1
|
Lv X, Li Y, Wang B, Wang Y, Xu Z, Hou D. Multisequence MRI-based radiomics signature as potential biomarkers for differentiating KRAS mutations in non-small cell lung cancer with brain metastases. Eur J Radiol Open 2024; 12:100548. [PMID: 38298532 PMCID: PMC10827674 DOI: 10.1016/j.ejro.2024.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Background Kirsten rat sarcoma virus (KRAS) has evolved from a genotype with predictive value to a therapeutic target recently. The study aimed to establish non-invasive radiomics models based on MRI to discriminate KRAS from epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) mutations in lung cancer patients with brain metastases (BM), then further explore the optimal sequence for prediction. Methods This retrospective study involved 317 patients (218 patients in training cohort and 99 patients in testing cohort) who had confirmed of KRAS, EGFR or ALK mutations. Radiomics features were separately extracted from T2WI, T2 fluid-attenuated inversion recovery (T2-FLAIR), diffusion weighted imaging (DWI) and contrast-enhanced T1-weighted imaging (T1-CE) sequences. The maximal information coefficient and recursive feature elimination method were used to select informative features. Then we built four radiomics models for differentiating KRAS from EGFR or ALK using random forest classifier. ROC curves were used to validate the capability of the models. Results The four radiomics models for discriminating KRAS from EGFR all worked well, especially DWI and T2WI models (AUCs: 0.942, 0.942 in training cohort, 0.949, 0.954 in testing cohort). When KRAS compared to ALK, DWI and T2-FLAIR models showed excellent performance in two cohorts (AUCs: 0.947, 0.917 in training cohort, 0.850, 0.824 in testing cohort). Conclusions Radiomics classifiers integrating MRI have potential to discriminate KRAS from EGFR or ALK, which are helpful to guide treatment and facilitate the discovery of new approaches capable of achieving this long-sought goal of cure in lung cancer patients with KRAS.
Collapse
Affiliation(s)
- Xinna Lv
- Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Ye Li
- Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Bing Wang
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Yichuan Wang
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Zexuan Xu
- Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Dailun Hou
- Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| |
Collapse
|
2
|
Zhang Y, Yan S, Li Y, Zhang J, Luo Y, Li P, Yang Y, Li Y, Huang Y, Wang E. Inhibin βA is an independent prognostic factor that promotes invasion via Hippo signaling in non‑small cell lung cancer. Mol Med Rep 2021; 24:789. [PMID: 34505633 PMCID: PMC8441965 DOI: 10.3892/mmr.2021.12429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/11/2021] [Indexed: 12/22/2022] Open
Abstract
Inhibin βA (INHBA) serves a prognostic and tumor-promoting role in numerous types of cancer. The present study aimed to determine the clinical significance of INHBA in non-small cell lung cancer (NSCLC) and the mechanisms underlying its potential tumor-promoting effect. INHBA expression was detected in clinical NSCLC samples using immunohistochemistry. In vivo loss- and gain-of-function studies were performed to determine the effects of INHBA on NSCLC invasion. In addition, protein and mRNA expression levels of INHBA, yes-associated protein (YAP), large tumor suppressor 1/2 kinase (LATS1/2), connective tissue growth factor, cysteine rich angiogenic inducer 61 and Merlin were assessed using western blotting and reverse transcription-quantitative PCR, respectively, to investigate the mechanism by which INHBA may affect the invasion of NSCLC. The present study revealed that INHBA was significantly upregulated in 238 clinical NSCLC samples compared with its expression levels in paired adjacent non-cancerous tissues, and in metastatic nodules compared with in primary tumors. Notably, high INHBA expression was statistically associated with clinicopathological features, including poor differentiation and advanced tumor stage. INHBA positivity was statistically related to decreased 5-year overall survival, for which INHBA was an independent prognostic factor. Furthermore, INHBA promoted NSCLC invasion in vitro. In NSCLC, INHBA expression was associated with the nuclear levels of YAP and INHBA overexpression enhanced the invasive abilities of NSCLC cells via inhibiting the Hippo pathway. Mechanistically, INHBA inhibited l LATS1/2 phosphorylation and induced YAP nuclear translocation by downregulating the protein expression levels of Merlin. In conclusion, INHBA may negatively regulate the Hippo pathway to act as a tumor promotor, and could represent a marker of prognosis in NSCLC.
Collapse
Affiliation(s)
- Yijun Zhang
- Department of Pathology, The First Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110010, P.R. China
| | - Shumei Yan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Yan Li
- Department of Radiotherapy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jiangbo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Yuan Luo
- Department of Pathology, The First Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110010, P.R. China
| | - Pengcheng Li
- Department of Pathology, The First Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110010, P.R. China
| | - Yuanzhong Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Yong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Yuhua Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Enhua Wang
- Department of Pathology, The First Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110010, P.R. China
| |
Collapse
|
3
|
Liver X receptor agonist T0901317 inhibits the migration and invasion of non-small-cell lung cancer cells in vivo and in vitro. Anticancer Drugs 2020; 30:495-500. [PMID: 30724772 PMCID: PMC6485493 DOI: 10.1097/cad.0000000000000758] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Liver X receptors are recognized as important regulators of cholesterol, fatty acid metabolism, inflammatory responses, and glucose homeostasis. The antineoplastic properties of synthetic liver X receptor (LXR) agonists (T0901317 and GW3965) have been reported in human carcinomas. Epidermal growth factor tyrosine kinase inhibitor (EGFR-TKI) is a first-line treatment for non-small-cell lung cancer patients with EGFR mutations. We used scratch and transwell assays to analyze cell migration and invasion. We evaluated tumor migration and invasion in vitro using a fluorescent orthotopic lung cancer model. An MMP9 (mouse) enzyme-linked immunosorbent assay kit was used to measure serum MMP9 concentrations. Protein expression was identified by western blot assays. In this study, we determined the effects of T0901317 and/or an EGFR-TKI on the lung cancer cell lines A549 and HCC827-8-1 in vitro and in vivo. We confirmed that the combination of the LXR agonist T0901317 and gefitinib can inhibit the migration and invasion of lung cancer both in vivo and in vitro, and this effect was possibly achieved by the inhibition of the ERK/MAPK signaling pathway. Our study showed that the combination of the LXR agonist T0901317 and gefitinib can inhibit the migration and invasion of lung cancer both in vivo and in vitro.
Collapse
|
4
|
Hopkins AM, Menz BD, Wiese MD, Kichenadasse G, Gurney H, McKinnon RA, Rowland A, Sorich MJ. Nuances to precision dosing strategies of targeted cancer medicines. Pharmacol Res Perspect 2020; 8:e00625. [PMID: 32662214 PMCID: PMC7358594 DOI: 10.1002/prp2.625] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 01/05/2023] Open
Abstract
Selecting the dose of a targeted cancer medicine that is most appropriate for a specific individual is a rational approach to maximize therapeutic outcomes and minimize toxicity. There are many different options for optimizing the dose of targeted cancer medicines and the purpose of this review is to provide a comprehensive comparison of the main options explored in prospective studies. Precision initial dose selection of targeted cancer therapies has been minimally explored to date; however, concentration, toxicity, and therapeutic outcome markers are used to guide on-therapy dose adaption of targeted cancer therapies across several medicines and cancers. While a specific concentration, toxicity, or therapeutic outcome marker commonly dominates an investigated precision on-therapy dose adaption strategy, greater attention to simultaneously account for exposure, toxicity, therapeutic outcomes, disease status, time since treatment initiation and patient preferences are required for optimal patient outcomes. To enable successful implementation of precision dosing strategies for targeted cancer medicines into clinical practice, future prospective studies aiming to develop strategies should consider these elements in their design.
Collapse
Affiliation(s)
- Ashley M. Hopkins
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Bradley D. Menz
- Division of PharmacySouthern Adelaide Local Health Network, Flinders Medical CentreAdelaideSouth AustraliaAustralia
| | - Michael D. Wiese
- School of Pharmacy and Medical SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Ganessan Kichenadasse
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Howard Gurney
- Department of Medical OncologyWestmead HospitalSydneyNew South WalesAustralia
| | - Ross A. McKinnon
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Michael J. Sorich
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
5
|
Yang G, Xu H, Yang L, Xu F, Zhang S, Yang Y, Wang Y. Apatinib in combination with pemetrexed-platinum chemotherapy for chemo-naive non-squamous non-small cell lung cancer: a phase II clinical study. Lung Cancer 2020; 147:229-236. [PMID: 32739743 DOI: 10.1016/j.lungcan.2020.07.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/08/2020] [Accepted: 07/19/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Apatinib showed efficacy in non-small cell lung cancer (NSCLC). We conducted this phase II clinical study to assess the efficacy and safety of apatinib in combination with pemetrexed-platinum chemotherapy in non-squamous NSCLC (Clinical Trial Registration: ChiCTR1800015920). MATERIALS AND METHODS Patients received oral apatinib (250 mg/d) with intravenous pemetrexed (500 mg/m2)-platinum (carboplatin AUC = 5 or cisplatin 75 mg/m2) chemotherapy every 21 days for 6 treatment cycles, and then maintained with apatinib 250 mg/d until progressive disease or intolerable toxicity. The primary endpoint was the objective response rate (ORR). The secondary endpoints included the progression-free survival (PFS), disease control rate (DCR), overall survival (OS) and safety. RESULTS Twenty advanced and chemo-naive non-squamous NSCLC patients were enrolled and evaluated. The ORR and DCR was 80% and 100%, respectively. The median PFS (mPFS) and median OS (mOS) for total patients was 7.7 (95%CI: 3.1-12.3) and 20.1 (95%CI: not available, NA) months. In the TKI-pretreated and treatment-naive subgroup, the ORR was 90% vs.70%, and the mPFS was 8.9 (95%CI: 5.5-12.3) vs.5.7 (95%CI: 0.1-11.3) months, respectively (P = 0.433). The mPFS in the responders without central nervous system (CNS) metastasis at baseline was 10.0 (95%CI: 6.1-13.9) months, and it was 3.8 (95%CI: 0.9-6.7) months in those with presence of CNS metastasis at baseline (P = 0.041, HR = 0.283, 95%CI: 0.084-0.948). Toxicities mainly included grade I-II hand-foot syndrome, hypertension, proteinuria and myelosuppression. CONCLUSION Apatinib in combination with pemetrexed-platinum chemotherapy showed good efficacy and tolerable toxicity in advanced non-squamous NSCLC, especially for those who failed to prior TKI targeted therapies.
Collapse
Affiliation(s)
- Guangjian Yang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haiyan Xu
- Department of Comprehensive Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lu Yang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fei Xu
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuyang Zhang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yaning Yang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
6
|
Raquel-Cunha A, Cardoso-Carneiro D, Reis RM, Martinho O. Current Status of Raf Kinase Inhibitor Protein (RKIP) in Lung Cancer: Behind RTK Signaling. Cells 2019; 8:cells8050442. [PMID: 31083461 PMCID: PMC6562953 DOI: 10.3390/cells8050442] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the most deadly neoplasm with the highest incidence in both genders, with non-small cell lung cancer (NSCLC) being the most frequent subtype. Somatic mutations within the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene are key drivers of NSCLC progression, with EGFR inhibitors being particularly beneficial for patients carrying the so-called “EGFR-sensitizing mutations”. However, patients eventually acquire resistance to these EGFR inhibitors, and a better knowledge of other driven and targetable proteins will allow the design of increasingly accurate drugs against patients’ specific molecular aberrations. Raf kinase inhibitory protein (RKIP) is an important modulator of relevant intracellular signaling pathways, including those controlled by EGFR, such as MAPK. It has been reported that it has metastasis suppressor activity and a prognostic role in several solid tumors, including lung cancer. In the present review, the potential use of RKIP in the clinic as a prognostic biomarker and predictor of therapy response in lung cancer is addressed.
Collapse
Affiliation(s)
- Ana Raquel-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3Bs-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal.
| | - Diana Cardoso-Carneiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3Bs-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal.
| | - Rui M Reis
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3Bs-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal.
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil.
| | - Olga Martinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3Bs-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal.
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil.
| |
Collapse
|
7
|
Ma JT, Sun J, Sun L, Zhang SL, Huang LT, Han CB. Efficacy and safety of apatinib in patients with advanced nonsmall cell lung cancer that failed prior chemotherapy or EGFR-TKIs: A pooled analysis. Medicine (Baltimore) 2018; 97:e12083. [PMID: 30170427 PMCID: PMC6392903 DOI: 10.1097/md.0000000000012083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/02/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Apatinib is a tyrosine kinase inhibitor (TKI) that selectively inhibits the vascular endothelial growth factor receptor-2. A weighted pooled analysis was performed to evaluate the clinical outcome, efficacy, and toxicity of apatinib in patients with advanced nonsmall cell lung cancer (NSCLC) that failed prior treatment with chemotherapy or epidermal growth factor receptor-TKIs (EGFR-TKIs). METHODS The literature published in PubMed, Embase, and Cochrane Library databases was searched (from inception to November 30, 2017) for eligible trials using the following search terms: apatinib AND (lung cancer OR NSCLC). Meeting abstracts were also reviewed to identify appropriate studies. Inclusion criteria were as follows: prospective or retrospective studies that evaluated efficacy and/or safety of apatinib in patients with advanced NSCLC that failed prior chemotherapy or EGFR-TKIs; primary outcome included one of these endpoints, progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), or adverse events (AEs); English language; and number of cases in the study ≥10 cases. RESULTS A total of 457 patients with advanced NSCLC were treated with apatinib in 14 studies (10 retrospective and 4 prospective studies) and were included in this pooled analysis. The pooled median PFS was 4.77 months [95% confidence interval (CI), 4.11-5.00] in all groups, 4.80 months (95% CI, 4.65-4.95) in the 750 mg apatinib (high-dose) group, and 3.88 months (95% CI, 3.11-4.65) in the 250 to 500 mg apatinib (low-dose) group. Median PFS stratified by single apatinib therapy or apatinib combined with continuous EGFR-TKIs was 4.76 months (95% CI, 3.66-5.06) and 5.20 months (95% CI, 3.66-6.74), respectively. The pooled median OS, ORR, and DCR values were 6.85 months, 18%, and 72%, respectively; pooled median ORR and DCR were 15% and 72% in the 750 mg apatinib group versus 20% and 72% in the 250 to 500 mg apatinib group. ORR and DCR stratified by therapeutic regimens were 14% and 70% for single-agent apatinib, 29% and 88% for apatinib combined with continuous EGFR-TKIs, and 26% and 63% for apatinib combined with chemotherapy, respectively. The pooled AE rates of grade 3/4 were hypertension (7%), proteinuria (3%), hand-foot-skin reaction (6%), fatigue (4%), decreased appetite (1.1%), oral mucositis (3%), and thrombocytopenia (3%). CONCLUSION Apatinib has promising antitumor activity and manageable toxicity profile in patients with advanced NSCLC that failed prior chemotherapy or EGFR-TKIs. This result needs to be confirmed through the ongoing Phase III clinical trial.
Collapse
|
8
|
Yuan XH, Yang J, Wang XY, Zhang XL, Qin TT, Li K. Association between EGFR/KRAS mutation and expression of VEGFA, VEGFR and VEGFR2 in lung adenocarcinoma. Oncol Lett 2018; 16:2105-2112. [PMID: 30008907 PMCID: PMC6036498 DOI: 10.3892/ol.2018.8901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 11/10/2017] [Indexed: 02/06/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (KRAS) are two of the most notable driver genes in lung cancer, whilst vascular endothelial growth factor (VEGF) signaling serves a critical function in tumor angiogenesis. However, few studies have focused on the potential connection between EGFR/KRAS mutational status, and VEGFA, VEGF receptor (VEGFR)1 and VEGFR2 expression in lung adenocarcinoma. EGFR (exon 19, 20 and 21) and KRAS (exon 2) mutations were detected using an amplification refractory mutation system technique, and the expression of VEGFA, VEGFR1 and VEGFR2 was analyzed using immunohistochemistry in 204 patients with lung adenocarcinoma. Associations between EGFR/KRAS mutational status and VEGFA, VEGFR1, and VEGFR2 expression was analyzed using Pearson χ2 tests. It was revealed that EGFR 21 exon (P=0.033) and EGFR 20 exon (P=0.002) mutated tumors exhibited a significantly higher level of expression of VEGFA. EGFR 21 exon mutant tumors additionally demonstrated a significantly higher level of co-expression of VEGFA and VEGFR1 (P<0.001). EGFR 19 exon mutation was significantly associated with low levels of VEGFR1 (P=0.008). KRAS mutation was significantly associated with a high level of co-expression of VEGFA, VEGFR1 and VEGFR2 (P=0.035), but no such association with the individual expression of VEGFA, VEGFR1 or VEGFR2 was identified. However, neither KRAS or EGFR mutations exhibited an association with the expression of VEGFR2. The present study may help in the treatment of various patients with KRAS or subtype of EGFR mutation with anti-angiogenesis therapy.
Collapse
Affiliation(s)
- Xiao-Han Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Jie Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xin-Yue Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xiao-Ling Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Ting-Ting Qin
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Kai Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
9
|
He X, Zhang C, Shi C, Lu Q. Meta-analysis of mRNA expression profiles to identify differentially expressed genes in lung adenocarcinoma tissue from smokers and non-smokers. Oncol Rep 2018; 39:929-938. [PMID: 29328493 PMCID: PMC5802042 DOI: 10.3892/or.2018.6197] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 12/29/2017] [Indexed: 11/24/2022] Open
Abstract
Compared to other types of lung cancer, lung adenocarcinoma patients with a history of smoking have a poor prognosis during the treatment of lung cancer. How lung adenocarcinoma-related genes are differentially expressed between smoker and non-smoker patients has yet to be fully elucidated. We performed a meta-analysis of four publicly available microarray datasets related to lung adenocarcinoma tissue in patients with a history of smoking using R statistical software. The top 50 differentially expressed genes (DEGs) in smoking vs. non‑smoking patients are shown using heat maps. Additionally, we conducted KEGG and GO analyses. In addition, we performed a PPI network analysis for 8 genes that were selected during a previous analysis. We identified a total of 2,932 DEGs (1,806 upregulated, 1,126 downregulated) and five genes (CDC45, CDC20, ANAPC7, CDC6, ESPL1) that may link lung adenocarcinoma to smoking history. Our study may provide new insights into the complex mechanisms of lung adenocarcinoma in smoking patients, and our novel gene expression signatures will be useful for future clinical studies.
Collapse
Affiliation(s)
- Xiaona He
- Department of Biostatistics and Epidemiology, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Cheng Zhang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chao Shi
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Quqin Lu
- Department of Biostatistics and Epidemiology, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
10
|
Ye M, Zhang X, Li N, Zhang Y, Jing P, Chang N, Wu J, Ren X, Zhang J. ALK and ROS1 as targeted therapy paradigms and clinical implications to overcome crizotinib resistance. Oncotarget 2017; 7:12289-304. [PMID: 26802023 PMCID: PMC4914285 DOI: 10.18632/oncotarget.6935] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022] Open
Abstract
During the past decade, more than 10 targetable oncogenic driver genes have been validated in non-small cell lung cancer (NSCLC). Anaplastic lymphoma kinase (ALK) and ROS1 kinase are two new driver genes implicated in ALK- and ROS1-rearranged NSCLC. Inhibition of ALK and ROS1 by crizotinib has been reported to be highly effective and well tolerated in these patients. However, resistance to crizotinib emerges years after treatment, and increasing efforts have been made to overcome this issue. Here, we review the biology of ALK and ROS1 and their roles in cancer progression. We also summarize the ongoing and completed clinical trials validating ALK and ROS1 as targets for cancer treatment. In the last section of the review, we will discuss the molecular mechanisms of crizotinib resistance and focus approaches to overcome it. This review describes an exciting new area of research and may provide new insights for targeted cancer therapies.
Collapse
Affiliation(s)
- Mingxiang Ye
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinxin Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Nan Li
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yong Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pengyu Jing
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ning Chang
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianxiong Wu
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinling Ren
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jian Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Sharma A. Chemoresistance in cancer cells: exosomes as potential regulators of therapeutic tumor heterogeneity. Nanomedicine (Lond) 2017; 12:2137-2148. [DOI: 10.2217/nnm-2017-0184] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Drug resistance in cancer cells remains a fundamental challenge. Be it nontargeted or targeted drugs, the presence of intrinsic or acquired cancer cell resistance remains a great obstacle in chemotherapy. Conventionally, a spectrum of cellular mechanisms defines drug resistance including overexpression of antiapoptotic proteins and drug efflux pumps, mutations in target and synergistic activation of prosurvival pathways in tumor cells. In addition to these well-studied routes, exosome-induced chemoresistance is emerging as a novel mechanism. Mechanistically, exosomes impart resistance by direct drug export, transport of drug efflux pumps and miRNAs exchange among cells. Moreover, exosome signaling creates ‘therapeutic tumor heterogeneity’ and favorably condition tumor microenvironment. Here, we discuss exosomes’ role in chemoresistance and possibilities of developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Aman Sharma
- ExoCan Healthcare Technologies Pvt Ltd, L4, 400 NCL Innovation Park, Dr Homi Bhabha Road, Pune 411008, India
| |
Collapse
|
12
|
Akbay EA, Koyama S, Liu Y, Dries R, Bufe LE, Silkes M, Alam MM, Magee DM, Jones R, Jinushi M, Kulkarni M, Carretero J, Wang X, Warner-Hatten T, Cavanaugh JD, Osa A, Kumanogoh A, Freeman GJ, Awad MM, Christiani DC, Bueno R, Hammerman PS, Dranoff G, Wong KK. Interleukin-17A Promotes Lung Tumor Progression through Neutrophil Attraction to Tumor Sites and Mediating Resistance to PD-1 Blockade. J Thorac Oncol 2017; 12:1268-1279. [PMID: 28483607 DOI: 10.1016/j.jtho.2017.04.017] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/24/2017] [Accepted: 04/21/2017] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Proinflammatory cytokine interleukin-17A (IL-17A) is overexpressed in a subset of patients with lung cancer. We hypothesized that IL-17A promotes a protumorigenic inflammatory phenotype and inhibits antitumor immune responses. METHODS We generated bitransgenic mice expressing a conditional IL-17A allele along with conditional KrasG12D and performed immune phenotyping of mouse lungs, a survival analysis, and treatment studies with antibodies either blocking programmed cell death 1 (PD-1) or IL-6 or depleting neutrophils. To support the preclinical findings, we analyzed human gene expression data sets and immune profiled patient lung tumors. RESULTS Tumors in IL-17:KrasG12D mice grew more rapidly, resulting in a significantly shorter survival as compared with that of KrasG12D mice. IL-6, granulocyte colony-stimulating factor (G-CSF), milk fat globule-EGF factor 8 protein, and C-X-C motif chemokine ligand 1 were increased in the lungs of IL17:Kras mice. Time course analysis revealed that levels of tumor-associated neutrophils were significantly increased, and lymphocyte recruitment was significantly reduced in IL17:KrasG12D mice as compared with in KrasG12D mice. In therapeutic studies PD-1 blockade was not effective in treating IL-17:KrasG12D tumors. In contrast, blocking IL-6 or depleting neutrophils with an anti-Ly-6G antibody in the IL17:KrasG12D tumors resulted in a clinical response associated with T-cell activation. In tumors from patients with lung cancer with KRAS mutation we found a correlation between higher levels of IL-17A and colony- stimulating factor 3 and a significant correlation among high neutrophil and lower T-cell numbers. CONCLUSIONS Here we have shown that an increase in a single cytokine, IL-17A, without additional mutations can promote lung cancer growth by promoting inflammation, which contributes to resistance to PD-1 blockade and sensitizes tumors to cytokine and neutrophil depletion.
Collapse
Affiliation(s)
- Esra A Akbay
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas; Simmons Comprehensive Cancer Center, Dallas, Texas
| | - Shohei Koyama
- Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yan Liu
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ruben Dries
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lauren E Bufe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael Silkes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Md Maksudul Alam
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas; Simmons Comprehensive Cancer Center, Dallas, Texas
| | - Dillon M Magee
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas; Simmons Comprehensive Cancer Center, Dallas, Texas
| | - Robert Jones
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Masahisa Jinushi
- Graduate School of Medicine, Institute for Advanced Medical Research, Keio University, Tokyo, Japan
| | - Meghana Kulkarni
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Julian Carretero
- Department of Physiology, University of Valencia, Valencia, Spain
| | - Xiaoen Wang
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jillian D Cavanaugh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Akio Osa
- Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Gordon J Freeman
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M Awad
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Raphael Bueno
- Thoracic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Peter S Hammerman
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Glenn Dranoff
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York.
| |
Collapse
|
13
|
Dalleau K, Marzougui Y, Da Silva S, Ringot P, Ndiaye NC, Coulet A. Learning from biomedical linked data to suggest valid pharmacogenes. J Biomed Semantics 2017; 8:16. [PMID: 28427468 PMCID: PMC5399403 DOI: 10.1186/s13326-017-0125-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 03/29/2017] [Indexed: 12/15/2022] Open
Abstract
Background A standard task in pharmacogenomics research is identifying genes that may be involved in drug response variability, i.e., pharmacogenes. Because genomic experiments tended to generate many false positives, computational approaches based on the use of background knowledge have been proposed. Until now, only molecular networks or the biomedical literature were used, whereas many other resources are available. Method We propose here to consume a diverse and larger set of resources using linked data related either to genes, drugs or diseases. One of the advantages of linked data is that they are built on a standard framework that facilitates the joint use of various sources, and thus facilitates considering features of various origins. We propose a selection and linkage of data sources relevant to pharmacogenomics, including for example DisGeNET and Clinvar. We use machine learning to identify and prioritize pharmacogenes that are the most probably valid, considering the selected linked data. This identification relies on the classification of gene–drug pairs as either pharmacogenomically associated or not and was experimented with two machine learning methods –random forest and graph kernel–, which results are compared in this article. Results We assembled a set of linked data relative to pharmacogenomics, of 2,610,793 triples, coming from six distinct resources. Learning from these data, random forest enables identifying valid pharmacogenes with a F-measure of 0.73, on a 10 folds cross-validation, whereas graph kernel achieves a F-measure of 0.81. A list of top candidates proposed by both approaches is provided and their obtention is discussed. Electronic supplementary material The online version of this article (doi:10.1186/s13326-017-0125-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kevin Dalleau
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France
| | - Yassine Marzougui
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France.,Ecole nationale supérieure des mines de Nancy, Campus Artem, Nancy, France
| | - Sébastien Da Silva
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France
| | - Patrice Ringot
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France
| | - Ndeye Coumba Ndiaye
- UMR U1122 IGE-PCV (INSERM, University of Lorraine), 30 Rue Lionnois, Nancy, France
| | - Adrien Coulet
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France.
| |
Collapse
|
14
|
Aguiar PN, Santoro IL, Tadokoro H, de Lima Lopes G, Filardi BA, Oliveira P, Castelo-Branco P, Mountzios G, de Mello RA. A pooled analysis of nivolumab for the treatment of advanced non-small-cell lung cancer and the role of PD-L1 as a predictive biomarker. Immunotherapy 2016; 8:1011-1019. [PMID: 27485075 DOI: 10.2217/imt-2016-0032] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/26/2016] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Recent studies with nivolumab (a monoclonal antibody against programmed cell death 1 [PD-1] receptor) have shown promise non-small-cell lung cancer (NSCLC) treatment. METHODS To review available clinical trials data in order to assess nivolumab efficacy and the role of tumoral PDL-1 expression as a biomarker. RESULTS Nine eligible studies included 2102 patients. In the second line setting, nivolumab achieved a 1-year survival rate of 41%; and in the first line, a 1-year survival rate of 76%. For those with PD-L1 expression <1%, nivolumab showed a trend for improved survival compared with docetaxel. CONCLUSIONS The available data reinforce nivolumab activity against NSCLC in first-line or subsequent lines. Although PD-L1 expression is related to greater response, PD-L1 negative patients had also some benefit.
Collapse
Affiliation(s)
- Pedro N Aguiar
- Division of Medical Oncology, Federal University of São Paulo, São Paulo, Brazil
| | - Ilka Lopes Santoro
- Division of Medical Oncology, Federal University of São Paulo, São Paulo, Brazil
| | - Hakaru Tadokoro
- Division of Medical Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Pedro Oliveira
- Department of Population Studies, Abel Salazar Biomedical Institute, University of Porto, Porto, Portugal
| | - Pedro Castelo-Branco
- Division of Oncology, Department of Biomedical Sciences & Medicine, University of Algarve, Faro, Portugal
| | - Giannis Mountzios
- Department of Medical Oncology, University of Athens Medical School, Athens, Greece
| | - Ramon Andrade de Mello
- Division of Oncology, Department of Biomedical Sciences & Medicine, University of Algarve, Faro, Portugal
- Department of Medicine, Faculty of Medicine, University of Porto, Porto, Portugal
- Research Center, Cearense School of Oncology, Instituto do Câncer do Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
15
|
de Mello RA. TG4010 immunotherapy: a novel weapon against advanced non-small cell lung cancer? ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:185. [PMID: 27275498 PMCID: PMC4876279 DOI: 10.21037/atm.2016.04.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 11/06/2022]
|
16
|
Sharma A, Khatun Z, Shiras A. Tumor exosomes: cellular postmen of cancer diagnosis and personalized therapy. Nanomedicine (Lond) 2016; 11:421-37. [PMID: 26784674 DOI: 10.2217/nnm.15.210] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nanosized (30-150 nm) extracellular vesicles 'exosomes' are secreted by cells for intercellular communication during normal and pathological conditions. Exosomes carry biomacromolecules from cell-of-origin and, therefore, represent molecular bioprint of the cell. Tumor-derived exosomes or TDEx modulate tumor microenvironment by transfer of macromolecules locally as well as at distant metastatic sites. Due to their biological stability, TDEx are rich source of biomarkers in cancer patients. TDEx focused cancer diagnosis allows liquid biopsy-based tumor typing and may facilitate therapy response monitoring by developing novel exosomes diagnostics. Therefore, efficient and specific capturing of exosomes for subsequent amplification of the biomessages; for example, DNA, RNA, miRNA can reinvent cancer diagnosis. Here, in this review, we discuss advancements in exosomes isolation strategies, presence of exosomes biomarkers and importance of TDEx in gauging tumor heterogeneity for their potential use in cancer diagnosis, therapy.
Collapse
Affiliation(s)
- Aman Sharma
- ExoCan Healthcare Technologies Pvt Ltd, L4, 100 NCL Innovation Park, Dr Homi Bhabha Road, Pune-411008, India.,National Centre for Cell Science, SP Pune University Campus, Ganeshkhind, Pune411007
| | - Zamila Khatun
- ExoCan Healthcare Technologies Pvt Ltd, L4, 100 NCL Innovation Park, Dr Homi Bhabha Road, Pune-411008, India
| | - Anjali Shiras
- National Centre for Cell Science, SP Pune University Campus, Ganeshkhind, Pune411007
| |
Collapse
|
17
|
Hu Y, Dong XZ, Liu X, Liu P, Chen YB. Enhanced Antitumor Activity of Cetuximab in Combination with the Jak Inhibitor CYT387 against Non-Small-Cell Lung Cancer with Various Genotypes. Mol Pharm 2015; 13:689-97. [PMID: 26685983 DOI: 10.1021/acs.molpharmaceut.5b00927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yuan Hu
- Department
of Clinical Pharmacology, Pharmacy Care Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xian-Zhe Dong
- Department
of Clinical Pharmacology, Pharmacy Care Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xu Liu
- Department
of Clinical Pharmacology, Pharmacy Care Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ping Liu
- Department
of Clinical Pharmacology, Pharmacy Care Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yi-Bang Chen
- Department
of Pharmacology and System Therapeutics, Mount Sinai School of Medicine; New York, New York 10029, United States
| |
Collapse
|
18
|
Davis PJ, Sudha T, Lin HY, Mousa SA. Thyroid Hormone, Hormone Analogs, and Angiogenesis. Compr Physiol 2015; 6:353-62. [PMID: 26756636 DOI: 10.1002/cphy.c150011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Modulation by thyroid hormone and hormone analogs of angiogenesis in the heart after experimental infarction, and in other organs, has been appreciated for decades. Description of a plasma membrane receptor for thyroid hormone on the extracellular domain of integrin αvβ3 on endothelial cells has revealed the complexity of the nongenomic regulation of angiogenesis by the hormone. From αvβ3, the hormone directs transcription of specific vascular growth factor genes, regulates growth factor receptor/growth factor interactions and stimulates endothelial cell migration to a vitronectin cue; these actions are implicated experimentally in tumor-relevant angiogenesis and angioproliferative pulmonary hypertension. Derived from L-thyroxine (T4), tetraiodothyroacetic acid (tetrac) can be covalently bound to a polymer and as Nanotetrac acts exclusively at the hormone receptor on αvβ3 to block actions of T4 and 3,5,3'-triiodo-L-thyronine (T3) on angiogenesis. Other antiangiogenic actions of Nanotetrac include disruption of crosstalk between integrin αvβ3 and adjacent cell surface vascular growth factor receptors, resulting in disordered vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF; FGF2) actions at their respective plasma membrane receptors. From αvβ3, Nanotetrac also downregulates expression of VEGFA and epidermal growth factor receptor (EGFR) genes, upregulates transcription of the angiogenesis suppressor gene, thrombospondin 1 (THBS1; TSP1) and decreases cellular abundance of Ang-2 protein and matrix metalloproteinase-9. Existence of this receptor provides new insights into the multiple mechanisms by which thyroid hormone and hormone analogs may regulate angiogenesis at the molecular level. The receptor also offers pharmacological opportunities for interruption of pathological angiogenesis via integrin αvβ3.
Collapse
Affiliation(s)
- Paul J Davis
- Department of Medicine, Albany Medical College, Albany, New York, USA.,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| | - Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| | - Hung-Yun Lin
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA.,Institute of Cancer Biology and Drug Discovery, School of Medical Technology, Taipei Medical University, Taipei, Taiwan
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| |
Collapse
|
19
|
Sia D, Moeini A, Labgaa I, Villanueva A. The future of patient-derived tumor xenografts in cancer treatment. Pharmacogenomics 2015; 16:1671-83. [PMID: 26402657 DOI: 10.2217/pgs.15.102] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Over the last decades, major technological advancements have led to a better understanding of the molecular drivers of human malignancies. Nonetheless, this progress only marginally impacted the cancer therapeutic approach, probably due to the limited ability of experimental models to predict efficacy in clinical trials. In an effort to offset this limitation, there has been an increasing interest in the development of patient-derived xenograft (PDX) models where human tumors are xenotransplanted into immunocompromised mice. Considering their high resemblance to human tumors and their stability, PDX models are becoming the preferred translational tools in preclinical studies. Nonetheless, several limitations hamper a wider use of PDX models and tarnish the concept that they might represent the missing piece in the personalized medicine puzzle.
Collapse
Affiliation(s)
- Daniela Sia
- Barcelona-Clínic Liver Cancer Group, HCC Translational Research Laboratory, Liver Unit, Hepato-biliary Surgery, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, CIBERehd, Universitat de Barcelona, C/Rossello 153, Barcelona, Catalonia, Spain.,Gastrointestinal Surgery & Liver Transplantation Unit, Department of Surgery, National Cancer Institute, via Venezian, 1, Milan, Italy.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue NY 10029, USA
| | - Agrin Moeini
- Barcelona-Clínic Liver Cancer Group, HCC Translational Research Laboratory, Liver Unit, Hepato-biliary Surgery, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, CIBERehd, Universitat de Barcelona, C/Rossello 153, Barcelona, Catalonia, Spain.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue NY 10029, USA
| | - Ismail Labgaa
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue NY 10029, USA
| | - Augusto Villanueva
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue NY 10029, USA.,Division of Hematology & Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, NY, USA
| |
Collapse
|
20
|
Li S, Zhao L, Zhang J, Zou Z, Du P. Inhibition of lung adenocarcinoma cells by insulin-like growth factor-I receptor and Kirsten rat sarcoma mutations: A mutation analysis with antisense oligodeoxynucleotide. Thorac Cancer 2015; 6:260-8. [PMID: 26273371 PMCID: PMC4448390 DOI: 10.1111/1759-7714.12176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/07/2014] [Indexed: 11/29/2022] Open
Abstract
Background Kirsten rat sarcoma (KRAS) mutations are widespread in lung adenocarcinoma patients. The combined utilization of KRAS antisense oligodeoxynucleotide (ASODN) and insulin-like growth factor-I receptor (IGF-IR) may inhibit the proliferation of A549 cell lines of lung adenocarcinoma. Methods Point mutations of the KRAS gene in A549 cells were detected by polymerase chain reaction with special sequence primers (PCR-SSP) and gene sequence analysis; ASODN was designed and synthesized according to the mutation specialty of KRAS; and the correlation of gene mutations and clinicopathological features were analyzed. Inhibition on the proliferation and morphostructure change were measured by methyl thiazolyl tetrazolium and colony-forming unit assays. Flow cytometry was used to evaluate the expression of KRAS and IGF-IR proteins and cell apoptosis and reverse transcriptase-polymerase chain reaction were used to detect the expression of KRAS and IGF-IR messenger ribonucleic acid (mRNA). Male nude mice were used to form the mice-human lung cancer model and show the inhibition of KRAS ASODN on A549 cells. Results PCR-SSP and gene sequence analysis results showed that the codon 12 of KRAS had changed from GGT to GTT. KRAS ASODN or IGF-IR ASODN could inhibit cell proliferation and promote apoptosis of A549 cells. However, the combined utilization of KRAS ASODN and IGF-IR ASODN could inhibit cell proliferation and promote apoptosis more powerfully than exclusive use of KRAS ASODN or IGF-IR ASODN. Conclusion The two ASODNs can inhibit the proliferation of lung adenocarcinoma cells through decreasing the expression of KRAS and IGF-IR mRNA and protein.
Collapse
Affiliation(s)
- Shaoming Li
- Departments of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University Xi'an, China
| | - Lei Zhao
- The First Affiliated Hospital of Dalian Medical University Dalian, China
| | | | | | - Peng Du
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai, China
| |
Collapse
|
21
|
Moschini I, Dell'Anna C, Losardo PL, Bordi P, D'Abbiero N, Tiseo M. Radiotherapy of non-small-cell lung cancer in the era of EGFR gene mutations and EGF receptor tyrosine kinase inhibitors. Future Oncol 2015; 11:2329-42. [DOI: 10.2217/fon.15.156] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) occurs, approximately, in 80–85% of all cases of lung cancer. The majority of patients present locally advanced or metastatic disease when diagnosed, with poor prognosis. The discovery of activating mutations in the EGFR gene has started a new era of personalized treatment for NSCLC patients. To improve the treatment outcome in patients with unresectable NSCLC and, in particular, EGFR mutated, a combined strategy of radiotherapy and medical treatment can be undertaken. In this review we will discuss preclinical data regarding EGF receptor (EGFR) tyrosine kinase inhibitors (TKIs) and radiotherapy, available clinical trials investigating efficacy and toxicity of combined treatment (thoracic or whole brain radiotherapy and EGFR-TKIs) and, also, the role of local radiation in mutated EGFR patients who developed EGFR-TKI resistance.
Collapse
Affiliation(s)
| | | | | | - Paola Bordi
- Medical Oncology Unit, University Hospital of Parma, Italy
| | | | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Italy
| |
Collapse
|
22
|
Aziz A, Kempkensteffen C, May M, Lebentrau S, Burger M, Chun FKH, Brookman-May S. Prognostic, predictive and potential surrogate markers in castration-resistant prostate cancer. Expert Rev Anticancer Ther 2015; 15:649-66. [DOI: 10.1586/14737140.2015.1038247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
de Mello RA, Pousa I, Pereira D. Nivolumab for advanced squamous cell lung cancer: what are the next steps? Lancet Oncol 2015; 16:234-235. [PMID: 25704436 DOI: 10.1016/s1470-2045(15)70074-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Ramon Andrade de Mello
- Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal; Department of Medical Oncology, Portuguese Oncology Institute, Porto, Portugal.
| | - Inés Pousa
- Department of Medical Oncology, Portuguese Oncology Institute, Porto, Portugal
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Oncology Institute, Porto, Portugal
| |
Collapse
|
24
|
Madureira P, de Mello RA, de Vasconcelos A, Zhang Y. Immunotherapy for lung cancer: for whom the bell tolls? Tumour Biol 2015; 36:1411-1422. [PMID: 25736929 DOI: 10.1007/s13277-015-3285-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 02/18/2015] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death and accounts for approximately 30% of all cancer deaths. Despite the recent developments in personalized therapy, the prognosis in lung cancer is still very poor. Immunotherapy is now emerging as a new hope for patients with lung cancer. It is well known that standard chemotherapeutic regimens have devastating effects for the patient's immune system. Therefore, the aim of immunotherapy is to specifically enhance the immune response against the tumour. Recently, many trials addressed the role of such therapies for metastatic non-small cell lung cancer (NSCLC) treatment: ipilimumab, tremelimumab, nivolumab and pembrolizumab are immunotherapeutic agents of high relevance in this field. Anti-tumour vaccines, as well as dendritic cell-based therapies, have emerged as potent inducers of immune response against the tumour. Herein, we will review some of the most promising cancer immunotherapies, highlighting their advantages and try to understand, in an immunological perspective, the missteps associated with the current treatments for cancer.
Collapse
Affiliation(s)
- Pedro Madureira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | | | | | | |
Collapse
|
25
|
Wang Z, Wu X, Shi Y, Han X, Cheng G, Cui D, Li L, Zhang Y, Mu X, Zhang L, Yang L, Di J, Yu Q, Liu D. Ventana immunohistochemistry ALK (D5F3) detection of ALK expression in pleural effusion samples of lung adenocarcinoma. Per Med 2015; 12:349-357. [PMID: 29771657 DOI: 10.2217/pme.15.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AIM To evaluate the Ventana IHC ALK (D5F3) assay for detecting anaplastic lymphoma kinase (ALK) protein expression in pleural effusion samples. METHODS Historical, selected (wild-type EGFR, K-RAS) pleural effusion cytologic blocks of lung adenocarcinoma samples (Study 1) and unselected lung adenocarcinoma pleural effusion cytologic blocks (Study 2) were tested by Ventana IHC ALK (D5F3) assay. Quantitative real-time-PCR was used to verify immunohistochemistry results. RESULTS A total of 17 out of 100 (Study 1) and ten out of 104 (Study 2) pleural effusion samples were ALK expression positive by the Ventana IHC ALK (D5F3) assay. The ALK fusion results with immunohistochemistry and quantitative real-time-PCR had a concordance rate of 87.5% (κ = 0.886; p < 0.001). CONCLUSION The Ventana IHC ALK (D5F3) assay is a reliable tool for detecting ALK protein expression in pleural effusion samples.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Pathology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Xiaonan Wu
- Department of Medical Oncology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Yuankai Shi
- Department of Medical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaohong Han
- Department of Medical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Cheng
- Department of Medical Oncology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Di Cui
- Department of Pathology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Lin Li
- Department of Medical Oncology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Yuhui Zhang
- Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xinlin Mu
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Li Zhang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Li Yang
- Department of Pathology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Jing Di
- Department of Pathology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Qi Yu
- Department of Pathology, Beijing Hospital of the Ministry of Health, Beijing, China
| | - Dongge Liu
- Department of Pathology, Beijing Hospital of the Ministry of Health, Beijing, China
| |
Collapse
|
26
|
Deng LL, Deng HB, Lu CL, Guo Y, Wang D, Yan CH, Lv X, Shao YX. Mutations of EGFR or KRAS and expression of chemotherapy-related genes based on small biopsy samples in stage IIIB and IV inoperable non-small cell lung cancer. J Cancer Res Clin Oncol 2014; 140:2097-105. [PMID: 24994038 DOI: 10.1007/s00432-014-1751-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/19/2014] [Indexed: 01/19/2023]
Abstract
PURPOSES Epidermal growth factor receptor (EGFR) and KRAS mutations may predict the outcome of targeted drug therapy and also may be associated with the efficacy of chemotherapy in patients with non-small cell lung cancer (NSCLC). This report investigated the relation of EGFR or KRAS mutation and expression of chemotherapy-related genes, including excision repair cross-complementing 1 (ERCC1), thymidylate synthetase (TYMS), ribonucleotide reductase subunit M1 (RRM1) and class III β-tubulin (TUBB3), as a potential explanation for these observations. METHODS A total of 143 patients with stage IIIB and IV NSCLC from bronchoscopy or percutaneous lung biopsy obtained tumor samples were analyzed concurrently for EGFR or KRAS mutations, and mRNA expression of ERCC1, TYMS, RRM1 and TUBB3. EGFR or KRAS mutations were detected with xTAG liquidchip technology (xTAG-LCT), and mRNA expression levels of four genes were detected by branched DNA-liquidchip technology (bDNA-LCT). RESULTS Of 143 patients, 63 tumors were positive for EGFR-activating mutations, and 16 tumors were positive for KRAS mutations. EGFR-activating mutations are more frequent in females, adenocarcinoma and non-smokers patients, and KRAS mutations are more frequent in smoking patients. ERCC1 mRNA levels were significantly associated with histological type and tumor differentiation, whereas TYMS levels were significantly associated with age. NSCLC specimens that harboring EGFR-activating mutations are more likely to express low ERCC1 and high TUBB3 mRNA levels, whereas tumors from patients with NSCLC harboring KRAS mutation are more likely to express high ERCC1 mRNA levels. CONCLUSIONS Mutations and expression of chemotherapy-related genes may provide a basis for the selection of suitable molecular markers for individual treatment in a population with stage IIIB and IV NSCLC.
Collapse
Affiliation(s)
- Li-Li Deng
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Clinical and metabolic parameters in non-small cell lung carcinoma and colorectal cancer patients with and without KRAS mutations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2014; 11:8645-60. [PMID: 25158139 PMCID: PMC4198984 DOI: 10.3390/ijerph110908645] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 07/28/2014] [Accepted: 08/15/2014] [Indexed: 12/12/2022]
Abstract
Lung cancer (LC) and colorectal cancer (CRC) are the first and second deadliest types of cancer worldwide. EGFR-based therapy has been used in the treatment of these cancers with variable success. Presence of mutations in the KRAS driver oncogene, possibly induced by environmental factors such as carcinogens in diet and cigarette smoke, may confer worse prognosis and resistance to treatment for reasons not fully understood. Data on possible associations between KRAS mutational status and clinical and metabolic parameters, which may help in clinical management, as well as in identifying risk factors for developing these cancers, are limited in the current literature. We sequenced the KRAS gene and investigated the associations of variations in 108 patients with non-small cell lung carcinoma (NSCLC), the most common form of LC, and in 116 patients with CRC. All of the mutations originated from the guanosine nucleotide and over half of all transversions in NSCLC and CRC were c.34 G>T and c.35 G>T, respectively. c.35 G>A was the most frequent type of transition in both cancers. Excluding smoking, the clinical and metabolic parameters in patients carrying mutant and wild type KRAS were similar except that the CRC patients with transversion mutations were 8.6 years younger than those carrying the transitions (P < 0.01). Dyslipidemia, hypertension, family cancer history, and age of diagnosis older than 60 years were more frequent in NSCLC than CRC (P ≤ 0.04). These results suggest that most of the clinical and metabolic parameters investigated in this study are probably not associated with the more aggressive phenotype and differences in response to EGFR-based treatment previously reported in patients with KRAS mutations. However, the increased rates of abnormal metabolic parameters in patients with NSCLC in comparison to CRC indicate that these parameters may be more important in the management of NSCLC. CRC patients carrying transition mutations are older than those carrying transversions, suggesting that age may determine the type of KRAS mutation in CRC patients.
Collapse
|
28
|
Madureira P, de Mello RA. BRAF and MEK gene rearrangements in melanoma: implications for targeted therapy. Mol Diagn Ther 2014; 18:285-291. [PMID: 24403169 DOI: 10.1007/s40291-013-0081-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The incidence of melanoma has been continuously increasing in the last decades, and faster than any other cancers. Melanoma is the leading cause of death from skin disease. It is estimated that 76,690 Americans will be diagnosed with melanoma in 2013 and 9,480 will die from the disease. Molecular mechanisms underlying melanoma pathogenesis have been extensively studied and novel therapeutic weapons developed. BRAF and MEK pathways emerged as key players in this field. Recently, novel drugs such as vemurafenib, dabrafenib and trametinib were approved for treatment of advanced disease harbouring BRAF V600E and V600K mutations. In addition, an effective strategy to build upon the successes seen with dabrafenib and trametinib monotherapies has been to combine these agents (CombiDT), with the goal of further improving response rates and delaying resistance. Our review gives an overall point of view concerning BRAF and MEK pathways as well as the role of BRAF and MEK testing in directing the personalised treatment of patients with metastatic melanoma.
Collapse
Affiliation(s)
- Pedro Madureira
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | | |
Collapse
|
29
|
Opitz I, Bueno R, Lim E, Pass H, Pastorino U, Boeri M, Rocco G. Biomolecular and clinical practice in malignant pleural mesothelioma and lung cancer: what thoracic surgeons should know. Eur J Cardiothorac Surg 2014; 46:602-6. [PMID: 24623168 DOI: 10.1093/ejcts/ezu048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Today, molecular-profile-directed therapy is a guiding principle of modern thoracic oncology. The knowledge of new biomolecular technology applied to the diagnosis, prognosis, and treatment of lung cancer and mesothelioma should be part of the 21st century thoracic surgeons' professional competence. The European Society of Thoracic Surgeons (ESTS) Biology Club aims at providing a comprehensive insight into the basic biology of the diseases we are treating. During the 2013 ESTS Annual Meeting, different experts of the field presented the current knowledge about diagnostic and prognostic biomarkers in malignant pleural mesothelioma including new perspectives as well as the role and potential application of microRNA and genomic sequencing for lung cancer, which are summarized in the present article.
Collapse
Affiliation(s)
- Isabelle Opitz
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Raphael Bueno
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric Lim
- Imperial College and the Academic Division of Thoracic Surgery, The Royal Brompton Hospital, London, UK
| | - Harvey Pass
- Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Ugo Pastorino
- Division of Thoracic Surgery, Istituto Nazionale Tumori, Milan, Italy
| | - Mattia Boeri
- Tumor Genomics Unit, Istituto Nazionale Tumori, Milan, Italy
| | - Gaetano Rocco
- Department of Thoracic Surgery and Oncology, Instituto Nazionale Tumori, Fondazione "G. Pascale", IRCCS, Naples, Italy
| | | |
Collapse
|
30
|
Abstract
Antibody-based immunotherapies are important therapy options in human oncology. Although human humoral specific immunity is constituted of five different immunoglobulin classes, currently only IgG-based immunotherapies have proceeded to clinical application. This review, however, discusses the benefits and difficulties of IgE-based immunotherapy of cancer, with special emphasis on how to translate promising preclinical results into clinical studies. Pursuing the “Comparative Oncology” approach, novel drug candidates are investigated in clinical trials with veterinary cancer patients, most often dogs. By this strategy drug development could be speeded up, animal experiments could be reduced and novel therapy options could be introduced benefitting humans as well as man’s best friend.
Collapse
Affiliation(s)
- Josef Singer
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, and University Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, and University Vienna, Vienna, Austria ; Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|