1
|
Kim J, Noh MH, Hur DY, Kim B, Kim YS, Lee HK. Celecoxib upregulates ULBP-1 expression in lung cancer cells via the JNK/PI3K signaling pathway and increases susceptibility to natural killer cell cytotoxicity. Oncol Lett 2020; 20:279. [PMID: 33014157 PMCID: PMC7520723 DOI: 10.3892/ol.2020.12142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/08/2020] [Indexed: 01/22/2023] Open
Abstract
Lung cancer has the highest cancer mortality rate in the world, and effective therapies are still required. Cyclooxygenase-2 (COX-2) is highly expressed in numerous types of cancer, and is therefore considered a possible target of cancer treatment. Celecoxib, a selective COX-2 inhibitor, has binding pockets that interact with COX-2 and disrupt its enzymatic activities. In addition, celecoxib is able to affect cellular functions in a COX-2-independent manner. The present study aimed to investigate if celecoxib affected natural killer (NK) cell receptors and susceptibility to NK cell toxicity. For this purpose, PCR, immunoblotting, flow cytometry analysis and NK cell cytotoxicity assays were performed. The present study revealed that sublethal concentrations of celecoxib increased the expression levels of UL16-binding protein 1 (ULBP-1), a natural-killer group 2 member D (NKG2D) ligand, in lung cancer A549 and H460 cell lines. ULBP-1 mRNA and protein expression was induced in a dose- and time-dependent manner after celecoxib treatment. Expression levels of other NKG2D ligands, such as ULBP-2, ULBP-3, MHC class I-related chain A (MICA) and MICB did not change considerably compared to ULBP-1 in response to celecoxib treatment. Fluorescence microscopic images revealed abundant ULBP-1 in the cytoplasm after celecoxib treatment. Both JNK and PI3K may be involved in the induction of ULBP-1 expression after celecoxib treatment in A549 and H460 cells. In a NK cytotoxicity assay, celecoxib increased the sensitivity to NK cell-mediated cytotoxicity via interaction with ULBP-1 in lung cancer cells. Overall, the present results demonstrated that celecoxib treatment induced ULBP-1 expression in lung cancer cells, thereby increasing their susceptibility to NK cell cytotoxicity. These results suggest that the effects of conventional anticancer therapy may potentially be enhanced by using celecoxib, which targets COX-2, to enhance the sensitivity of lung cancer cells to NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Medical Science, Pusan National University School of Medicine, Yangsan, South Gyeongsang 50612, Republic of Korea
| | - Min Hye Noh
- Department of Anatomy, Inje University College of Medicine, Busan 47396, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy, Inje University College of Medicine, Busan 47396, Republic of Korea
| | - Bomi Kim
- Department of Pathology, Inje University Haewoondae Paik Hospital, Busan 48108, Republic of Korea
| | - Yeong Seok Kim
- Department of Anatomy, Inje University College of Medicine, Busan 47396, Republic of Korea
| | - Hyun-Kyung Lee
- Division of Pulmonology, Department of Internal Medicine, Inje University Pusan Paik Hospital, Busan 47396, Republic of Korea
| |
Collapse
|
2
|
Li D, He S. MAGE3 and Survivin activated dendritic cell immunotherapy for the treatment of non-small cell lung cancer. Oncol Lett 2018; 15:8777-8783. [PMID: 29805617 DOI: 10.3892/ol.2018.8362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/24/2016] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell (DC) immunotherapy is an optimal cancer treatment, resulting in its emergence as a therapeutic choice; however, there are limited studies investigating dual antigen-pulsed DC immunotherapy in non-small cell lung cancer (NSCLC). In order to determine the effect of a recombinant melanoma-associated antigen (rMAGE-3) and recombinant Survivin (rSurvivin) peptide-pulsed DC immunotherapy in patients with NSCLC, the present clinical study was performed. DC immunotherapy was generated from the monocytes of patients with NSCLC and primed with rMAGE-3 and rSurvivin peptides. The present open-label, non-randomised study enrolled 16 patients with histologically confirmed stage I-IIIB NSCLC between December 2013 and October 2014. A prime immunotherapy (9.1×107 cells/dose) and a single boost (8.2×107 cells/dose) were administered 1 month apart intradermally and the patients were evaluated for immunological and clinical response. DC immunotherapy was well tolerated, with no serious adverse events. There was a single incidence of grade 1 fever, chills and fatigue. Out of the 16 patients enrolled, 11 patients showed stable disease and 5 showed disease progression. There was a significant increase in IFN-γ expression on day 60 vs. day 0 (P=0.048). An increasing trend in the mean cluster of differentiation (CD)4:CD8 values of day 30 and day 90 was observed, but this was not significant. The present study established that DCs primed with rMAGE-3 and rSurvivin may be used in NSCLC treatment. However, a larger study is required to address prominent issues, including secretion of immunosuppressive cytokines and mechanisms of tumour escape from immune surveillance. Several factors associated with the manufacturing and quality of immunotherapy also require standardisation.
Collapse
Affiliation(s)
- Dong Li
- Department of Cardiothoracic Surgery, Central Hospital of Zibo, Zibo, Shandong 250012, P.R. China
| | - Song He
- Maanshan Center for Clinical Laboratory, Maanshan, Anhui 243000, P.R. China.,Maanshan Municipal Hospital Group, Maanshan, Anhui 243000, P.R. China
| |
Collapse
|
3
|
Kim B, Kim J, Kim YS. Celecoxib induces cell death on non-small cell lung cancer cells through endoplasmic reticulum stress. Anat Cell Biol 2017; 50:293-300. [PMID: 29354301 PMCID: PMC5768566 DOI: 10.5115/acb.2017.50.4.293] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) is an enzyme induced by various proinflammatory and mitogenic stimuli. Celecoxib is a selective inhibitor of COX-2 that have been shown to affect cell growth and apoptosis. Lung cancer cells expressing COX-2 is able to be a target of celecoxib, this study focuses on investigating that celecoxib induces apoptosis via endoplasmic reticulum (ER) stress on lung cancer cells. We investigated whether celecoxib induced apoptosis on non-small cell lung cancer cell line, A549 and H460. The 50 µM of celecoxib increased apoptotic cells and 100 µM of celecoxib significantly induced apoptosis. To check involvement of caspase cascade, pretreatment of z-VAD-fmk blocked celecoxib-induced apoptosis. However, caspase-3, -8, and -9 were not activated, but cleavage of non-classical caspase-4 was detected using western blot. As checking ER stress associated molecules, celecoxib did not increase expressions of growth arrest and DNA damage inducible protein 34, activating transcription factor 4, and spliced X-box binding protiens-1, but increase of both glucose-regulated protein 78 (GRP78) and C/EBP homologous transcription factor were detected. Salubrinal, inhibitor of eIF2 and siRNA for IRE1 did not alter celecoxib-induced apoptosis. Instead, celecoxib-induced apoptosis might be deeply associated with ER stress depending on GRP78 because siRNA for GRP78 enhanced apoptosis. Taken together, celecoxib triggered ER stress on lung cancer cells and celecoxib-induced apoptosis might be involved in both non-classical caspase-4 and GRP78.
Collapse
Affiliation(s)
- Bomi Kim
- Department of Pathology, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Jayoung Kim
- Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine, Busan, Korea
| | - Yeong Seok Kim
- Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
4
|
Kim J, Hong SW, Kim S, Kim D, Hur DY, Jin DH, Kim B, Kim YS. Cyclooxygenase-2 expression is induced by celecoxib treatment in lung cancer cells and is transferred to neighbor cells via exosomes. Int J Oncol 2017; 52:613-620. [PMID: 29345286 DOI: 10.3892/ijo.2017.4227] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/08/2017] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is one of most common types of cancer worldwide. Lung cancer results in a death higher rate each year compared to colon, breast and prostate cancer combined. Celecoxib is a selective inhibitor of cyclooxygenase-2 (COX‑2), an enzyme of which the expression is induced by various stimuli, such as inflammation. In addition, celecoxib triggers COX-2 loading on exosomes. Exosomes are small vesicles composed of a lipid bilayer membrane and are found in most biological fluids, such as blood breast milk and urine. In this study, we focused on exosomes containing COX-2 proteins from lung cancer cells to determine their involvement in the interaction with neighbor cells following treatment with celecoxib. We found that celecoxib induced COX-2 expression in both the cytosol and exosomes in lung cancer cells. Exosomes from celecoxib-treated lung cancer cell culture supernatant were isolated and incubated with several types of cells. The THP-1, monocytic leukemia cell line effectively absorbed COX-2 by lung cancer cell-derived exosomes. Following incubation with exosomes, the COX-2 protein level was increased in the THP-1 cells; however, COX-2 mRNA expression was not affected. Moreover, prostaglandin E2 (PGE2) and vascular endothelial growth factor (VEGF) production by THP-1 cells was increased following incubation with exosomes from celecoxib-treated lung cancer cells. Conditioned medium from THP-1 following incubation with exosomes promoted formation in EA.hy926 cells. Taken together, our findings suggest that celecoxib induces COX-2 expression in lung cancer cells, and that highly expressed COX-2 in exosomes can be transferred to other cells.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Anatomy and Research Center for Tumor Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Seung-Woo Hong
- Department of Anatomy and Research Center for Tumor Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Seonghan Kim
- Department of Anatomy and Research Center for Tumor Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Daejin Kim
- Department of Anatomy and Research Center for Tumor Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy and Research Center for Tumor Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Dong-Hoon Jin
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Bomi Kim
- Department of Pathology, Haeundae Paik Hospital, College of Medicine, Inje University, Busan 48108, Republic of Korea
| | - Yeong Seok Kim
- Department of Anatomy and Research Center for Tumor Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| |
Collapse
|
5
|
Phase I study of sorafenib and tipifarnib for recurrent glioblastoma: NABTC 05-02. J Neurooncol 2017; 136:79-86. [PMID: 28988377 DOI: 10.1007/s11060-017-2624-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/22/2017] [Indexed: 10/18/2022]
Abstract
Recurrent glioblastoma (GBM) has a very low 6-month progression free survival (PFS) with currently available treatments. Combination chemotherapy to target multiple cell signaling pathways is currently being investigated in order to improve prognosis for recurrent disease. The purpose of this phase I study was to determine the maximum tolerated dose (MTD) for the combination of tipifarnib and sorafenib for the treatment of recurrent GBM. Patients with pathologically proven WHO grade IV GBM and radiographically proven tumor recurrence were eligible for this study. Treatments included sorafenib at twice daily and escalating dosages of tipifarnib. Dose-limiting toxicity (DLT) was determined over the first 28-days of treatments, and the MTD was determined in a 3 + 3 study design. We enrolled 24 patients, and 21 patients completed the MTD period. The study was stopped early with no MTD determination for excessive toxicities. The last dose level reached was sorafenib at 200 mg twice a day and tipifarnib 100 mg twice a day on an alternating week schedule. The DLTs included diarrhea, lipase elevation, hypophosphatemia, and arthralgia. The combination of sorafenib and tipifarnib has excessive toxicities and full single agent dosages could not be achieved in combination.
Collapse
|
6
|
Ahmed M, Carrascosa LG, Ibn Sina AA, Zarate EM, Korbie D, Ru KL, Shiddiky MJ, Mainwaring P, Trau M. Detection of aberrant protein phosphorylation in cancer using direct gold-protein affinity interactions. Biosens Bioelectron 2017; 91:8-14. [DOI: 10.1016/j.bios.2016.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/27/2016] [Accepted: 12/06/2016] [Indexed: 01/06/2023]
|
7
|
Zhao S, Qiu ZX, Zhang L, Li WM. Prognostic values of ERK1/2 and p-ERK1/2 expressions for poor survival in non-small cell lung cancer. Tumour Biol 2015; 36:4143-50. [PMID: 25596700 DOI: 10.1007/s13277-015-3048-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/02/2015] [Indexed: 02/05/2023] Open
Abstract
The extracellular-regulated kinase (ERK) 1/2, as a member of the mitogen-activated protein kinase family, plays a crucial role in the development of cancer. However, little is known about the prognostic value of ERK1/2 and phosphorylated ERK1/2 (p-ERK1/2) in non-small cell lung cancer (NSCLC). Thus, we investigated their prognostic values and analyzed the associations between their expressions and clinicopathological features in NSCLC patients. We examined ERK1/2 and p-ERK1/2 expressions via immunohistochemistry in 183 NSCLC samples. The prognostic significances of protein expression were evaluated with univariate and multivariate survival analysis. Of the specimens, 44.8 and 44.3 % revealed positive staining for ERK1/2 and p-ERK1/2, respectively. There were 24.6 % specimens with both ERK1/2 and p-ERK1/2-positive expression. The results showed p-ERK1/2-positive expression was an independent prognostic factor for poor overall survival (OS) in NSCLC patients on both univariate analysis (p < 0.0001) and multivariate analysis (p = 0.0000). Meanwhile, the positive expression of both proteins was also associated with poor OS (p = 0.002). With respect to clinicopathological features, the tumor differentiation was significantly associated with the positivity of ERK1/2, p-ERK1/2, and both proteins, while histological type was only related to ERK1/2. However, there were no significant differences between the expressions and other clinical features, such as gender, age, smoking, tumor-node-metastasis (TNM) stage, lymph node metastasis, and treatments. The p-ERK1/2-positive expression was associated with adverse outcomes, and the positive expression of both ERK1/2 and p-ERK1/2 proteins was also related to poor OS. Therefore, the positivity of p-ERK1/2 expression may serve as a vital biomarker in the development of NSCLC.
Collapse
Affiliation(s)
- Shuang Zhao
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China,
| | | | | | | |
Collapse
|
8
|
Cho YS, Park SY. Harnessing of Programmed Necrosis for Fighting against Cancers. Biomol Ther (Seoul) 2014; 22:167-75. [PMID: 25009696 PMCID: PMC4060077 DOI: 10.4062/biomolther.2014.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/11/2014] [Accepted: 05/12/2014] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy has long been considered as one of useful strategies for cancer treatment. It is primarily based on the apoptosis that can selectively kill cancer cells. However, cancer cells can progressively develop an acquired resistance to apoptotic cell death, rendering refractory to chemo- and radiotherapies. Although the mechanism by which cells attained resistance to drug remains to be clarified, it might be caused by either pumping out of them or interfering with apoptotic signal cascades in response to cancer drugs. In case that cancer cells are defective in some part of apoptotic machinery by repeated exposure to anticancer drugs, alternative cell death mechanistically distinct from apoptosis could be adopted to remove cancer cells refractory to apoptosis-inducing agents. This review will mainly deal with harnessing of necrotic cell death, specifically, programmed necrosis and practical uses. Here, we begin with various defects of apoptotic death machinery in cancer cells, and then provide new perspective on programmed necrosis as an alternative anticancer approach.
Collapse
Affiliation(s)
- Young Sik Cho
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Seung Yeon Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| |
Collapse
|
9
|
Yang Y, Zhao W, Xu QW, Wang XS, Zhang Y, Zhang J. IQGAP3 promotes EGFR-ERK signaling and the growth and metastasis of lung cancer cells. PLoS One 2014; 9:e97578. [PMID: 24849319 PMCID: PMC4029748 DOI: 10.1371/journal.pone.0097578] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/21/2014] [Indexed: 12/23/2022] Open
Abstract
Proteins of the IQGAP family display complicated and often contradictory activities in tumorigenesis. IQGAP1 has well documented oncogenic potential and IQGAP2 has putative tumor-suppressive function. IQGAP3 is the latest addition to this family and its role in cancer development remains to be defined. Here we demonstrate IQGAP3 expression is markedly increased in lung cancer tissues at both mRNA and protein levels. Overexpression of IQGAP3 promoted tumor cell growth, and migration and invasion, whereas knockdown of IQGAP3 exhibited opposite effects. Moreover, suppression of IQGAP3 in a lung cancer cell line caused a reduction in the tumorigenicity of these cells in lung tissue after intravenous injection. Furthermore, we showed that IQGAP3 is able to interact with ERK1 and enhance its phosphorylation following treatment with EGF. These data suggest that IQGAP3 may contribute to the pathogenesis of lung cancer by modulating EGFR-ERK signaling.
Collapse
Affiliation(s)
- Ying Yang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, P. R. China
| | - Wei Zhao
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, P. R. China
| | - Qing-Wen Xu
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, P. R. China
| | - Xiao-Song Wang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, P. R. China
| | - Yu Zhang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, P. R. China
- * E-mail: (YZ); (JZ)
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Public Health, Peking University Health Science Center, Beijing, P. R. China
- * E-mail: (YZ); (JZ)
| |
Collapse
|
10
|
Chen YT, Feng B, Chen LB. Update of research on drug resistance in small cell lung cancer chemotherapy. Asian Pac J Cancer Prev 2013; 13:3577-81. [PMID: 23098422 DOI: 10.7314/apjcp.2012.13.8.3577] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Small cell lung cancer (SCLC) is characterized by a short cell doubling time, rapid progression and early occurrence of blood-borne and lymph metastasis. The malignancy is the highest of all lung cancer types. Although SCLC has a relatively good initial response to chemotherapy as well as radiotherapy, relapse or disease progression may occur quickly after the initial treatment. Drug resistance, especially multi-drug resistance, is the most important cause of failure of SCLC chemotherapy. This article provides a brief update of research on mechanisms of drug resistance in SCLC and reversal strategies.
Collapse
Affiliation(s)
- Yi-Tian Chen
- Department of Medical Oncology, Jinling Hospital, Nanjing, China
| | | | | |
Collapse
|
11
|
Tang J, Salama R, Gadgeel SM, Sarkar FH, Ahmad A. Erlotinib resistance in lung cancer: current progress and future perspectives. Front Pharmacol 2013; 4:15. [PMID: 23407898 PMCID: PMC3570789 DOI: 10.3389/fphar.2013.00015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 01/25/2013] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is the most common cancer in the world. Despite modern advancements in surgeries, chemotherapies, and radiotherapies over the past few years, lung cancer still remains a very difficult disease to treat. This has left the death rate from lung cancer victims largely unchanged throughout the past few decades. A key cause for the high mortality rate is the drug resistance that builds up for patients being currently treated with the chemotherapeutic agents. Although certain chemotherapeutic agents may initially effectively treat lung cancer patients, there is a high probability that there will be a reoccurrence of the cancer after the patient develops resistance to the drug. Erlotinib, the epidermal growth factor receptor (EGFR)-targeting tyrosine kinase inhibitor, has been approved for localized as well as metastatic non-small cell lung cancer where it seems to be more effective in patients with EGFR mutations. Resistance to erlotinib is a common observation in clinics and this review details our current knowledge on the subject. We discuss the causes of such resistance as well as innovative research to overcome it. Evidently, new chemotherapy strategies are desperately needed in order to better treat lung cancer patients. Current research is investigating alternative treatment plans to enhance the chemotherapy that is already offered. Better insight into the molecular mechanisms behind combination therapy pathways and even single molecular pathways may help improve the efficacy of the current treatment options.
Collapse
Affiliation(s)
- Joy Tang
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Rasha Salama
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Shirish M. Gadgeel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Fazlul H. Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| | - Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, MI, USA
| |
Collapse
|
12
|
de Mello RA, Marques DS, Medeiros R, Araújo AM. Epidermal growth factor receptor and K-Ras in non-small cell lung cancer-molecular pathways involved and targeted therapies. World J Clin Oncol 2011; 2:367-76. [PMID: 22087435 PMCID: PMC3215775 DOI: 10.5306/wjco.v2.i11.367] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 10/10/2011] [Accepted: 10/17/2011] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is currently the leading cause of cancer death in Western nations. Non-small cell lung cancer (NSCLC) represents 80% of all lung cancers, and adenocarcinoma is the predominant histological type. Despite the intensive research carried out on this field and therapeutic advances, the overall prognosis of these patients remains unsatisfactory, with a 5-year overall survival rate of less than 15%. Nowadays, pharmacogenetics and pharmacogenomics represent the key to successful treatment. Recent studies suggest the existence of two distinct molecular pathways in the carcinogenesis of lung adenocarcinoma: one associated with smoking and activation of the K-Ras oncogene and the other not associated with smoking and activation of the epidermal growth factor receptor (EGFR). The K-ras mutation is mainly responsible for primary resistance to new molecules which inhibit tyrosine kinase EGFR (erlotinib and gefitinib) and most of the EGFR mutations are responsible for increased tumor sensitivity to these drugs. This article aims to conduct a systematic review of the literature regarding the molecular pathways involving the EGFR, K-Ras and EGFR targeted therapies in NSCLC tumor behavior.
Collapse
Affiliation(s)
- Ramon Andrade de Mello
- Ramon Andrade de Mello, Dânia Sofia Marques, Department of Medical Oncology, Portuguese Oncology Institute, Porto 4200-072, Portugal
| | | | | | | |
Collapse
|
13
|
Li W, Mu D, Song L, Zhang J, Liang J, Wang C, Liu N, Tian F, Li X, Zhang W, Wang X. Molecular mechanism of silymarin-induced apoptosis in a highly metastatic lung cancer cell line anip973. Cancer Biother Radiopharm 2011; 26:317-24. [PMID: 21711112 DOI: 10.1089/cbr.2010.0892] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Silymarin, the main flavonoid constituent element extracted from Silybum marianum possessing antioxidant activity, is already known to be able to block the NF-κB activation process and result in cell apoptosis, implicating silymarin's potential to control cancer cell growth. MATERIALS AND METHODS In this study, based upon the above assumption, silymarin was administered to a highly metastatic lung cancer cell line Anip973 to test silymarin's role in cancer cell proliferation. RESULTS Silymarin had significant inhibitory effects on the proliferation of Anip973 cells in a dose-dependent and time-response manner within 48 hours. Silymarin can induce Anip973 apoptosis. CONCLUSIONS Silymarin may in vitro inhibit the proliferation of the human lung adenocarcinoma cell line Anip973 and induce apoptosis via the mitochondria-dependent caspase cascade pathway.
Collapse
Affiliation(s)
- Wenhai Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shannxi, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Korpanty G, Smyth E, Carney DN. Update on anti-angiogenic therapy in non-small cell lung cancer: Are we making progress? J Thorac Dis 2011; 3:19-29. [PMID: 22263059 PMCID: PMC3256499 DOI: 10.3978/j.issn.2072-1439.2010.11.11] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 11/18/2010] [Indexed: 01/08/2023]
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of death worldwide among patients diagnosed with malignancy. Despite new chemotherapy regimens and new cytotoxic combinations investigated in multiple clinical trials in recent years, no significant improvement in the prognosis of patients with lung cancer was achieved. The five-year survival rate for all patients diagnosed with NSCLC is about 15%, only 5% better than that of more than 40 years ago. New therapeutic approaches that target various different aspects of tumor progression and metastasis are of particular interest in to NSCLC patients. Drugs that block tumor vascularization (angiogenesis) or interfere with the activity of growth factor receptors and molecular pathways that are triggered by activation of these receptors are already used in clinical practice. In this review we will briefly discuss briefly the basic mechanisms of lung cancer angiogenesis, rationale for using drugs that block this process and present the most current recent data on their clinical efficacy.
Collapse
Affiliation(s)
- Grzegorz Korpanty
- Department of Medical Oncology, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | | | | |
Collapse
|
15
|
EGFR inhibitor C225 increases the radiosensitivity of human lung squamous cancer cells. Cancer Cell Int 2010; 10:39. [PMID: 20969791 PMCID: PMC2972262 DOI: 10.1186/1475-2867-10-39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 10/23/2010] [Indexed: 11/10/2022] Open
Abstract
Background The purpose of the present study is to investigate the direct biological effects of the epidermal growth factor receptor (EGFR) inhibitor C225 on the radiosensitivity of human lung squamous cancer cell-H520. H520 cells were treated with different dosage of 60Co γ ray irradiation (1.953 Gy/min) in the presence or absence of C225. The cellular proliferation, colony forming capacity, apoptosis, the cell cycle distribution as well as caspase-3 were analyzed in vitro. Results We found that C225 treatment significantly increased radiosensitivity of H-520 cells to irradiation, and led to cell cycle arrest in G1 phase, whereas 60Co γ ray irradiation mainly caused G2 phase arrest. H-520 cells thus displayed both the G1 and G2 phase arrest upon treatment with C225 in combination with 60Co γ ray irradiation. Moreover, C225 treatment significantly increased the apoptosis percentage of H-520 cells (13.91% ± 1.88%) compared with the control group (5.75% ± 0.64%, P < 0.05). Conclusion In this regard, C225 treatment may make H-520 cells more sensitive to irradiation through the enhancement of caspase-3 mediated tumor cell apoptosis and cell cycle arrest.
Collapse
|
16
|
Cho Y, Park MJ, Park M, Min SS, Yee J, Kim C, Han MS, Han SH. Effects of CAY10404 on the PKB/Akt and MAPK pathway and apoptosis in non-small cell lung cancer cells. Respirology 2009; 14:850-8. [PMID: 19703066 DOI: 10.1111/j.1440-1843.2009.01563.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Lung cancer is the most common cause of cancer death in men and women worldwide. The mechanism of cell death induced by CAY10404, a highly selective cyclooxygenase-2 inhibitor, was evaluated in three non-small cell lung cancer (NSCLC) cell lines (H460, H358, H1703). METHODS To measure the effects of CAY10404 on proliferation of NSCLC cells, 3 x 10(3) cells/well were plated in 96-well plates and allowed to adhere overnight at 37 degrees C. After treatment with CAY10404 for 3 days, cell proliferation was measured by the 3- (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. In the H460 NSCLC cells, evidence of apoptosis was sought using the terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labelling (TUNEL) assay and western blot analysis. RESULTS Treatment with CAY10404 in the range of 10-100 microM caused dose-dependent growth inhibition, with an average 50% inhibitory concentration (IC(50)) of 60-100 micromol/L, depending on the cell line. Western blot analysis of CAY10404-treated cells showed cleavage of poly (ADP-ribose) polymerase (PARP) and procaspase-3, signifying caspase activity and apoptotic cell death. CAY10404 treatment inhibited the phosphorylation of Akt, glycogen synthase kinase-3beta and extracellular signal-regulated kinases 1/2 in H460 and H358 cells. CONCLUSIONS These results suggest that CAY10404 is a potent inducer of apoptosis in NSCLC cells, and that it may act by suppressing multiple protein kinase B/Akt and mitogen-activated protein kinase pathways.
Collapse
Affiliation(s)
- Yongseon Cho
- Department of Internal Medicine, Medical Sciences Research Institute, School of Medicine, Eulji University, Jung-Gu, Daejeon, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Paleari L, Sessa F, Catassi A, Servent D, Mourier G, Doria-Miglietta G, Ognio E, Cilli M, Dominioni L, Paolucci M, Calcaterra A, Cesario A, Margaritora S, Granone P, Russo P. Inhibition of non-neuronal alpha7-nicotinic receptor reduces tumorigenicity in A549 NSCLC xenografts. Int J Cancer 2009; 125:199-211. [PMID: 19326440 DOI: 10.1002/ijc.24299] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nicotinic acetylcholine receptors (nAChR) are expressed on bronchial epithelial and non-small cell lung cancer cells and are involved in cell growth regulation. Nicotine (classical nAChR agonist) induced cell proliferation, whereas nAChR antagonists, d- tubocurarine or alpha-cobratoxin (alpha-CbT), induced cell death. In the current study, we further explored the antitumor potential mechanisms and activities of alpha-CbT. NOD/SCID mice were grafted intraperitoneally or orthotopically and treated with alpha-CbT. alpha-CbT treatment [0.04 ng/kg or 0.12 ng/kg] induced a strong reduction in tumor size ( approximately 90%) in comparison with mice treated with the vehicle alone. Tumor inhibition was related to severe induction of apoptosis. Moreover, neoangiogenesis was strongly inhibited (reduction of cells positive to vascular endothelial growth factor and CD31). Biochemical analyses of the cells, isolated by the primary lung tumor in alpha-CbT-treated mice, showed apoptosis features characterized by: (i) inhibition of BAD phosphorylation at Ser(112) and Ser(136); (ii) BAD dissociation from 14-3-3; (iii) BAD association with BCL-XL; and (iv) cleavage of caspase-9. Moreover, these cells were unable to grow in soft agar and develop tumor, when reinjected into mice. The small interfering RNA-mediated silencing of the alpha7-nAChR gene confirmed that alpha-CbT specifically inhibited the alpha7-nAChR-mediated survival pathway in A549 cells. Furthermore, the specificity of alpha-CbT is reinforced by the lack of effect of short chain toxin (Erabutoxin-a). Once more, the no effect of the low-affinity R33E-modified alpha-CbT strengthened the specificity of this inhibition. Although alpha7-nAChR antagonists, such as alpha-CbT, are unlikely to be a primary therapy, it may provide lead compounds for the design of clinically useful drugs.
Collapse
Affiliation(s)
- Laura Paleari
- Lung Cancer Unit, National Cancer Research Institute, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kishino D, Kiura K, Takigawa N, Katayama H, Kuyama S, Sato K, Okada T, Ohashi K, Tanimoto M. Effect of gefitinib on N-nitrosamine-4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone induced lung tumorigenesis in A/J mice. Lung Cancer 2009; 65:284-9. [PMID: 19152984 DOI: 10.1016/j.lungcan.2008.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 11/26/2008] [Accepted: 11/27/2008] [Indexed: 11/30/2022]
Abstract
Gefitinib is an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). N-Nitrosamine-4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a potent carcinogen found in tobacco smoke, induces lung tumors in A/J mice. NNK induces cellular transformation resulting in the over-expression of EGFR. Accordingly, EGFR may be a target for cancer prevention. In this study, we investigated the effect of gefitinib on NNK-induced tumorigenesis and the carcinogenicity of gefitinib in A/J mice. A total of 180 four-week-old female A/J mice were randomly divided into six groups: group 1 (controls), treated with deionized water; group 2, treated with 5 mg/kg p.o. gefitinib; group 3, treated with 50 mg/kg p.o. gefitinib (to test the carcinogenicity of gefitinib); group 4 (controls for NNK treatment), treated with deionized water; group 5, treated with 5 mg/kg p.o. gefitinib; and group 6, treated with 50 mg/kg p.o. gefitinib and injected with NNK once at 8 weeks of age to test the chemopreventive activity of gefitinib. Gefitinib was given once a day, 5 days a week by gavage, beginning at 4 weeks of age and continuing for 26 weeks. All mice were sacrificed at 30 weeks of age. The multiplicities of the NNK-induced lung tumors were significantly suppressed in a dose-dependent manner. Gefitinib had no effect on body weight at a low dose. The administration of gefitinib alone for 26 weeks did not induce tumorigenesis; instead, it significantly suppressed the incidence of spontaneous tumors in the mice, in contrast with other anti-cancer agents. Gefitinib did not induce lung fibrosis when compared with control mice by Azan-Mallory staining. Our results suggest that gefitinib has a weak but significant chemopreventive effect with no carcinogenicity or pulmonary toxicity in A/J mice.
Collapse
Affiliation(s)
- Daizo Kishino
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Qian J, Zou Y, Rahman JS, Lu B, Massion PP. Synergy between phosphatidylinositol 3-kinase/Akt pathway and Bcl-xL in the control of apoptosis in adenocarcinoma cells of the lung. Mol Cancer Ther 2009; 8:101-9. [PMID: 19139118 PMCID: PMC3110728 DOI: 10.1158/1535-7163.mct-08-0973] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adenocarcinomas of the lung commonly show an increase in the activity of phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, yet many are resistant to apoptosis induced by the inhibition of PI3K. We hypothesized that Bcl-xL would have a synergistic effect on the apoptotic response induced by inhibition of the PI3K/Akt pathway in lung adenocarcinoma. To test this, we examined the effect of the PI3K inhibitor (LY294002) on lung adenocarcinoma cell lines expressing varying levels of Bcl-xL. We found that cells that overexpress Bcl-xL are resistant to LY294002-induced apoptosis, whereas cells that express little Bcl-xL readily are not. Restoring Bcl-xL expression in cells that express low level of Bcl-xL conferred resistance to apoptosis in response to LY294002. The simultaneous inhibition of the PI3K/Akt pathway by LY294002 or Akt1 small interfering RNA and Bcl-xL function by ABT-737 or Bcl-xL small interfering RNA greatly enhanced the apoptotic response. Moreover, this response was associated with the induction of proapoptotic BH3-only Bcl-2 family member Bim. Our data suggest that PI3K/Akt and Bcl-xL pathways control cell death in lung adenocarcinoma cells in a synergistic manner. Modulation of Bcl-xL expression may represent one important strategy to optimize the efficacy of therapeutic agents targeting the PI3K/Akt pathway in adenocarcinoma of the lung.
Collapse
Affiliation(s)
- Jun Qian
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, Nashville VA Medical Center, Nashville, TN
| | - Yong Zou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, Nashville VA Medical Center, Nashville, TN
| | - Jamshedur S.M. Rahman
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, Nashville VA Medical Center, Nashville, TN
| | - Bo Lu
- Department of Radiation Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, Nashville VA Medical Center, Nashville, TN
| | - Pierre P. Massion
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, Nashville VA Medical Center, Nashville, TN
| |
Collapse
|
20
|
Rossi A, Maione P, Palazzolo G, Sacco PC, Ferrara ML, Falanga M, Gridelli C. New Targeted Therapies and Small-Cell Lung Cancer. Clin Lung Cancer 2008; 9:271-9. [DOI: 10.3816/clc.2008.n.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
21
|
Lee JM, Yanagawa J, Peebles KA, Sharma S, Mao JT, Dubinett SM. Inflammation in lung carcinogenesis: new targets for lung cancer chemoprevention and treatment. Crit Rev Oncol Hematol 2008; 66:208-17. [PMID: 18304833 PMCID: PMC2483429 DOI: 10.1016/j.critrevonc.2008.01.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 12/23/2007] [Accepted: 01/16/2008] [Indexed: 10/22/2022] Open
Abstract
Lung carcinogenesis is a complex process involving the acquisition of genetic mutations that confer cancer development and the malignant phenotype, and is critically linked to apoptosis resistance, unregulated proliferation, invasion, metastasis, and angiogenesis. Epithelial mesenchymal transition (EMT) in cancer is an unregulated process in a host environment with deregulated inflammatory response that impairs cell-mediated immunity and permits cancer progression. Given the immunosuppressive tumor environment, strategies to reverse these events by stimulating host immune responses are an important area of investigation. Cyclooxygenase 2 (COX-2) and its downstream signaling pathways are potential targets for lung cancer chemoprevention and therapy. Clinical trials are underway to evaluate COX-2 inhibitors as adjuvants to chemotherapy in patients with lung cancer and to determine efficacy in prevention of bronchogenic carcinoma. The understanding of molecular mechanisms involved in inflammation and lung carcinogenesis provide insight for new drug development that target reversible, non-mutational events in the chemoprevention and treatment of lung cancer.
Collapse
Affiliation(s)
- Jay M. Lee
- Division of Cardiothoracic Surgery, Department of Surgery, UCLA Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, Phone: (310) 794-7333, Fax: (310) 794-7335, E-mail:
| | - Jane Yanagawa
- Division of Pulmonary and Critical Care Medicine and Hospitalists, Department of Medicine, UCLA Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, Phone: (310) 267-2725, Fax: (310) 267-2829, E-mail:
| | - Katherine A. Peebles
- Division of Pulmonary and Critical Care Medicine and Hospitalists, Department of Medicine, UCLA Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, Phone: (310) 794-6566, Fax: (310) 267-2725, E-mail:
| | - Sherven Sharma
- Division of Pulmonary and Critical Care Medicine and Hospitalists, Department of Medicine, UCLA Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, West Los Angeles VA, Los Angeles, CA 90073, Phone: (310) 478-3711 x 41863, Fax: (310) 268-4164, E-mail:
| | - Jenny T. Mao
- Division of Pulmonary and Critical Care Medicine and Hospitalists, Department of Medicine, UCLA Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Phone: (310) 825-3100, Fax: (310) 206-8622, E-mail:
| | - Steven M. Dubinett
- Division of Pulmonary and Critical Care Medicine and Hospitalists, Department of Medicine, UCLA Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, Phone: (310) 267-2725, Fax: (310) 267-2829, E-mail:
| |
Collapse
|
22
|
Chiappori AA, Haura E, Rodriguez FA, Boulware D, Kapoor R, Neuger AM, Lush R, Padilla B, Burton M, Williams C, Simon G, Antonia S, Sullivan DM, Bepler G. Phase I/II study of atrasentan, an endothelin A receptor antagonist, in combination with paclitaxel and carboplatin as first-line therapy in advanced non-small cell lung cancer. Clin Cancer Res 2008; 14:1464-9. [PMID: 18316570 DOI: 10.1158/1078-0432.ccr-07-1508] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Endothelins and their cell membrane receptors (ET(A)R and ET(B)R) are implicated in neoplastic pathogenesis. atrasentan, a potent, selective ET(A)R antagonist, has a direct effect on tumor proliferation, apoptosis, and angiogenesis. This study was designed to assess the influence of atrasentan on paclitaxel pharmacokinetics and to determine the safety and efficacy of atrasentan in combination with paclitaxel-carboplatin. EXPERIMENTAL DESIGN Chemonaive patients with stage IIIB (malignant pleural effusion) and IV non-small cell lung cancer were enrolled. Toxicity and response were determined using the National Cancer Institute Common Toxicity Criteria version 2.0 and Response Evaluation Criteria in Solid Tumors criteria, respectively. Treatment consisted of paclitaxel (225 mg/m(2)) and carboplatin (area under the curve, 6) administered on day 1 every 3 weeks. A fixed 10 mg daily oral dose ofAtrasentan was administered continuously, starting on day 4 of cycle 1. Paclitaxel clearance was calculated during the first two cycles (pre- and post-atrasentan) in the first 10 patients. RESULTS All 44 patients were evaluable for survival, toxicity, and response. No significant change in mean paclitaxel clearance was detected (mean +/- SD, 21.2 +/- 4.5 L/h versus 21.3 +/- 4.9 L/h) for pre- and post-atrasentan values, respectively (P = 0.434). Grade 3/4 toxicities > or = 10% were lymphopenia (22.7%), neutropenia (20.5%), dyspnea (11.4%), and hyperglycemia (11.4%). Response rate was 18.2%, with progression-free survival of 4.2 months, median survival of 10.6 months, and 1-year survival of 43%. CONCLUSION Atrasentan plus paclitaxel-carboplatin was safe and well tolerated, with no apparent paclitaxel-atrasentan pharmacokinetic interaction. Efficacy and survival in advanced non-small cell lung cancer were comparable with studies of chemotherapy alone.
Collapse
Affiliation(s)
- Alberto A Chiappori
- Thoracic Oncology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Khawli LA, Hu P, Epstein AL. Cytokine, chemokine, and co-stimulatory fusion proteins for the immunotherapy of solid tumors. Handb Exp Pharmacol 2008:291-328. [PMID: 18071951 DOI: 10.1007/978-3-540-73259-4_13] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
This chapter describes the generation of novel reagents for the treatment of cancer using fusion proteins constructed with natural ligands of the immune system. Immunotherapy is a powerful therapeutic modality that has not been fully harnessed for the treatment of cancer. We and others have hypothesized that if the proper immunoregulatory ligands can be targeted to the tumor, an effective immune response can be mounted to treat both established primary tumors and distant metastatic lesions. Though it is generally believed that immunotherapy has the potential to treat only residual disease, we offer evidence that this approach can, by itself, destroy large tumor masses and produce lasting remissions of experimental solid tumors. From these studies, three major classes of immune activators, namely, cytokines, chemokines, and costimulatory molecules, have been shown to generate antitumor responses in animal models. In addition, the reversal of immune tolerance by the deletion of T regulatory (Treg) cells has been shown to be equally important for effective immunotherapy. In an attempt to identify reagents that can provide an enhanced immune stimulation and treatment of cancer, our laboratory has developed a novel monoclonal antibody targeting approach, designated Tumor Necrosis Therapy (TNT), which utilizes stable intracellular antigens present in all cell types but which are only accessible in dead and/or dying cells. Since tumors contain necrotic and degenerating regions that account for 30-80% of the tumor mass, this targeting approach can be used to deliver therapeutic reagents to the core of tumors, a site abundant in tumor antigens. In our first set of reagents, a panel of cytokine fusion proteins was genetically engineered using monoclonal antibody chimeric TNT-3 (chTNT-3) directed against necrotic regions of tumors (single-stranded DNA) fused with IL-2, or GM-CSF, or TNFalphaa, or IFNgamma. Tested against different solid tumors, these reagents were found to mount an effective although transient immune response to tumor especially when used in combination. To improve upon these results, additional chTNT-3 fusion proteins using the liver-expression chemokine (LEC) and the costimulatory molecule B7.1 were constructed. Both of these reagents were found to work significantly better than the above cytokine fusion proteins due to their ability to stimulate multiple arms of the immune system deemed useful for cancer immunotherapy. Finally, the Tumor Necrosis Factor Superfamily (TNFSF) gene DC137L was used to generate chTNT-3 antibody (targeted) and soluble Fc (untargeted) fusion proteins. When used alone, both forms of costimulatory fusion proteins were found to produce in a s dose-dependent manner, complete regression of murine solid tumors. Evidence is presented to show that Treg cells play an important role in suppressing antitumor immunity since the deletion of these cells, when used in combination with LEC or costimulatory fusion proteins, produced profound and effective treatment with sustained memory. It is hoped that these data will further the preclinical development of soluble Fc and antibody based fusion proteins fro the immunotherapy of cancer.
Collapse
Affiliation(s)
- L A Khawli
- Department of Pathology, Keck School of Medicine at the University of Southern California, 2011 Zonal Avenue, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
24
|
Bigioni M, Benzo A, Irrissuto C, Lopez G, Curatella B, Maggi CA, Manzini S, Crea A, Caroli S, Cubadda F, Binaschi M. Antitumour effect of combination treatment with Sabarubicin (MEN 10755) and cis-platin (DDP) in human lung tumour xenograft. Cancer Chemother Pharmacol 2007; 62:621-9. [PMID: 18038274 DOI: 10.1007/s00280-007-0645-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Accepted: 11/09/2007] [Indexed: 11/29/2022]
Abstract
PURPOSE Sabarubicin (MEN 10755), a new disaccaride anthracycline, has shown greater efficacy than Doxorubicin in a large panel of preclinical models and now it is in phase II clinical trials. Its promising antitumour activity promoted considerable interest to combine Sabarubicin with other antitumour agents. Thus, the purpose of this study was to evaluate in vitro cytotoxic effects and in vivo antitumour activities produced by the combination of Sabarubicin and cisplatin (DDP). METHODS The antitumour effect of Sabarubicin and DDP association was investigated, in vitro and in vivo, in preclinical models of lung cancer i.e.: the non-small cell lung carcinoma (NSCLC) H460 and the small-cell lung carcinoma (SCLC) GLC4 in terms of synergism, additivity or antagonism in order to establish the best schedule for the combined treatment. Further, the correlation between antitumour activity and the pharmacokinetic parameters of the studied combination was also evaluated. RESULTS The drug combination in vitro was in general more cytotoxic than the single drug alone, indicating the presence of a synergistic effect in both tumour cell lines. Also, in the xenograft experiments a superior antitumoral effect was observed when Sabarubicin was combined with DDP. The antitumour efficacy of Sabarubicin (6 mg/kg q4d x 5) combined with DDP (6 mg/kg q4d x 3) greatly depended on the schedule of administration. In H460 tumour line, the sequential combination was more effective than the simultaneous administration of the two agents, although the antitumour efficacy was not dependent on the sequence of combination. On the other hand, a strong sequence-dependent effect was observed when Sabarubicin was combined with DDP in SCLC, GLC4. In particular, the highest value of LCK = 6.7 was obtained when administration of DDP followed by 24 h that of Sabarubicin. Pharmacokinetics of Sabarubicin in combination with DDP was evaluated at 6 mg/kg for both drugs with different sequential schedule. The experimental data showed no evidence for pharmacokinetics drug-drug interaction. CONCLUSION These preclinical results indicate the potential for a strong antitumour activity in lung tumours of the combination Sabarubicin and DDP. In particular, in SCLC the best response should be given by a sequence with administration of Sabarubicin followed 24 h later by that of DDP. Clinical trials based on these results are ongoing.
Collapse
Affiliation(s)
- M Bigioni
- Department of Pharmacology and Pharmacokinetics, Menarini Ricerche S.p.A, Via Tito Speri 10, 00040, Rome, Pomezia, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ho C, Gautschi O, Lara PN, Gandara DR, Davies AM. Lung Cancer Vaccines. Lung Cancer 2007. [DOI: 10.3109/9781420020359.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Lee JM, Mao JT, Krysan K, Dubinett SM. Significance of cyclooxygenase-2 in prognosis, targeted therapy and chemoprevention of NSCLC. Future Oncol 2007; 3:149-53. [PMID: 17381414 DOI: 10.2217/14796694.3.2.149] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Evaluation of: Tsubochi H, Nobuyuki S, Hiyama M et al.: Combined analysis of cyclooxygenase-2 expression with p53 and Ki-67 in nonsmall cell lung cancer. Ann. Thorac. Surg. 82(4), 1198–1204 (2006).The report by Tsubochi and colleagues adds to the growing evidence indicating that tumor cyclo-oxygenase (Cox)-2 has a multifaceted role in conferring the malignant phenotype of non-small-cell lung cancer (NSCLC), and provides insight into the use of markers to provide prognostic information. Cox-2 has been implicated in apoptosis resistance, angiogenesis, decreased host immunity and enhanced invasion and metastasis, and, thus, has a critical involvement in carcinogenesis. This study, as well as others, has contributed to providing an insight into opportunities for targeted therapies in NSCLC. Cox-2 is one of the novel targets being studied for lung cancer therapy and chemoprevention.
Collapse
Affiliation(s)
- Jay M Lee
- Division of Cardiothoracic Surgery, Department of Surgery, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
27
|
Abstract
Targeted therapies will advance the treatment of NSCLC as we improve our understanding of the underlying biology of NSCLC and enhance our ability to clinically target the optimal therapy to an individual's cancer. Ongoing translational research including tissue arrays, genomic, and proteomic studies will help to identify clinically useful biomarkers that will allow further classification of NSCLC and may allow accurate prediction of response to specific chemotherapeutic regimens. Advances in targeted therapy in NSCLC are already yielding exciting results, and promises to become an increasingly important adjunct to surgical management of NSCLC.
Collapse
Affiliation(s)
- Philip W Smith
- University of Virginia, School of Medicine, P.O. Box 800679, Lee Street, Charlottesville, VA 22908-0679, USA
| | | | | |
Collapse
|
28
|
Pizzo B. New Directions in Oncology Nursing Care: Focus on Gefitinib in Patients With Lung Cancer. Clin J Oncol Nurs 2007; 8:385-92. [PMID: 15354925 DOI: 10.1188/04.cjon.385-392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Treatment of solid tumors with chemotherapy regimens commonly is associated with debilitating or life-threatening side effects. Careful patient management, appropriate and prompt management of side effects, and interruption of therapy frequently are required for patients receiving chemotherapy. Furthermore, the systemic toxicity associated with chemotherapy may result in irreversible and incapacitating side effects, such as peripheral neuropathy, that lead to poor quality of life in patients. Gefitinib (Iressa, ZD1839, AstraZeneca Pharmaceuticals LP, Wilmington, DE) is a biologically based, molecular targeted therapy with a novel mechanism of action: selective inhibition of the epidermal growth factor receptortyrosine kinase (EGFR-TK) activity. Once-daily oral treatment with gefitinib is well tolerated. In clinical trials, treatment with gefitinib resulted in durable tumor responses and improvement in lung cancer-related symptoms in patients with advanced non-small cell lung cancer who had received prior chemotherapy. Trials are under way to explore the full potential of gefitinib and additional EGFR-TK inhibitors for other solid tumors and in other treatment settings, including prevention. Biologically based, molecular-targeted therapies such as gefitinib are providing new treatment options for patients and adding a new dimension to clinical practice for oncology nurses.
Collapse
Affiliation(s)
- Barbara Pizzo
- Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
29
|
Park JS, Jun HJ, Cho MJ, Cho KH, Lee JS, Zo JI, Pyo H. Radiosensitivity enhancement by combined treatment of celecoxib and gefitinib on human lung cancer cells. Clin Cancer Res 2006; 12:4989-99. [PMID: 16914589 DOI: 10.1158/1078-0432.ccr-05-2259] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To characterize the radiation-enhancing effects and underlying mechanisms of combined treatment with celecoxib, a cyclooxygenase-2 selective inhibitor, and gefitinib, an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, in human lung cancer cells. EXPERIMENTAL DESIGN Clonogenic cytotoxicity assays and clonogenic radiation survival assays after treatments with celecoxib and gefitinib with or without radiation were done on three human lung cancer cell lines. Synergisms after combined treatment with celecoxib, gefitinib, and radiation were investigated using isobologram and statistical analyses according to an independent action model. Alterations in apoptosis and cell cycle were measured to identify the mechanisms underlying the cell killing or radiation-enhancing effects of celecoxib and gefitinib combination treatment. Western blots for phosphorylated EGFR, EGFR, cyclooxygenase-2, and G(2) checkpoint molecules were conducted after treatment with celecoxib and/or gefitinib with or without radiation. RESULTS Combination celecoxib, gefitinib, and radiation treatments were shown to be synergistic in causing clonogenic cell deaths in all cell lines tested, but the nature of synergism was cell type specific. The combined drug treatments induced apoptosis in an additive manner in A549 cells and in a synergistic manner in NCI-H460 and VMRC-LCD cells. Celecoxib or gefitinib attenuated radiation-induced G(2)-M arrest, and combined drug treatment additively attenuated radiation-induced G(2)-M arrest in all cell lines. Radiation-induced checkpoint kinase (Chk) 1 and Chk2 phosphorylation were inhibited by celecoxib and gefitinib treatment, respectively. CONCLUSIONS Combined celecoxib and gefitinib treatments were shown to synergistically enhance the effect of radiation on lung cancer cells. The mechanisms underlying these synergistic effects seem to involve the synergistic enhancement of apoptosis and cooperative attenuation of radiation-induced G(2)-M arrest, possibly via Chk1 and Chk2 inhibition, by the combined drug treatments.
Collapse
Affiliation(s)
- Ji Sun Park
- Research Institute and Hospital, National Cancer Center, Goyang, Gyeonggi, Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Chiappori AA, Ellis PM, Hamm JT, Bitran JD, Eiseman I, Lovalvo J, Burnett D, Olson S, Lenehan P, Zinner RG. A Phase I Evaluation of Oral CI-1033 in Combination with Paclitaxel and Carboplatin as First-Line Chemotherapy in Patients with Advanced Non-small Cell Lung Cancer. J Thorac Oncol 2006. [DOI: 10.1016/s1556-0864(15)31635-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
A Phase I Evaluation of Oral CI-1033 in Combination with Paclitaxel and Carboplatin as First-Line Chemotherapy in Patients with Advanced Non-small Cell Lung Cancer. J Thorac Oncol 2006. [DOI: 10.1097/01243894-200611000-00015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Pentheroudakis G, Razis E, Athanassiadis A, Pavlidis N, Fountzilas G. Paclitaxel-carboplatin combination chemotherapy in advanced breast cancer: accumulating evidence for synergy, efficacy, and safety. Med Oncol 2006; 23:147-60. [PMID: 16720915 DOI: 10.1385/mo:23:2:147] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Revised: 11/30/1999] [Accepted: 10/04/2005] [Indexed: 11/11/2022]
Abstract
Patients with metastatic breast cancer receive multiple lines of cytotoxic chemotherapy, with taxane and anthracycline-based regimens being the most active. Anthracyclines carry the risk of significant cardiotoxicity at high cumulative doses and when combined with trastuzumab, an anti-HER2 antibody. Carboplatin has shown promising single-agent activity in advanced breast cancer, is not a P-glycoprotein substrate, and is conveniently administered on an outpatient basis. Preclinical experiments demonstrated schedule-dependent synergistic cytotoxic effects of the paclitaxel first/carboplatin last (PC) combination. Pharmacokinetic parameters of paclitaxel and carboplatin were studied by Hellenic Cooperative Oncology Group (HECOG) and no significant interaction or correlation with clinical parameters were found. We assessed PC both as salvage as well as first-line treatment of advanced breast cancer patients in phase II studies which disclosed 40-60% response rates and median survival times of 12-20 mo with manageable toxicity. These results were confirmed by other groups and prompted us to the first randomized phase III trial comparing PC to the standard of epirubicin/paclitaxel (EP), a trial that showed equivalent efficacy and tolerable toxicity for PC. Registry retrospective analysis identified factors prognostic for improved outcome: good performance status, low tumor burden, lack of anthracycline exposure and of hormonal maintenance therapy. PC combinations with HER1 or HER2 modulators are being evaluated both by HECOG and by international groups. Paclitaxel coupled with carboplatin provides an alternative therapeutic option for anthracycline-exposed patients and warrants further clinical research in the direction of anthracycline-free management of metastatic breast cancer.
Collapse
Affiliation(s)
- G Pentheroudakis
- Department of Medical Oncology, Ioannina University Hospital, Ioannina, Greece.
| | | | | | | | | |
Collapse
|
33
|
Azim HA, Ganti AK. Targeted therapy in advanced non-small cell lung cancer (NSCLC): where do we stand? Cancer Treat Rev 2006; 32:630-6. [PMID: 17034953 DOI: 10.1016/j.ctrv.2006.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Revised: 07/18/2006] [Accepted: 07/19/2006] [Indexed: 11/17/2022]
Abstract
Cytotoxic chemotherapy has helped improve the outcomes in patients with advanced non-small cell lung cancer (NSCLC), but we seem to have reached a plateau with respect to the benefit obtained. Also, a large subset of elderly patients and those with a poor performance status cannot tolerate these drugs at recommended doses. There is a growing need to incorporate newer drugs with different mechanisms of action and better safety profile. The epidermal growth factor receptor family (EGFR) and vascular endothelial growth factor (VEGF) have been identified as potential targets and agents acting specifically against these targets have been developed with the hope of improving outcomes. Although recent data with the small molecule EGFR tyrosine kinase inhibitors have been disappointing, there have been instances of dramatic responses thereby raising questions about the ideal patient to whom these drugs should be administered. Cetuximab, the anti-EGFR antibody has shown promising results. Bevacizumab, the anti-VEGF antibody was the first drug to demonstrate a survival benefit in first line treatment when added to chemotherapy. This review will briefly discuss the important trials using these targeted agents in advanced NSCLC.
Collapse
Affiliation(s)
- Hatem A Azim
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | | |
Collapse
|
34
|
Sonnweber B, Dlaska M, Skvortsov S, Dirnhofer S, Schmid T, Hilbe W. High predictive value of epidermal growth factor receptor phosphorylation but not of EGFRvIII mutation in resected stage I non-small cell lung cancer (NSCLC). J Clin Pathol 2006; 59:255-9. [PMID: 16505275 PMCID: PMC1860345 DOI: 10.1136/jcp.2005.027615] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Overexpression and mutation of epidermal growth factor regulator (EGFR) are frequently found in the carcinogenesis of non-small cell lung cancer (NSCLC). Because targeting of this receptor has proven therapeutic efficacy, studying EGFR has become a matter of particular scientific interest. The present study analysed the EGFR receptor, rate of EGFRvIII mutations, and rate of activated phosphorylated EGFR (pEGFR) by immunohistochemistry on cryostat sections. METHODS Surgically obtained tumour specimens of a series of 78 NSCLC patients and 66 adjacent tumour free specimens were examined immunohistochemically using monoclonal antibodies to stain EGFR, pEGFR, and EGFRvIII. RESULTS EGFRvIII and pEGFR expression was found in 42% and 26% of the tumours respectively and both were increased significantly compared with tumour free samples. EGFR, pEGFR, and EGFRvIII expression did not correlate with any of the previously tested markers (c-erbB-2, c-erbB-3, p53, ki-67, and microvessel density). Similar distributions of immunohistochemical profiles were seen, regardless of histological subtype, age, or sex. In stage I patients, EGFR phosphorylation at tyrosine residue 845 proved to be an independent prognostic factor. CONCLUSION Because pEGFR correlated with poor prognosis, it can be speculated that it plays a crucial biological role in the pathogenesis of NSCLC.
Collapse
Affiliation(s)
- B Sonnweber
- Department of Internal Medicine, Division of General Internal Medicine, Medical University Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
35
|
Jensen AD, Münter MW, Bischoff H, Haselmann R, Timke C, Krempien R, Sterzing F, Nill S, Heeger S, Hoess A, Haberkorn U, Huber PE, Steins M, Thomas M, Debus J, Herfarth KK. Treatment of non-small cell lung cancer with intensity-modulated radiation therapy in combination with cetuximab: the NEAR protocol (NCT00115518). BMC Cancer 2006; 6:122. [PMID: 16681848 PMCID: PMC1524801 DOI: 10.1186/1471-2407-6-122] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 05/08/2006] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Even today, treatment of Stage III NSCLC still poses a serious challenge. So far, surgical resection is the treatment of choice. Patients whose tumour is not resectable or who are unfit to undergo surgery are usually referred to a combined radio-chemotherapy. However, combined radio-chemotherapeutic treatment is also associated with sometimes marked side effects but has been shown to be more efficient than radiation therapy alone. Nevertheless, there is a significant subset of patients whose overall condition does not permit administration of chemotherapy in a combined-modality treatment. It could be demonstrated though, that NSCLCs often exhibit over-expression of EGF-receptors hence providing an excellent target for the monoclonal EGFR-antagonist cetuximab (Erbitux) which has already been shown to be effective in colorectal as well as head-and-neck tumours with comparatively mild side-effects. METHODS/DESIGN The NEAR trial is a prospective phase II feasibility study combining a monoclonal EGF-receptor antibody with loco-regional irradiation in patients with stage III NSCLC. This trial aims at testing the combination's efficacy and rate of development of distant metastases with an accrual of 30 patients. Patients receive weekly infusions of cetuximab (Erbitux) plus loco-regional radiation therapy as intensity-modulated radiation therapy. After conclusion of radiation treatment patients continue to receive weekly cetuximab for 13 more cycles. DISCUSSION The primary objective of the NEAR trial is to evaluate toxicities and feasibility of the combined treatment with cetuximab (Erbitux) and IMRT loco-regional irradiation. Secondary objectives are remission rates, 3-year-survival and local/systemic progression-free survival.
Collapse
Affiliation(s)
- AD Jensen
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
- Clinical Co-operation Unit Radiation Oncology, German Cancer Research Centre (DKFZ), INF 280, 69120 Heidelberg, Germany
| | - MW Münter
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
- Clinical Co-operation Unit Radiation Oncology, German Cancer Research Centre (DKFZ), INF 280, 69120 Heidelberg, Germany
| | - H Bischoff
- Dept. of Medical Oncology, Thoraxklinik Heidelberg, Amalienstr. 5, 69126 Heidelberg, Germany
| | - R Haselmann
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
| | - C Timke
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
| | - R Krempien
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
| | - F Sterzing
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
| | - S Nill
- Dept. of Medical Physics, German Cancer Research Centre (DKFZ), INF 280, 69120 Heidelberg, Germany
| | | | - A Hoess
- Dept. of Medical Physics, German Cancer Research Centre (DKFZ), INF 280, 69120 Heidelberg, Germany
| | - U Haberkorn
- Dept. of Nuclear Medicine, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
| | - PE Huber
- Clinical Co-operation Unit Radiation Oncology, German Cancer Research Centre (DKFZ), INF 280, 69120 Heidelberg, Germany
| | - M Steins
- Dept. of Medical Oncology, Thoraxklinik Heidelberg, Amalienstr. 5, 69126 Heidelberg, Germany
| | - M Thomas
- Dept. of Medical Oncology, Thoraxklinik Heidelberg, Amalienstr. 5, 69126 Heidelberg, Germany
| | - J Debus
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
| | - KK Herfarth
- Dept. of Radiation Oncology, Clinical Radiology, University of Heidelberg Medical School, INF 400, 69120 Heidelberg, Germany
- Clinical Co-operation Unit Radiation Oncology, German Cancer Research Centre (DKFZ), INF 280, 69120 Heidelberg, Germany
| |
Collapse
|
36
|
Norsa A, Martino V. Somatostatin, retinoids, melatonin, vitamin D, bromocriptine, and cyclophosphamide in advanced non-small-cell lung cancer patients with low performance status. Cancer Biother Radiopharm 2006; 21:68-73. [PMID: 16480333 DOI: 10.1089/cbr.2006.21.68] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The prognosis of low performance status (PS) patients with advanced non-small-cell-lung cancer (NSCLC) is dismal. In these patients, we have determined the survival, clinical benefits, and toxicity of a multidrug regimen, based on cyclophosphamide and biotherapeutical agents. METHODS Patients with a diagnosis of stage IIIB or stage IV NSCLC, no previous surgery or chemoradiotherapy, and an Eastern Cooperative Oncology Group (ECOG) PS equal to or greater than 2 received a daily combination of somatostatin, retinoids, melatonin, vitamin D, bromocriptine, and cyclophosphamide. RESULTS Twenty-eight (28) patients were enrolled. The median age was 64 years (range, 35-74). The PS was 2 and 3 in 78.6% and 21.4% of patients, respectively. The median overall survival (intent-to-treat analysis) was 12.9 months (range, 1.5-33.5 months), The overall survival rates at 1 and 2 years were 51.2% and 21.1%, respectively. The side-effects were very mild, mostly consisting of diarrhoea, nausea/vomiting, and drowsiness of grade 1-2. Most patients experienced an improvement of both respiratory (cough and dyspnoea) and general (pain, fatigue, and insomnia) symptoms. CONCLUSIONS Low PS patients with advanced NSCLC may benefit from a combination of somatostatin, retinoids, melatonin, vitamin D, bromocriptine, and cyclophosphamide, in terms of survival and quality of life, with very low side-effects.
Collapse
Affiliation(s)
- Achille Norsa
- Thoracic Surgery Unit, Ospedale Maggiore Azienda Ospedaliera, Verona, Italy.
| | | |
Collapse
|
37
|
Davies AM, Ho C, Lara PN, Mack P, Gumerlock PH, Gandara DR. Pharmacodynamic Separation of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors and Chemotherapy in Non–Small-Cell Lung Cancer. Clin Lung Cancer 2006; 7:385-8. [PMID: 16800963 DOI: 10.3816/clc.2006.n.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) given concurrently with chemotherapy do not improve patient outcomes compared with chemotherapy alone in advanced-stage non-small-cell lung cancer (NSCLC). One potential explanation for this lack of benefit is a negative interaction or antagonism between chemotherapy and EGFR TKIs when delivered concomitantly. Support for this line of reasoning is provided by preclinical data demonstrating that EGFR TKIs induce primarily a cytostatic effect resulting from a G1 cell cycle arrest in cell lines with wild-type EGFR, reducing cell cycle phase-dependent activity of chemotherapy, whereas they induce apoptotic cell death in tumors with EGFR-activating mutations. Because the great majority of NSCLC tumors consist of wild-type EGFR, sequence-specific interactions of EGFR TKI/chemotherapy combinations might negatively influence the efficacy of these regimens in patients with NSCLC. Further evidence is provided by EGFR mutational analysis in patient tumor specimens from the TRIBUTE study. Herein we provide the preclinical and clinical rationale for studies examining the concept of pharmacodynamic separation as a means for overcoming hypothesized antagonism of EGFR TKIs and chemotherapy.
Collapse
Affiliation(s)
- Angela M Davies
- Division of Hematology/Oncology, University of California Davis Cancer Center, Sacramento, CA 95817, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Rossi G, Cavazza A, Marchioni A, Longo L, Migaldi M, Sartori G, Bigiani N, Schirosi L, Casali C, Morandi U, Facciolongo N, Maiorana A, Bavieri M, Fabbri LM, Brambilla E. Role of chemotherapy and the receptor tyrosine kinases KIT, PDGFRalpha, PDGFRbeta, and Met in large-cell neuroendocrine carcinoma of the lung. J Clin Oncol 2006; 23:8774-85. [PMID: 16314638 DOI: 10.1200/jco.2005.02.8233] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Pulmonary large-cell neuroendocrine carcinoma (LCNEC) is a relatively uncommon, high-grade neuroendocrine tumor sharing several features with small-cell lung carcinoma (SCLC) but currently considered as a variant of non-SCLC and accordingly treated with poor results. Little is known about the optimal therapy of LCNEC and the possible therapeutic molecular targets. PATIENTS AND METHODS We reviewed 83 patients with pure pulmonary LCNEC to investigate their clinicopathologic features, therapeutic strategy, and immunohistochemical expression and the mutational status of the receptor tyrosine kinases (RTKs) KIT, PDGFRalpha, PDGFRbeta, and Met. RESULTS LCNEC histology predicted a dismal outcome (overall median survival, 17 months) even in stage I patients (5-year survival rate, 33%). LCNEC strongly expressed RTKs (KIT in 62.7% of patients, PDGFRalpha in 60.2%, PDGFRbeta in 81.9%, and Met in 47%), but no mutations were detected in the exons encoding for the relevant juxtamembrane domains. Tumor stage and size (> or = 3 cm) and Met expression were significantly correlated with survival. At univariate and multivariate analysis, SCLC-based chemotherapy (platinum-etoposide) was the most important variable correlating with survival, both in the adjuvant and metastatic settings (P < .0001). CONCLUSION Pulmonary LCNEC represents an aggressive tumor requiring multimodal treatment even for resectable stage I disease, and LCNEC seems to respond to adjuvant platinum-etoposide-based chemotherapy. Patients who received this therapy had the best survival rate. Despite our failure in finding mutational events in the tested RTKs, the strong expression of KIT, PDGFRalpha, PDGFRbeta, and Met in tumor cells suggests an important role of these RTKs in LCNEC, and these RTKs seem to be attractive therapeutic targets.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carboplatin/administration & dosage
- Carcinoma, Large Cell/drug therapy
- Carcinoma, Large Cell/enzymology
- Carcinoma, Large Cell/mortality
- Carcinoma, Neuroendocrine/drug therapy
- Carcinoma, Neuroendocrine/enzymology
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Small Cell/drug therapy
- Carcinoma, Small Cell/enzymology
- Carcinoma, Small Cell/mortality
- Cisplatin/administration & dosage
- DNA Mutational Analysis
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Female
- Follow-Up Studies
- Humans
- Immunohistochemistry
- Lung Neoplasms/drug therapy
- Lung Neoplasms/enzymology
- Lung Neoplasms/mortality
- Male
- Middle Aged
- Multivariate Analysis
- Neoplasm Staging
- Paclitaxel/administration & dosage
- Polymerase Chain Reaction
- Proto-Oncogene Proteins c-kit/biosynthesis
- Proto-Oncogene Proteins c-kit/genetics
- Proto-Oncogene Proteins c-met/biosynthesis
- Proto-Oncogene Proteins c-met/genetics
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor, Platelet-Derived Growth Factor alpha/biosynthesis
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor beta/biosynthesis
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Survival Analysis
- Survival Rate
- Treatment Outcome
- Gemcitabine
Collapse
Affiliation(s)
- Giulio Rossi
- Integrated Department of Diagnostic and Laboratory Services and Legal Medicine, Section of Pathologic Anatomy, Respiratory Disease Clinic, University of Modena and Reggio Emilia, via del Pozzo, 71-41100, Modena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yu L, Ju DW, Chen W, Li T, Xu Z, Jiang C, Chen S, Tao Q, Ye D, Hu P, Khawli LA, Taylor CR, Epstein AL. 131I-chTNT Radioimmunotherapy of 43 Patients with Advanced Lung Cancer. Cancer Biother Radiopharm 2006; 21:5-14. [PMID: 16480326 DOI: 10.1089/cbr.2006.21.5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
UNLABELLED The treatment of advanced lung cancer remains a major challenge in clinical medicine, justifying an urgent need for new therapeutic approaches. In a rather unique international collaboration, 43 patients with advanced lung cancer were treated using iodine-131-labeled tumor necrosis therapy chimeric antibody (131I-chTNT). METHODS Patients were treated either with intravenous (i.v.) infusion (n = 22), intratumoral injection using a computer tomography (CT)-guided catheter (n = 16), or combination i.v. and intratumoral infusion (n = 5). All patients, regardless of route of administration, received 2 doses of 131I-chTNT on days 1 and 14. RESULTS The results showed that of those patients receiving i.v. injection alone, 2 achieved partial response (PR) (9%), 16 had stable disease (73%), and 4 progressed (18%). Of those patients receiving intratumoral injection only, 1 had a complete response (CR) (6%), 8 achieved PR (50%), 7 had stable disease (44%), and none (0%) progressed. Finally, of those patients receiving both i.v. and intratumoral administration, 1 had a CR (20%), 1 achieved PR (20%), 2 had stable disease (40%), and 1 (20%) showed progression. CONCLUSIONS These promising results demonstrate that sufficient doses of radiolabeled antibody can be safely delivered to tumors to cause significant therapeutic effects in advanced lung cancer.
Collapse
Affiliation(s)
- Like Yu
- Nanjing Pulmonary Hospital, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kim YC, Kim KS. Drugs for Lung Cancer Treatment. Tuberc Respir Dis (Seoul) 2006. [DOI: 10.4046/trd.2006.60.2.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Young-Chul Kim
- Division of Pulmonology, Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Korea
| | - Kyu-Sik Kim
- Division of Pulmonology, Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Korea
| |
Collapse
|
41
|
Abstract
Growth factor signals are propagated from the cell surface to intracellular processes that control critical functions such as growth, differentiation, angiogenesis, and inhibition of apoptosis via sequential kinase signaling. These kinases are receptor kinases, which are transmembrane proteins such as epidermal growth factor receptor or cytoplasmic kinases such as Src kinase. In malignancies, these signaling pathways are often exploited to optimize tumor growth and metastasis. Thus, they represent attractive targets for cancer therapy. This review will summarize current knowledge of the small-molecule multiple-kinase inhibitors in lung cancer therapy. These inhibitors generally hinder the phosphorylation of several protein kinases of membrane receptors, such as vascular endothelial growth factor receptors, platelet-derived growth factor receptors, the human epidermal growth factor receptor family, and cytoplasmic receptors such as c-Kit, Raf kinase, and FLT3. These inhibitors include ZD6474, SU11248, AEE 788, sorafenib, vatalanib, and AG-013736.
Collapse
Affiliation(s)
- Alex A Adjei
- Division of Medical Oncology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
42
|
Abstract
Neuroendocrine pulmonary and thymic tumors constitute a distinct category of tumors collectively disclosing morphologic and biologic neuroendocrine features. They are classified in 4 histopathological types and 3 malignancy grades. The typical carcinoids are of low grade, the atypical carcinoids of intermediate grade and the large cell neuroendocrine carcinoma with the small cell carcinoma are high grade neuroendocrine tumors. Their distinction relies on objective morphologic and phenotypic criteria of strong clinical significance and predictive prognostic value.
Collapse
|
43
|
Robert F, Blumenschein G, Herbst RS, Fossella FV, Tseng J, Saleh MN, Needle M. Phase I/IIa study of cetuximab with gemcitabine plus carboplatin in patients with chemotherapy-naive advanced non-small-cell lung cancer. J Clin Oncol 2005; 23:9089-96. [PMID: 16301597 DOI: 10.1200/jco.2004.00.1438] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE This multicenter, open-label, phase I/IIa study was undertaken to establish the safety/toxicity profile of cetuximab in combination with gemcitabine and carboplatin in patients with chemotherapy-naïve, epidermal growth factor receptor-positive, stage IV non-small-cell lung cancer. Secondary objectives were to gather preliminary evidence of efficacy including tumor response rate, time to progression, and overall survival. PATIENTS AND METHODS Thirty-five patients received a total of 264 3-week cycles of treatment with cetuximab, carboplatin, and gemcitabine. An initial dose of cetuximab 400 mg/m2 intravenously was administered the first week, followed by weekly doses of 250 mg/m2. Carboplatin (area under the curve = 5, day 1) and gemcitabine 1,000 mg/m2 on days 1 and 8 were administered every 3 weeks. Patients were evaluated for tumor response after every two cycles of therapy. RESULTS The most frequently reported adverse events related to cetuximab included an acne-like rash (88.6%), dry skin (34.3%), asthenia and skin disorders (31.4%), mucositis/stomatitis (25.7%), fever/chills (20%), and nausea/vomiting (17.1%). The majority of these toxicities were mild to moderate. One patient withdrew from the study because of a grade 3 allergic reaction. Myelosuppression was the most frequently observed toxicity related to chemotherapy. Responses among 35 assessable patients included 10 partial responses (28.6%). Twenty-one patients had stable disease. The median time to progression was 165 days, and the median overall survival was 310 days. CONCLUSION The combination of cetuximab, carboplatin, and gemcitabine was well tolerated with an acceptable toxicity profile. Most grade 3 adverse events were attributable to chemotherapy. The response rate and median survival are encouraging and warrant additional investigation.
Collapse
Affiliation(s)
- Francisco Robert
- University of Alabama at Birmingham, Comprehensive Cancer Center, Birmingham, AL 35294-3300, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Vansteenkiste J, Canon JL, Riska H, Pirker R, Peterson P, John W, Mali P, Lahn M. Randomized phase II evaluation of aprinocarsen in combination with gemcitabine and cisplatin for patients with advanced/metastatic non-small cell lung cancer. Invest New Drugs 2005; 23:263-9. [PMID: 15868384 DOI: 10.1007/s10637-005-6736-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Aprinocarsen is a specific antisense oligonucleotide inhibitor of protein kinase C-alpha. This study aimed to evaluate the response rate to combination therapy with aprinocarsen, gemcitabine and cisplatin, in chemonaive patients with advanced/metastatic NSCLC. Secondary objectives included comparison of response rate, time to event efficacy parameters, and toxicities on the 2 treatment arms. Patients with stage IV, or stage IIIB disease (N3 and/or pleural/pericardial effusion), were randomized to either control or experimental arm. Patients on both arms received gemcitabine 1250 mg/m2 on days 1 and 8, and cisplatin 80 mg/m2 on day 1 of a 3-week cycle. Additionally, on the experimental arm, aprinocarsen was administered as 2 mg/kg continuous iv infusion on days 1-14, every 21 days. A total of 18 enrolled patients were randomized on the 2 arms. Further enrollment was terminated in March 2003 as a result of a phase III trial suggesting that aprinocarsen did not have an added survival benefit when combined with paclitaxel and carboplatin therapy in patients with NSCLC. Patients received a median of 4 cycles on control arm and 2.5 cycles on experimental arm. The response rate was 16.7% in the experimental arm and 44.4% in the control arm. Most frequent grade 3/4 toxicities were hematologic, with a higher incidence of thrombocytopenia in the experimental arm (87.5% vs. 33.3%). Despite the 14-day continuous infusion schedule, infection rate was not increased in the experimental arm. The present study did not show any advantage, in response rate or secondary endpoints, with aprinocarsen; however, the toxicity was not unduly increased, and aprinocarsen regimen was safely administered.
Collapse
|
45
|
Kanashiro CA, Schally AV, Varga JL, Hammann B, Halmos G, Zarandi M. Antagonists of growth hormone releasing hormone and bombesin inhibit the expression of EGF/HER receptor family in H-69 small cell lung carcinoma. Cancer Lett 2005; 226:123-31. [PMID: 16039952 DOI: 10.1016/j.canlet.2005.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2004] [Revised: 12/08/2004] [Accepted: 01/07/2005] [Indexed: 01/03/2023]
Abstract
Effects of in vivo treatment with antagonists of growth hormone-releasing hormone (GHRH), JV-1-65 and MZ-J-7-110, and bombesin/gastrin-releasing peptide antagonist RC-3940-II, on the EGF receptor (EGFR) family, were investigated in H-69 SCLC. Tumors were analyzed by RT-PCR, immunoblotting and binding assays. Treatment with these analogs reduced the binding capacity of EGFR by 18-64%, and inhibited the mRNA expression for EGFR, HER-2 and -3 by 27-75.4, 17-26.3, and 13.8-46.6%, respectively. The antagonists also decreased the protein levels for EGFR by 21-34%, HER-2 by 36-68% and HER-3 by 43-49%. This is the first demonstration that antiproliferative effects of GHRH antagonists are associated with a downregulation of EGF/HER receptors.
Collapse
Affiliation(s)
- Celia A Kanashiro
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, 1601 Perdido Street, New Orleans, LA 70112, USA
| | | | | | | | | | | |
Collapse
|
46
|
Hespanhol VP. Prognóstico do cancro do pulmão: Novos factores biológicos e moleculares. REVISTA PORTUGUESA DE PNEUMOLOGIA 2005. [DOI: 10.1016/s0873-2159(15)30548-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
47
|
Péqueux C, Breton C, Hagelstein MT, Geenen V, Legros JJ. Oxytocin receptor pattern of expression in primary lung cancer and in normal human lung. Lung Cancer 2005; 50:177-88. [PMID: 16043261 DOI: 10.1016/j.lungcan.2005.05.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Revised: 05/23/2005] [Accepted: 05/25/2005] [Indexed: 12/29/2022]
Abstract
In order to assess if oxytocin- and vasopressin-induced mitogenic effects detected on small-cell lung carcinoma (SCLC) cell lines could be transposed on primary SCLC, the aim of the present work was to identify mediators of these mitogenic actions on primary tumours samples. This was addressed on normal human lung tissue, on SCLC and on non-SCLC (NSCLC). Herein, we observe, in normal human lung, that OTR is colocalized with vascular endothelial cells of the lung and is not expressed by lung cells of epithelial nature. We detected mRNA amplification of V1aR, V2R and of a V2R variant. We observed that 86% of SCLC biopsies analyzed expressed at least the OTR and that 71% expressed the OTR, the V1aR and the V2R altogether. Comparatively, 50% of NSCLC biopsies tested expressed at least the OTR and 32% expressed the OTR, the V1aR and the V2R altogether. The occurrence of the V1bR/V3R is of 28 and 18% for SCLC and NSCLC, respectively. Nevertheless, for the SCLC biopsies analyzed in this study, V1bR/V3R expression correlates, in all cases, with the expression of all the other neurohypophysial peptide receptors. Our results suggest that neurohypophysial peptide antagonists may offer promise as a potential new therapeutic modality for the treatment of lung cancer expressing at least one of the neurhypophysial peptide receptor subtypes.
Collapse
Affiliation(s)
- Christel Péqueux
- University of Liege, Institute of Pathology CHU-B23, +4, Center of Immunology & Laboratory of Neuroendocrinology, B-4000 Liege 1-Sart Tilman, Belgium.
| | | | | | | | | |
Collapse
|
48
|
Abstract
This article reviews novel therapies that are under development for non-small cell lung cancer and small cell lung cancer.
Collapse
Affiliation(s)
- Olwen Hahn
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Cancer Research Center, Pritzker School of Medicine, 5841 South Maryland Avenue, MC 2115, Chicago, IL 60637, USA
| | | |
Collapse
|
49
|
Choong NW, Ma PC, Salgia R. Therapeutic targeting of receptor tyrosine kinases in lung cancer. Expert Opin Ther Targets 2005; 9:533-59. [PMID: 15948672 DOI: 10.1517/14728222.9.3.533] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lung cancer is a difficult illness with a poor overall survival. Even though combination strategies with chemotherapy, radiation therapy and surgery have all been utilised, the overall outcome for this disease continues to be relatively disappointing. In order to make a difference in the treatment of lung cancer, novel therapeutics will have to be developed. Through basic biological studies, a number of receptor tyrosine kinases have been implicated in the pathogenesis and progression of lung cancer. In this review, the authors summarise the mechanisms of several major receptor tyrosine kinases in lung cancer, especially epidermal growth factor receptor, Her2/neu, MET, vascular endothelial growth factor and KIT. The biology associated with these receptors is described, and the various novel therapeutic inhibitory strategies that are ongoing in preclinical and clinical studies for lung cancer are detailed. Through understanding of receptor tyrosine kinases and the utilisation of specific inhibitors, it is hopeful that a dramatic impact will be made on the biology and therapy for lung cancer.
Collapse
Affiliation(s)
- Nicholas W Choong
- University of Chicago Medical Center, Pritzker School of Medicine, MC 2115, 5841, S. Maryland Avenue, Chicago, IL 60615, USA
| | | | | |
Collapse
|
50
|
Gasparini G, Meo S, Comella G, Stani SC, Mariani L, Gamucci T, Avallone A, Lo Vullo S, Mansueto G, Bonginelli P, Gattuso D, Gion M. The Combination of the Selective Cyclooxygenase-2 Inhibitor Celecoxib with Weekly Paclitaxel Is a Safe and Active Second-Line Therapy for Non-Small Cell Lung Cancer. Cancer J 2005; 11:209-16. [PMID: 16053664 DOI: 10.1097/00130404-200505000-00007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The selection of effective schedules of treatment for metastatic non-small cell lung cancer still remains a challenge for the oncologist. The present multicentric phase II study was designed in order to investigate the activity and safety of the combination of weekly paclitaxel and celecoxib as second-line treatment for non-small cell lung cancer. As a secondary endpoint, the possible correlation of biomarkers with objective response was investigated in a subset of patients. PATIENTS AND METHODS Patients with platinum-refractory non-small cell lung cancer and Eastern Cooperative Oncology Group performance status 0-2 entered the present phase II study. Paclitaxel was administered at the dose of 80 mg/m(2) i.v. weekly for 6 weeks, followed by a 2-week rest, and celecoxib, 400 mg p.o. b.i.d. administered continuously. A cycle consisted of 8 weeks of treatment. Determination of circulating vascular endothelial growth factor and interleukin 6 was performed at baseline and every two cycles. RESULTS Fifty-eight patients were enrolled: median age, 60 years (range, 30-77 years); male/female ratio = 44/14; performance status, 0, 31 patients; 1, 25 patients; and 2, two patients. Predominant histotype was adenocarcinoma (34 cases), and most patients had at least two sites of disease. According to the intent-to-treat analysis, 14/58 objective responses (24.1%) and 24/58 (41.3%) stabilizations of disease were observed, with a median duration of 4 months (range, 2-22+ months) and 5 months (range, 1-13 months), respectively. Median time to progression and median overall survival were 5 and 11 months, respectively. One-year survival was 42.5%. The main toxicity was neuropathy (4% of grade 3). Preliminary results suggest that decrease in serum vascular endothelial growth factor level is significantly associated with clinical response. DISCUSSION Combination of celecoxib and weekly paclitaxel is safe and active new regimen in pretreated non-small cell lung cancer. Toxicity appears not to be worsened by the addition of celecoxib. According to preliminary results, serum vascular endothelial growth factor level seems to be predictive of response, suggesting that it should be further investigated as a surrogate marker of response.
Collapse
|