CCA patients; however, clinical trials in this specific CCA group are required.
Collapse
Affiliation(s)
- Kulthida Vaeteewoottacharn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 123 Mitraparb Road, Khon Kaen, 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | | | | | | | | |
Collapse
|
169
|
Kloth M, Buettner R. Changing histopathological diagnostics by genome-based tumor classification. Genes (Basel) 2014; 5:444-59. [PMID: 24879454 PMCID: PMC4094942 DOI: 10.3390/genes5020444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 05/05/2014] [Accepted: 05/08/2014] [Indexed: 12/18/2022] Open
Abstract
Traditionally, tumors are classified by histopathological criteria, i.e., based on their specific morphological appearances. Consequently, current therapeutic decisions in oncology are strongly influenced by histology rather than underlying molecular or genomic aberrations. The increase of information on molecular changes however, enabled by the Human Genome Project and the International Cancer Genome Consortium as well as the manifold advances in molecular biology and high-throughput sequencing techniques, inaugurated the integration of genomic information into disease classification. Furthermore, in some cases it became evident that former classifications needed major revision and adaption. Such adaptations are often required by understanding the pathogenesis of a disease from a specific molecular alteration, using this molecular driver for targeted and highly effective therapies. Altogether, reclassifications should lead to higher information content of the underlying diagnoses, reflecting their molecular pathogenesis and resulting in optimized and individual therapeutic decisions. The objective of this article is to summarize some particularly important examples of genome-based classification approaches and associated therapeutic concepts. In addition to reviewing disease specific markers, we focus on potentially therapeutic or predictive markers and the relevance of molecular diagnostics in disease monitoring.
Collapse
Affiliation(s)
- Michael Kloth
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, Cologne D-50937, Germany.
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, Cologne D-50937, Germany.
| |
Collapse
|
170
|
El-Chaar NN, Piccolo SR, Boucher KM, Cohen AL, Chang JT, Moos PJ, Bild AH. Genomic classification of the RAS network identifies a personalized treatment strategy for lung cancer. Mol Oncol 2014; 8:1339-54. [PMID: 24908424 DOI: 10.1016/j.molonc.2014.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 05/09/2014] [Indexed: 01/06/2023] Open
Abstract
Better approaches are needed to evaluate a single patient's drug response at the genomic level. Targeted therapy for signaling pathways in cancer has met limited success in part due to the exceedingly interwoven nature of the pathways. In particular, the highly complex RAS network has been challenging to target. Effectively targeting the pathway requires development of techniques that measure global network activity to account for pathway complexity. For this purpose, we used a gene-expression-based biomarker for RAS network activity in non-small cell lung cancer (NSCLC) cells, and screened for drugs whose efficacy was significantly highly correlated to RAS network activity. Results identified EGFR and MEK co-inhibition as the most effective treatment for RAS-active NSCLC amongst a panel of over 360 compounds and fractions. RAS activity was identified in both RAS-mutant and wild-type lines, indicating broad characterization of RAS signaling inclusive of multiple mechanisms of RAS activity, and not solely based on mutation status. Mechanistic studies demonstrated that co-inhibition of EGFR and MEK induced apoptosis and blocked both EGFR-RAS-RAF-MEK-ERK and EGFR-PI3K-AKT-RPS6 nodes simultaneously in RAS-active, but not RAS-inactive NSCLC. These results provide a comprehensive strategy to personalize treatment of NSCLC based on RAS network dysregulation and provide proof-of-concept of a genomic approach to classify and target complex signaling networks.
Collapse
Affiliation(s)
- Nader N El-Chaar
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| | - Stephen R Piccolo
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA; Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Kenneth M Boucher
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| | - Adam L Cohen
- Department of Medicine, Division of Oncology, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Medical School, Houston 77030, USA.
| | - Philip J Moos
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA.
| | - Andrea H Bild
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
171
|
Han JY, Kim SH, Lee YS, Lee SY, Hwang JA, Kim JY, Yoon SJ, Lee GK. Comparison of targeted next-generation sequencing with conventional sequencing for predicting the responsiveness to epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) therapy in never-smokers with lung adenocarcinoma. Lung Cancer 2014; 85:161-7. [PMID: 24857785 DOI: 10.1016/j.lungcan.2014.04.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/23/2014] [Accepted: 04/14/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE To investigate the clinical utility of targeted next-generation sequencing (NGS) for predicting the responsiveness to epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) therapy, we compared the efficacy with conventional sequencing in never-smokers with lung adenocarcinoma (NSLAs). PATIENTS AND METHODS We obtained DNA from 48 NSLAs who received gefitinib or erlotinib for their recurrent disease after surgery. Sanger sequencing and peptide nucleic acid clamp polymerase chain reaction (PCR) were used to analyze EGFR, KRAS, BRAF, and PIK3CA mutations. We analyzed ALK, RET, and ROS1 rearrangements by fluorescent in situ hybridization or reverse transcriptase-PCR and quantitative real-time PCR. After molecular screening, Ion Torrent NGS was performed in 31 cases harboring only EGFR exon 19 deletions (19DEL), an L858R mutation, or none of the above mutations. RESULTS The 31 samples were divided into four groups: (1) responders to EGFR-TKIs with only 19DEL or L858R (n=15); (2) primary resistance to EGFR-TKI with only 19DEL or L858R (n=4); (3) primary resistance to EGFR-TKI without any mutations (n=8); (4) responders to EGFR-TKI without any mutations (n=4). With NGS, all conventionally detected mutations were confirmed except for one L858R in group 2. Additional uncovered predictive mutations with NGS included one PIK3CA E542K in group 2, two KRAS (G12V and G12D), one PIK3CA E542K, one concomitant PIK3CA and EGFR L858R in group 3, and one EGFR 19DEL in group 4. CONCLUSIONS Targeted NGS provided a more accurate and clinically useful molecular classification of NSLAs. It may improve the efficacy of EGFR-TKI therapy in lung cancer.
Collapse
Affiliation(s)
- Ji-Youn Han
- Lung Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea.
| | - Sun Hye Kim
- Lung Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Yeon-Su Lee
- Functional Genomic Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | | | - Jung-Ah Hwang
- Functional Genomic Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Jin Young Kim
- Lung Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Sung Jin Yoon
- Lung Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Geon Kook Lee
- Lung Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
172
|
Mbogning C, Perdry H, Toussile W, Broët P. A novel tree-based procedure for deciphering the genomic spectrum of clinical disease entities. J Clin Bioinforma 2014; 4:6. [PMID: 24739673 PMCID: PMC4129184 DOI: 10.1186/2043-9113-4-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/08/2014] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Dissecting the genomic spectrum of clinical disease entities is a challenging task. Recursive partitioning (or classification trees) methods provide powerful tools for exploring complex interplay among genomic factors, with respect to a main factor, that can reveal hidden genomic patterns. To take confounding variables into account, the partially linear tree-based regression (PLTR) model has been recently published. It combines regression models and tree-based methodology. It is however computationally burdensome and not well suited for situations for which a large number of exploratory variables is expected. METHODS We developed a novel procedure that represents an alternative to the original PLTR procedure, and considered different selection criteria. A simulation study with different scenarios has been performed to compare the performances of the proposed procedure to the original PLTR strategy. RESULTS The proposed procedure with a Bayesian Information Criterion (BIC) achieved good performances to detect the hidden structure as compared to the original procedure. The novel procedure was used for analyzing patterns of copy-number alterations in lung adenocarcinomas, with respect to Kirsten Rat Sarcoma Viral Oncogene Homolog gene (KRAS) mutation status, while controlling for a cohort effect. Results highlight two subgroups of pure or nearly pure wild-type KRAS tumors with particular copy-number alteration patterns. CONCLUSIONS The proposed procedure with a BIC criterion represents a powerful and practical alternative to the original procedure. Our procedure performs well in a general framework and is simple to implement.
Collapse
Affiliation(s)
- Cyprien Mbogning
- Abirisk consortium WP4, 14-16 Avenue Paul-Vaillant-Couturier, 94807 Villejuif, France
- Inserm U669, 14-16 Avenue Paul-Vaillant-Couturier, 94807 Villejuif, France
| | - Hervé Perdry
- Inserm U669, 14-16 Avenue Paul-Vaillant-Couturier, 94807 Villejuif, France
- Faculty of Medicine Paris-Sud, 63 rue Gabriel Peri, 94276 Le Kremlin-Bicêtre, France
| | - Wilson Toussile
- Inserm U669, 14-16 Avenue Paul-Vaillant-Couturier, 94807 Villejuif, France
- Faculty of Medicine Paris-Sud, 63 rue Gabriel Peri, 94276 Le Kremlin-Bicêtre, France
| | - Philippe Broët
- Abirisk consortium WP4, 14-16 Avenue Paul-Vaillant-Couturier, 94807 Villejuif, France
- Inserm U669, 14-16 Avenue Paul-Vaillant-Couturier, 94807 Villejuif, France
- Faculty of Medicine Paris-Sud, 63 rue Gabriel Peri, 94276 Le Kremlin-Bicêtre, France
- Assistance Publique – Hôpitaux de Paris, Hôpital Paul Brousse, Villejuif, France
| |
Collapse
|
173
|
|
174
|
Kohno T, Tsuchihara K, Ogiwara H, Ichikawa H. RET and other genes: therapeutic targets in lung adenocarcinoma. Lung Cancer Manag 2014. [DOI: 10.2217/lmt.13.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY: The RET fusion gene was recently identified as a new druggable driver gene present in 1–2% of lung adenocarcinomas (LADCs). Vandetanib (ZD6474) and cabozantininb (XL184), two RET tyrosine kinase inhibitors approved by US FDA for the therapy of medullary thyroid cancer, have demonstrated therapeutic effectiveness in a few RET fusion-positive LADC patients. Several clinical trials are under way to address the therapeutic effects of RET tyrosine kinase inhibitors, including these two drugs. Multiplex diagnosis of aberrations in druggable driver oncogenes, such as EGFR, ALK, RET, ROS1, HER2/ERBB2, BRAF and others, in clinical samples will facilitate the design of personalized therapies for LADC based on protein kinase inhibitors. The development of therapeutic methods targeting aberrations of other genes, such as chromatin remodeling genes, is necessary to further improve the treatment of LADC.
Collapse
Affiliation(s)
- Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045 & 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Katsuya Tsuchihara
- Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045 & 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Hideaki Ogiwara
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hitoshi Ichikawa
- Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045 & 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| |
Collapse
|
175
|
Lennon FE, Salgia R. Mitochondrial dynamics: biology and therapy in lung cancer. Expert Opin Investig Drugs 2014; 23:675-92. [PMID: 24654596 DOI: 10.1517/13543784.2014.899350] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Lung cancer mortality rates remain at unacceptably high levels. Although mitochondrial dysfunction is a characteristic of most tumor types, mitochondrial dynamics are often overlooked. Altered rates of mitochondrial fission and fusion are observed in lung cancer and can influence metabolic function, proliferation and cell survival. AREAS COVERED In this review, the authors outline the mechanisms of mitochondrial fission and fusion. They also identify key regulatory proteins and highlight the roles of fission and fusion in metabolism and other cellular functions (e.g., proliferation, apoptosis) with an emphasis on lung cancer and the interaction with known cancer biomarkers. They also examine the current therapeutic strategies reported as altering mitochondrial dynamics and review emerging mitochondria-targeted therapies. EXPERT OPINION Mitochondrial dynamics are an attractive target for therapeutic intervention in lung cancer. Mitochondrial dysfunction, despite its molecular heterogeneity, is a common abnormality of lung cancer. Targeting mitochondrial dynamics can alter mitochondrial metabolism, and many current therapies already non-specifically affect mitochondrial dynamics. A better understanding of mitochondrial dynamics and their interaction with currently identified cancer 'drivers' such as Kirsten-Rat Sarcoma Viral Oncogene homolog will lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Frances E Lennon
- University of Chicago, Department of Medicine, Section of Hematology/Oncology , 5841 S. Maryland Avenue, MC 2115 Chicago, IL 60637 , USA +1 773 702 4399 ; +1 773 834 1798 ;
| | | |
Collapse
|
176
|
Guan YS, He Q, Li M. Icotinib: activity and clinical application in Chinese patients with lung cancer. Expert Opin Pharmacother 2014; 15:717-28. [PMID: 24588695 DOI: 10.1517/14656566.2014.890183] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Icotinib (BPI-2009H, Conmana) is a novel oral quinazoline compound that has proven survival benefit in Chinese patients with lung cancer, for which several therapies are currently available often with unsatisfactory results. Icotinib is the first self-developed small molecular drug in China for targeted therapy of lung cancer. AREAS COVERED The authors' experience in the clinical application of icotinib is reviewed in combination with related publications in the literature. Antitumor activities were observed in non-small-cell lung cancer and others in several recent studies. On 7 June 2011, icotinib was approved by the State Food and Drug Administration of China for the treatment of local advanced or metastatic non-small-cell lung cancer based on the results of a nationwide, of 27 centers, randomized, double-blind, double-modulated, parallel-controlled, Phase III trial with single agent icotinib in lung cancer patients after failure of chemotherapy. EXPERT OPINION Icotinib is a generic drug. Compared to the other two commercially available EGFR tyrosine kinase inhibitors, gefitinib and erlotinib, icotinib is similar to them in chemical structure, mechanism of activity and therapeutic effects but less expensive. Better safety as well as a wider therapeutic window has also been proven in several Chinese studies. Future studies on cost effectiveness are warranted.
Collapse
Affiliation(s)
- Yong-Song Guan
- West China Hospital of Sichuan University, Department of Oncology , Chengdu 610041 , China
| | | | | |
Collapse
|
177
|
Lower Ras expression as an independent predictor of patient outcomes in lung cancer treated with bevacizumab plus chemotherapy. Cancer Gene Ther 2014; 21:110-4. [PMID: 24577128 DOI: 10.1038/cgt.2014.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 01/26/2014] [Accepted: 01/28/2014] [Indexed: 01/22/2023]
Abstract
The objective of this study was to analyze the predictive roles of VEGF/KDR/Ras/MAPK gene expression in patients with advanced non-small-cell lung cancer (NSCLC) treated with bevacizumab plus chemotherapy. Twenty-five patients participating in an open-label phase IV trial (SAiL, MO19390) with available tumor tissues were analyzed. The mRNA expression levels of VEGF, kinase insert domain receptor (KDR), Ras, and mitogen-activated protein kinase (MAPK) in tumor tissues were detected using real-time quantitative PCR methods. The relationships between gene expression and disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) were assessed. Patients with lower Ras expression had a longer PFS and OS than patients with higher expression (median PFS, 9.9 vs 5.5 months, χ(2)=3.944, P=0.047; OS, 19.3 vs 7.1 months, χ(2)=9.384, P=0.002). The PFS and OS of patients with lower and higher MAPK expression exhibited a marginal and significant difference (median PFS, 9.9 vs 5.5 months, χ(2)=3.464, P=0.063; OS, 19.3 vs 9.7 months, χ(2)=5.298, P=0.021), respectively. Multivariate analyses using Cox's proportional hazards model showed that Ras is an independent predictor of OS (χ(2)=9.384, P=0.002). No differences in DCR were found according to Ras expression level. The results indicate that Ras is an independent predictor of OS. Thus, patients with lower Ras expression are most likely to benefit from bevacizumab plus chemotherapy treatment regimen. Patients with higher levels of Ras should receive other inhibitors that target Ras. The results also suggest that gene therapies that decrease RAS expression combined with bevacizumab may improve lung cancer treatment. Although there is a very important implication to patient selection in the target therapy, the data in this study are very preliminary owing to the too small sample size. Therefore, further research involving large numbers of patients and a prospective assessment of low and high RAS mRNA expressions getting the same treatments need to be done before conclusions can be made.
Collapse
|
178
|
Establishment and characterization of a lung cancer cell line, SMC-L001, from a lung adenocarcinoma. In Vitro Cell Dev Biol Anim 2014; 50:519-26. [PMID: 24569940 DOI: 10.1007/s11626-014-9736-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/21/2014] [Indexed: 01/22/2023]
Abstract
Lung cancer cell lines are a valuable tool for elucidating lung tumorigenesis and developing novel therapies. However, the majority of cell lines currently available were established from tumors in patients of Caucasian origin, limiting our ability to investigate how cancers in patients of different ethnicities differ from one another in terms of tumor biology and drug responses. In this study, we established a human non-small cell lung carcinoma cell line, SMC-L001, and characterized its genome and tumorigenic potential. SMC-L001 cells were isolated from a Korean lung adenocarcinoma patient (male, pStage IIb) and were propagated in culture. SMC-L001 cells were adherent. DNA fingerprinting analysis indicated that the SMC-L001 cell line originated from parental tumor tissue. Comparison of the genomic profile of the SMC-L001 cell line and the original tumor revealed an identical profile with 739 mutations in 46 cancer-related genes, including mutations in TP53 and KRAS. Furthermore, SMC-L001 cells were highly tumorigenic, as evidenced by the induction of solid tumors in immunodeficient mice. In summary, we established a new lung cancer cell line with point mutations in TP53 and KRAS from a Korean lung adenocarcinoma patient that will be useful for investigating ethnic differences in lung cancer biology and drug response.
Collapse
|
179
|
Gibault L, Cazes A, Narjoz C, Blons H. [Molecular profiling of non-small cell lung cancer]. REVUE DE PNEUMOLOGIE CLINIQUE 2014; 70:47-62. [PMID: 24566035 DOI: 10.1016/j.pneumo.2013.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 10/24/2013] [Accepted: 11/01/2013] [Indexed: 06/03/2023]
Abstract
The management of locally advanced and metastatic non-small cell lung cancer has been revolutionized thanks to recent progress in pathology and molecular biology. The first molecular subgroup is defined by activating mutations of the epidermal growth factor receptor (EGFR), and a dramatic response to specific tyrosine kinase inhibitors. Since then, multiple genetic alterations (KRAS, HER2, BRAF, PIK3CA, ALK, ROS, RET…) have been identified as potential target of novel therapies, and molecular profiling has become common practice. This review focus on the molecular alterations associated with non-small cell lung cancer, including molecular profiling and response to targeted therapies.
Collapse
Affiliation(s)
- L Gibault
- Service de pathologie, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France
| | - A Cazes
- Service de pathologie, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France; Université Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - C Narjoz
- Service de biochimie, UF de pharmacogénétique et oncologie moléculaire, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France
| | - H Blons
- Université Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France; Service de biochimie, UF de pharmacogénétique et oncologie moléculaire, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France; UMR-S775, Inserm, centre universitaire des Saints-Pères, 46, rue des Saints-Pères, 75006 Paris, France.
| |
Collapse
|
180
|
Liu H, Park J, Manning C, Goehlmann HW, Marshall DJ. Metastatic signature in lung cancer is associated with sensitivity to anti-integrin αVmonoclonal antibody intetumumab. Genes Chromosomes Cancer 2014; 53:349-57. [DOI: 10.1002/gcc.22145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 01/02/2014] [Accepted: 01/03/2014] [Indexed: 11/10/2022] Open
Affiliation(s)
- Huiqing Liu
- Janssen Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson; Spring House PA 19477 USA
| | - Jaehong Park
- Janssen Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson; Spring House PA 19477 USA
| | - Carol Manning
- Janssen Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson; Spring House PA 19477 USA
| | - Hinrich W.H. Goehlmann
- Janssen Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson; Turnhoutseweg 30 2340 Beerse Belgium
| | - Deborah J. Marshall
- Janssen Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson; Spring House PA 19477 USA
| |
Collapse
|
181
|
Bosutinib inhibits migration and invasion via ACK1 in KRAS mutant non-small cell lung cancer. Mol Cancer 2014; 13:13. [PMID: 24461128 PMCID: PMC3930897 DOI: 10.1186/1476-4598-13-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/21/2014] [Indexed: 01/13/2023] Open
Abstract
The advent of effective targeted therapeutics has led to increasing emphasis on precise biomarkers for accurate patient stratification. Here, we describe the role of ACK1, a non-receptor tyrosine kinase in abrogating migration and invasion in KRAS mutant lung adenocarcinoma. Bosutinib, which inhibits ACK1 at 2.7 nM IC50, was found to inhibit cell migration and invasion but not viability in a panel of non-small cell lung cancer (NSCLC) cell lines. Knockdown of ACK1 abrogated bosutinib-induced inhibition of cell migration and invasion specifically in KRAS mutant cells. This finding was further confirmed in an in vivo zebrafish metastatic model. Tissue microarray data on 210 Singaporean lung adenocarcinomas indicate that cytoplasmic ACK1 was significantly over-expressed relative to paired adjacent non-tumor tissue. Interestingly, ACK1 expression in “normal” tissue adjacent to tumour, but not tumour, was independently associated with poor overall and relapse-free survival. In conclusion, inhibition of ACK1 with bosutinib attenuates migration and invasion in the context of KRAS mutant NSCLC and may fulfil a therapeutic niche through combinatorial treatment approaches.
Collapse
|
182
|
Chung HW, Lee KY, Kim HJ, Kim WS, So Y. FDG PET/CT metabolic tumor volume and total lesion glycolysis predict prognosis in patients with advanced lung adenocarcinoma. J Cancer Res Clin Oncol 2014; 140:89-98. [PMID: 24194352 DOI: 10.1007/s00432-013-1545-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 10/23/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE We investigated fluorine-18 fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT)-assessed metabolic tumor volume (MTV) and total lesion glycolysis (TLG) as prognostic factors in lung adenocarcinoma patients. METHODS This retrospective study included 106 patients (19 stage I/II and 87 stage III/IV lung adenocarcinoma) who underwent FDG PET/CT before treatment. Standardized uptake value (SUV), MTV, and TLG (MTV × mean SUV) of each malignant lesion were measured. Whole MTV and whole TLG were the summation of all the MTV and TLG values in each patient. Survival analysis and FDG PET/CT parameters regarding epidermal growth factor receptor (EGFR) gene mutation status were evaluated. RESULTS Univariate survival analysis of stage III/IV patients identified high whole MTV (≥90), high whole TLG (≥600), and stage IV as significant predictors of poor progression-free survival. For overall survival, high whole MTV (≥90), high whole TLG (≥600), EGFR mutation-negative, and stage IV were significant poor prognostic predictors. After multivariate survival analysis, high whole MTV (P = 0.001), high whole TLG (P = 0.027), and stage IV (P = 0.006) were independent predictors of poor progression-free survival. High whole MTV (P < 0.001), high whole TLG (P = 0.001), and EGFR mutation-negative (P = 0.001) were independent prognostic predictors for poor overall survival. In a survival analysis of stage I/II patients, none was an independent prognostic predictor. No significant differences were found in FDG PET/CT parameters for EGFR mutation-negative and EGFR mutation-positive patients. CONCLUSIONS Assessment of MTV and TLG by FDG PET/CT in advanced lung adenocarcinoma patients provides useful information regarding prognosis.
Collapse
Affiliation(s)
- Hyun Woo Chung
- Department of Nuclear Medicine, Konkuk University Medical Center, Research Institute of Biomedical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Hwayang-dong, Gwangjin-gu, Seoul, 143-729, Korea,
| | | | | | | | | |
Collapse
|
183
|
Hensing T, Chawla A, Batra R, Salgia R. A personalized treatment for lung cancer: molecular pathways, targeted therapies, and genomic characterization. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 799:85-117. [PMID: 24292963 DOI: 10.1007/978-1-4614-8778-4_5] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lung cancer is a heterogeneous, complex, and challenging disease to treat. With the arrival of genotyping and genomic profiling, our simple binary division of lung cancer into non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC) is no longer acceptable. In the past decade and with the advent of personalized medicine, multiple advances have been made in understanding the underlying biology and molecular mechanisms of lung cancer. Lung cancer is no longer considered a single disease entity and is now being subdivided into molecular subtypes with dedicated targeted and chemotherapeutic strategies. The concept of using information from a patient's tumor to make therapeutic and treatment decisions has revolutionized the landscape for cancer care and research in general.Management of non-small-cell lung cancer, in particular, has seen several of these advances, with the understanding of activating mutations in EGFR, fusion genes involving ALK, rearrangements in ROS-1, and ongoing research in targeted therapies for K-RAS and MET. The next era of personalized treatment for lung cancer will involve a comprehensive genomic characterization of adenocarcinoma, squamous-cell carcinoma, and small-cell carcinoma into various subtypes. Future directions will involve incorporation of molecular characteristics and next generation sequencing into screening strategies to improve early detection, while also having applications for joint treatment decision making in the clinics with patients and practitioners. Personalization of therapy will involve close collaboration between the laboratory and the clinic. Given the heterogeneity and complexity of lung cancer treatment with respect to histology, tumor stage, and genomic characterization, mind mapping has been developed as one of many tools which can assist physicians in this era of personalized medicine. We attempt to utilize the above tool throughout this chapter, while reviewing lung cancer epidemiology, lung cancer treatment, and the genomic characterization of lung cancer.
Collapse
Affiliation(s)
- Thomas Hensing
- Department of Medicine, Section of Hematology/Oncology, NorthShore University Health System, Kellogg Cancer Center, 2650 Ridge Avenue, Evanston, IL, 60201, USA,
| | | | | | | |
Collapse
|
184
|
Reungwetwattana T, Dy GK. Targeted therapies in development for non-small cell lung cancer. J Carcinog 2013; 12:22. [PMID: 24574860 PMCID: PMC3927069 DOI: 10.4103/1477-3163.123972] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/15/2013] [Indexed: 12/11/2022] Open
Abstract
The iterative discovery in various malignancies during the past decades that a number of aberrant tumorigenic processes and signal transduction pathways are mediated by "druggable" protein kinases has led to a revolutionary change in drug development. In non-small cell lung cancer (NSCLC), the ErbB family of receptors (e.g., EGFR [epidermal growth factor receptor], HER2 [human epidermal growth factor receptor 2]), RAS (rat sarcoma gene), BRAF (v-raf murine sarcoma viral oncogene homolog B1), MAPK (mitogen-activated protein kinase) c-MET (c-mesenchymal-epithelial transition), FGFR (fibroblast growth factor receptor), DDR2 (discoidin domain receptor 2), PIK3CA (phosphatidylinositol-4,5-bisphosphate3-kinase, catalytic subunit alpha)), PTEN (phosphatase and tensin homolog), AKT (protein kinase B), ALK (anaplastic lym phoma kinase), RET (rearranged during transfection), ROS1 (reactive oxygen species 1) and EPH (erythropoietin-producing hepatoma) are key targets of various agents currently in clinical development. These oncogenic targets exert their selective growth advantage through various intercommunicating pathways, such as through RAS/RAF/MEK, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin and SRC-signal transduction and transcription signaling. The recent clinical studies, EGFR tyrosine kinase inhibitors and crizotinib were considered as strongly effective targeted therapies in metastatic NSCLC. Currently, five molecular targeted agents were approved for treatment of advanced NSCLC: Gefitinib, erlotinib and afatinib for positive EGFR mutation, crizotinib for positive echinoderm microtubule-associated protein-like 4 (EML4)-ALK translocation and bevacizumab. Moreover, oncogenic mutant proteins are subject to regulation by protein trafficking pathways, specifically through the heat shock protein 90 system. Drug combinations affecting various nodes in these signaling and intracellular processes are predicted and demonstrated to be synergistic and advantageous in overcoming treatment resistance compared with monotherapy approaches. Understanding the role of the tumor microenvironment in the development and maintenance of the malignant phenotype provided additional therapeutic approaches as well. More recently, improved knowledge on tumor immunology has set the stage for promising immunotherapies in NSCLC. This review will focus on the rationale for the development of targeted therapies in NSCLC and the various strategies employed in preventing or overcoming the inevitable occurrence of treatment resistance.
Collapse
Affiliation(s)
- Thanyanan Reungwetwattana
- Department of Internal Medicine, Division of Medical Oncology, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Grace Kho Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
185
|
Sun W, Sun Y, Zhu M, Wang Z, Zhang H, Xin Y, Jiang G, Guo X, Zhang Z, Liu Y. The role of plasma cell-free DNA detection in predicting preoperative chemoradiotherapy response in rectal cancer patients. Oncol Rep 2013; 31:1466-72. [PMID: 24378613 DOI: 10.3892/or.2013.2949] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 11/27/2013] [Indexed: 11/05/2022] Open
Abstract
In the present study, we studied the relationship between plasma cell-free DNA and the effect of preoperative chemoradiotherapy in patients with rectal cancer. The concentration, KRAS mutation and O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status of cell-free DNA were measured by using polymerase chain reaction (PCR) analyses. The response to chemoradiotherapy was assessed using tumor regression grading (TRG) scores. The cell-free DNA concentrations in patients with rectal cancer (n=34) were significantly higher compared to healthy controls (n=10). The 400-base pair (bp) DNA concentration, 400-/100-bp DNA ratio decreased significantly after chemoradiotherapy in the good response group. The incidence of KRAS mutation decreased significantly after chemoradiotherapy in both good and poor response groups. Higher MGMT promoter methylation status at baseline DNA was associated with a better tumor response. Therefore, cell-free DNA detection may be useful in evaluating the effect of preoperative chemoradiotherapy in patients with rectal cancer.
Collapse
Affiliation(s)
- W Sun
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Y Sun
- Cancer Research Institute of Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - M Zhu
- Cancer Research Institute of Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Z Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - H Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Y Xin
- Cancer Research Institute of Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - G Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - X Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Z Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Y Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| |
Collapse
|
186
|
New treatment options for lung adenocarcinoma--in view of molecular background. Pathol Oncol Res 2013; 20:11-25. [PMID: 24306880 DOI: 10.1007/s12253-013-9719-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 10/29/2013] [Indexed: 12/22/2022]
Abstract
Lung cancer is the leading cause of cancer related mortality all over the world, and a number of developments have indicated future clinical benefit recently. The development of molecular pathology methods has become increasingly important in the prediction of chemotherapy sensitivity and mutation analysis to identify driver mutations as important targets of new therapeutic agents. The most significant changes in the treatment of NSCLC revealed in new pathologic classification and in the introduction of molecularly targeted therapies, which include monoclonal antibodies and small molecule tyrosine kinase inhibitors. The side effects of these agents are generally better tolerated than those of conventional chemotherapy and show higher efficacy. The most important factor follows: histology subtypes, gene mutation status, patients' selection, drug toxicities and occurence of drug resistance. In the advanced disease, the hope of cure is less than 3%, but improvements in survival have been clearly achieved. Some years ago the median lung cancer survival rate was 10-12 months, now in case of available specific molecular targets, a significant increase in median survival rates to 24-36 months has been achieved. These agents give an opportunity to provide a new standard of care. Therefore testing EGFR mutations and ALK rearrangements in patients with advanced lung adenocarcinoma should be incorporated into routine clinical practice. This review focuses on the rationale for targeted agents and new treatment possibilities in case of advanced lung adenocarcinoma.
Collapse
|
187
|
Gadgeel SM, Bepler G. Prognostic and predictive markers for personalized adjuvant therapy for non-small-cell lung cancer patients. Future Oncol 2013; 9:1909-21. [DOI: 10.2217/fon.13.160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adjuvant chemotherapy following surgical resection in non-small-cell lung cancer patients with large tumors (>4 cm) or tumors with lymph node metastases has emerged as a standard of care. The benefits of adjuvant chemotherapy are modest, with a 5-year survival improvement of only 5–10%. In addition, survival rates of 25% or greater with surgery alone suggest that some patients do not need adjuvant therapy. Therefore, there is a need to develop prognostic and predictive markers to identify patients in need of adjuvant therapy and the patients likely to benefit from such therapy. Many factors have been evaluated for this purpose and some of these factors, such as visceral pleural invasion, can influence the decision to use adjuvant chemotherapy. However, most of the available data are retrospective, which limits the utility of these markers in current practice. Ongoing trials are evaluating many promising markers and may guide adjuvant therapy in the future.
Collapse
Affiliation(s)
- Shirish M Gadgeel
- Department of Oncology, Wayne State University & Molecular Theapeutics Program, Karmanos Cancer Institute, 4100 John R, Mailcode: HW02EA, Detroit, MI 48201, USA
| | - Gerold Bepler
- Department of Oncology, Wayne State University & Molecular Theapeutics Program, Karmanos Cancer Institute, 4100 John R, Mailcode: HW02EA, Detroit, MI 48201, USA
| |
Collapse
|
188
|
Zhou W, Yue C, Deng J, Hu R, Xu J, Feng L, Lan Q, Zhang W, Ji D, Wu J, Liu Q, Liu A. Autophagic protein Beclin 1 serves as an independent positive prognostic biomarker for non-small cell lung cancer. PLoS One 2013; 8:e80338. [PMID: 24260370 PMCID: PMC3829868 DOI: 10.1371/journal.pone.0080338] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 10/02/2013] [Indexed: 12/19/2022] Open
Abstract
Beclin 1, a key regulator of autophagy, has been found to be aberrantly expressed in a variety of human malignancies. Herein, we employed immunohistochemistry (IHC) to detect the protein expression of Beclin 1 in non-small cell lung cancer (NSCLC) and paired normal adjacent lung tissues, and analyzed its clinicopathological/prognostic significance in NSCLC. Receiver operating characteristic (ROC) curve analysis was utilized to determine a cutoff point (>2 VS. ≤2) for Beclin 1 expression in a training set (n = 105). For validation, the ROC-derived cutoff value was subjected to analysis of the association of Beclin 1 with patients’ clinical characteristics and outcome in a testing set (n = 111) and the overall patient cohort (n = 216). Our data showed that Beclin 1 was significantly lower in NSCLC tissues compared with the adjacent normal tissues, negatively associating with tumor recurrence rate (65.8% VS 32.3%; p < 0.001). In the testing set and the overall patient cohort, low expression of Beclin 1 showed significantly inferior overall survival (OS) (p < 0.001) and progression-free survival (PFS) (p < 0.001) compared to high expression of Beclin 1. In the testing set and the overall patient cohort, the median duration of OS for patients with high and low expression of Beclin 1 was 108 VS. 24.5 months (p < 0.001) and 108 VS. 28 months (p < 0.001), respectively. Furthermore, low expression of Beclin 1 was also a poor prognostic factor within each stage of NSCLC patients. Multivariate analysis identified that Beclin 1 was an independent prognostic factor for NSCLC. Our findings in the present study provided evidence that Beclin 1 may thus emerge as an independent prognostic biomarker in this tumor entity in the future.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Caifeng Yue
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Jinyun Deng
- Department of the Sixth Internal Medicine, Jiangxi Province Cancer Hospital, Nanchang, China
| | - Ronghuan Hu
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jie Xu
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Long Feng
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Qiongyu Lan
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Wenfeng Zhang
- Department of Infectious Disease, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Dexiang Ji
- Department of Hematology, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jianbing Wu
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Quentin Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (AWL); (QTL)
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
- * E-mail: (AWL); (QTL)
| |
Collapse
|
189
|
Abstract
Lung cancer remains the most common cause of cancer-related death in the United States. At presentation, the majority of patients have regional or systemic metastases and therefore require systemic therapy. For years, chemotherapy was the only systemic therapy option. A major paradigm shift has occurred in recent years with the identification of driver genetic alterations in some non-small cell lung cancers (NSCLCs). It is part of current standard of care to assess epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) translocations in tumors of patients with advanced NSCLC. Drugs targeting these mutations provide significant clinical benefit and are the preferred therapeutic option in these patients. Ongoing clinical trials are assessing the clinical benefit from targeting other driver genetic alterations. Further therapeutic targets have been identified through greater understanding of the variety of molecular processes that facilitate tumor formation and progression. Some of these new therapeutic targets are heat shock proteins and targets that can allow enhanced anti-tumor immune response. It is expected that these advances will allow personalized management of NSCLC patients and move us away from approaching all NSCLC patients with the same therapeutic tools.
Collapse
|
190
|
Genetic alterations defining NSCLC subtypes and their therapeutic implications. Lung Cancer 2013; 82:179-89. [PMID: 24011633 DOI: 10.1016/j.lungcan.2013.07.025] [Citation(s) in RCA: 260] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/20/2013] [Accepted: 07/29/2013] [Indexed: 01/03/2023]
Abstract
Lung cancer is the leading cause of cancer death worldwide, accounting for more deaths than breast, prostate and colon cancer combined. While treatment decisions are determined primarily by stage, therapeutically non small cell lung cancer (NSCLC) has traditionally been treated as a single disease. However, recent findings have led to the recognition of histology and molecular subtypes as important determinants in treatment selection. Identifying the genetic differences that define these molecular and histological subtypes has the potential to impact treatment and as such is currently the focus of much research. Microarray and genomic sequencing efforts have provided unparalleled insight into the genomes of lung cancer subtypes, specifically adenocarcinoma (AC) and squamous cell carcinoma (SqCC), revealing subtype specific genomic alterations and molecular subtypes as well as differences in cell signaling pathways. In this review, we discuss the recurrent genomic alterations characteristic of AC and SqCC (including molecular subtypes), their therapeutic implications and emerging clinical practices aimed at tailoring treatments based on a tumor's molecular alterations with the hope of improving patient response and survival.
Collapse
|
191
|
Zaorsky NG, Sun Y, Wang Z, Palmer J, Fortina PM, Solomides C, Werner-Wasik M, Dicker AP, Axelrod R, Campling B, Evans N, Cowan S, Lu B. Identification of a KRAS mutation in a patient with non-small cell lung cancer treated with chemoradiotherapy and panitumumab. Cancer Biol Ther 2013; 14:883-7. [PMID: 23917487 DOI: 10.4161/cbt.25942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RTOG 0839 is a Phase II study of pre-operative chemoradiotherapy with or without panitumumab in potentially operable locally advanced non-small cell lung cancer (NSCLC). The investigational agent, panitumumab, is an anti-epithelial growth factor receptor (EGFR) antibody that improves progression-free survival in chemorefractory metastatic colorectal cancer (mCRC). Recently, both KRAS mutational status (i.e., mutated or not) and subtype (i.e., activating or inactivating) have been shown to be predictive of response to anti-EGFR therapy in mCRC. However, in NSCLC, it is unknown if KRAS mutational status or subtype predict benefit to anti-EGFR therapies because of unique genetic and epigenetic factors unique to each cancer. We present a patient with stage III NSCLC containing a KRAS G12D activating mutation who had a partial pathologic response, with disappearance of a minor KRAS mutant clone. This case suggests possible eradication of the G12D KRAS lung cancer clones by concurrent chemoradiation with panitumumab.
Collapse
Affiliation(s)
- Nicholas G Zaorsky
- Department of Radiation Oncology; Fox Chase Cancer Center; Philadelphia, PA USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Garassino MC, Martelli O, Broggini M, Farina G, Veronese S, Rulli E, Bianchi F, Bettini A, Longo F, Moscetti L, Tomirotti M, Marabese M, Ganzinelli M, Lauricella C, Labianca R, Floriani I, Giaccone G, Torri V, Scanni A, Marsoni S. Erlotinib versus docetaxel as second-line treatment of patients with advanced non-small-cell lung cancer and wild-type EGFR tumours (TAILOR): a randomised controlled trial. Lancet Oncol 2013; 14:981-8. [PMID: 23883922 DOI: 10.1016/s1470-2045(13)70310-3] [Citation(s) in RCA: 402] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Erlotinib is registered for treatment of all patients with advanced non-small-cell lung cancer (NSCLC). However, its efficacy for treatment of patients whose tumours are EGFR wild-type-which includes most patients-is still contentious. We assessed the efficacy of erlotinib compared with a standard second-line chemotherapy in such patients. METHODS We did this randomised controlled trial in 52 Italian hospitals. We enrolled patients who had metastatic NSCLC, had had platinum-based chemotherapy, and had wild-type EGFR as assessed by direct sequencing. Patients were randomly assigned centrally (1:1) to receive either erlotinib orally 150 mg/day or docetaxel intravenously 75 mg/m(2) every 21 days or 35 mg/m(2) on days 1, 8, and 15, every 28 days. Randomisation was stratified by centre, stage, type of first-line chemotherapy, and performance status. Patients and investigators who gave treatments or assessed outcomes were not masked to treatment allocation, investigators who analysed results were. The primary endpoint was overall survival in the intention-to-treat population. The study is registered at ClinicalTrials.gov, number NCT00637910. FINDINGS We screened 702 patients, of whom we genotyped 540. 222 patients were enrolled (110 assigned to docetaxel vs 112 assigned to erlotinib). Median overall survival was 8·2 months (95% CI 5·8-10·9) with docetaxel versus 5·4 months (4·5-6·8) with erlotinib (adjusted hazard ratio [HR] 0·73, 95% CI 0·53-1·00; p=0·05). Progression-free survival was significantly better with docetaxel than with erlotinib: median progression-free survival was 2·9 months (95% CI 2·4-3·8) with docetaxel versus 2·4 months (2·1-2·6) with erlotinib (adjusted HR 0·71, 95% CI 0·53-0·95; p=0·02). The most common grade 3-4 toxic effects were: low absolute neutrophil count (21 [20%] of 104 in the docetaxel group vs none of 107 in the erlotinib group), skin toxic effects (none vs 15 [14%]), and asthenia (ten [10%] vs six [6%]). INTERPRETATION Our results show that chemotherapy is more effective than erlotinib for second-line treatment for previously treated patients with NSCLC who have wild-type EGFR tumours.
Collapse
|
193
|
Farhat FS, Houhou W. Targeted therapies in non-small cell lung carcinoma: what have we achieved so far? Ther Adv Med Oncol 2013; 5:249-70. [PMID: 23858333 PMCID: PMC3707340 DOI: 10.1177/1758834013492001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The search for innovative therapeutic agents in non-small cell lung cancer (NSCLC) has witnessed a swift evolution. The number of targeted drugs that can improve patient outcomes with an acceptable safety profile is steadily increasing. In this review, we highlight current drugs that have already been approved or are under evaluation for the treatment of patients with NSCLC, either in monotherapy or combined therapy for both the first- and second-line settings. Experience with drugs targeting the vascular endothelial growth factor and its receptor, as well as the epidermal growth factor receptor is summarized. Moreover, we provide an overview of more novel targets in NSCLC and initial experience with the respective therapeutic agents.
Collapse
Affiliation(s)
- Fadi S Farhat
- Hematology-Oncology Division (Head), Hammoud Hospital University Medical Center, Ghassan Hammoud Street, 652, Saida, Lebanon
| | | |
Collapse
|
194
|
Ou SHI. Lung cancer in never-smokers. Does smoking history matter in the era of molecular diagnostics and targeted therapy? J Clin Pathol 2013; 66:839-46. [PMID: 23661716 DOI: 10.1136/jclinpath-2012-201296] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer in never-smokers was recognised as a distinct clinical entity around the mid-2000s because these patients tended to be Asian women and diagnosed at a younger age with a preponderance of adenocarcinoma and better survival outcome despite a more advanced stage of presentation. It was soon discovered that lung cancer in never-smokers had a higher prevalence of activating EGFR mutations and we tend to classify lung cancer by smoking status for screening purpose. With the discoveries of many actionable driver mutations such as activating EGFR mutations and ALK rearrangement in adenocarcinoma of the lung we have switched to classifying non-small cell lung cancer into different individual molecular subgroups based on the presence of a dominant driver mutation. Although many actionable driver mutations are found in never-smokers with adenocarcinoma, this review will summarise that a substantial proportion of patients with these actionable driver mutations had a previous smoking history. Alternatively among the driver mutations that are associated with smoking history, a fair amount of these patients were never-smokers. Thus smoking status should not be used as a screen strategy for identifying driver mutations in clinical practice. Finally smoking history may have predictive and/or prognostic significance within individual molecular subgroups and identifying the difference according to smoking history may help optimise future targeted therapy.
Collapse
|