1
|
Muñoz-Moreno L, Gómez-Calcerrada MI, Arenas MI, Carmena MJ, Prieto JC, Schally AV, Bajo AM. Antagonist of Growth Hormone-Releasing Hormone Receptor MIA-690 Suppresses the Growth of Androgen-Independent Prostate Cancers. Int J Mol Sci 2024; 25:11200. [PMID: 39456984 PMCID: PMC11508372 DOI: 10.3390/ijms252011200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The development of resistance remains the primary challenge in treating castration-resistant prostate cancer (CRPC). GHRH receptors (GHRH-R), which are coupled to G-proteins (GPCRs), can mediate EGFR transactivation, offering an alternative pathway for tumour survival. This study aimed to evaluate the effects of the GHRH-R antagonist MIA-690, in combination with the EGFR inhibitor Gefitinib, on cell viability, adhesion, gelatinolytic activity, and the cell cycle in advanced prostate cancer PC-3 cells. The findings demonstrate a synergistic effect between MIA-690 and Gefitinib, leading to the inhibition of cell viability, adhesion, and metalloprotease activity. Cell cycle analysis suggests that both compounds induce cell cycle arrest, both individually and in combination. Furthermore, similar effects of the GHRH-R antagonist MIA-690 combined with Gefitinib were observed in PC-3 tumours developed by subcutaneous injection in athymic nude mice 36 days post-inoculation. These results indicate that combined therapy with a GHRH-R antagonist and an EGFR inhibitor exerts a stronger antitumor effect compared to monotherapy by preventing transactivation between EGFR and GHRH-R in CRPC.
Collapse
Affiliation(s)
- Laura Muñoz-Moreno
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - M. Isabel Gómez-Calcerrada
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - M. Isabel Arenas
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Biología Celular, Departamento de Biomedicina y Biotecnología, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - M. José Carmena
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - Juan C. Prieto
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - Andrew V. Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33125, USA
- Department of Pathology and Medicine, Division of Hematology/Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ana M. Bajo
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| |
Collapse
|
2
|
Modica TME, Dituri F, Mancarella S, Pisano C, Fabregat I, Giannelli G. Calcium Regulates HCC Proliferation as well as EGFR Recycling/Degradation and Could Be a New Therapeutic Target in HCC. Cancers (Basel) 2019; 11:cancers11101588. [PMID: 31635301 PMCID: PMC6826902 DOI: 10.3390/cancers11101588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/13/2019] [Indexed: 01/08/2023] Open
Abstract
Calcium is the most abundant element in the human body. Its role is essential in physiological and biochemical processes such as signal transduction from outside to inside the cell between the cells of an organ, as well as the release of neurotransmitters from neurons, muscle contraction, fertilization, bone building, and blood clotting. As a result, intra- and extracellular calcium levels are tightly regulated by the body. The liver is the most specialized organ of the body, as its functions, carried out by hepatocytes, are strongly governed by calcium ions. In this work, we analyze the role of calcium in human hepatoma (HCC) cell lines harboring a wild type form of the Epidermal Growth Factor Receptor (EGFR), particularly its role in proliferation and in EGFR downmodulation. Our results highlight that calcium is involved in the proliferative capability of HCC cells, as its subtraction is responsible for EGFR degradation by proteasome machinery and, as a consequence, for EGFR intracellular signaling downregulation. However, calcium-regulated EGFR signaling is cell line-dependent. In cells responding weakly to the epidermal growth factor (EGF), calcium seems to have an opposite effect on EGFR internalization/degradation mechanisms. These results suggest that besides EGFR, calcium could be a new therapeutic target in HCC.
Collapse
Affiliation(s)
- Teresa Maria Elisa Modica
- Department of Biomedical Science and Human Oncology, Università degli Studi di Bari Aldo Moro, 70121 Bari, Italy.
- Biogem S.C.A.R.L., 83031 Ariano Irpino (AV), Italy.
| | | | | | | | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL) L'Hospitalet, 08907 Barcelona, Spain.
- Faculty of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain.
- Oncology Program, CIBEREHD, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | | |
Collapse
|
3
|
Shen W, Xi H, Li C, Bian S, Cheng H, Cui J, Wang N, Wei B, Huang X, Chen L. Endothelin-A receptor in gastric cancer and enhanced antitumor activity of trastuzumab in combination with the endothelin-A receptor antagonist ZD4054. Ann N Y Acad Sci 2019; 1448:30-41. [PMID: 30937921 DOI: 10.1111/nyas.14053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/30/2018] [Accepted: 02/21/2019] [Indexed: 12/25/2022]
Abstract
Endothelin-A receptor (ETAR) is overexpressed in cancers and can function through transactivation of the epidermal growth factor receptor. We explored ETAR in gastric cancer and investigated the antitumor effect of trastuzumab in combination with the ETAR antagonist ZD4054. The expression of ETAR was significantly correlated with the expression of vascular endothelial growth factor. Univariate and multivariate analyses further showed that ETAR expression correlated with reduced survival in gastric cancer patients. In vitro, ZD4054 increased the antiproliferative effect of trastuzumab in gastric cancer cell lines. Moreover, the addition of ZD4054 to trastuzumab significantly increased apoptosis in gastric cancer cell lines. In vivo, tumor growth was considerably inhibited by treatment with ZD4054 and trastuzumab, and the tumor volume in the trastuzumab and ZD4054 combination group was smaller than in the other groups. The detection of ETAR could help predict the prognosis of gastric cancer patients. Additionally, this study provides support for the therapeutic use of the combination of ZD4054 and trastuzumab as an anticancer treatment, especially for gastric cancer.
Collapse
Affiliation(s)
- Weisong Shen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongqing Xi
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chenyang Li
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shibo Bian
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haidong Cheng
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jianxin Cui
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ning Wang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Bo Wei
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaohui Huang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lin Chen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
4
|
Kim JS, Kim MW, Kang SJ, Jeong HY, Park SI, Lee YK, Kim HS, Kim KS, Park YS. Tumor-specific delivery of therapeutic siRNAs by anti-EGFR immunonanoparticles. Int J Nanomedicine 2018; 13:4817-4830. [PMID: 30214190 PMCID: PMC6118344 DOI: 10.2147/ijn.s161932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Efficient target-specific siRNA delivery has always been a primary concern in the field of siRNA clinical application. Purpose In this study, four different types of anti-epidermal growth factor receptor (EGFR) antibody-conjugated immunonanoparticles were prepared and tested for cancer cell-targeted therapeutic siRNA delivery. Materials and methods The prepared nanoparticles encapsulating siRNAs were character-ized by gel retardation and particle analysis using a Zetasizer. In vitro transfection and reduction of target genes, vimentin and JAK3, were determined using quantitative reverse transcription polymerase chain reaction. In vivo tumor targeting and antitumoral efficacies of the nanoparticles were evaluated in mice carrying tumors. Results Among these immunonanoparticles, anti-EGFR immunolipoplexes and immunoviroplexes exhibited remarkable cell binding and siRNA delivery to EGFR-expressing tumor cells compared to immunoliposomes and immunovirosomes. Especially, the anti-EGFR immunoviroplexes exhibited the most efficient siRNA transfection to target tumor cells. Therefore, antitumoral vimentin and Janus kinase-3 siRNAs were loaded in the anti-EGFR immunolipoplexes and immunoviroplexes, which were tested in mice carrying SK-OV-3 tumor xenografts. In fact, the therapeutic siRNAs were efficiently delivered to the tumor tissues by both delivery vehicles, resulting in significant inhibition of tumor growth. Moreover, administration of doxorubicin in combination with anti-EGFR immunoviroplexes resulted in remarkable and synergistic tumor growth inhibition. Conclusion This study provides experimental proof that cancer cell-targeted immunoviroplexes are an efficient siRNA delivery system for cancer therapy. Moreover, this study also suggests that a combination of conventional chemotherapy and tumor-directed anticancer siRNA therapy would be a better modality for cancer treatment.
Collapse
Affiliation(s)
- Jung Seok Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| | - Min Woo Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| | - Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| | - Hwa Yeon Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| | - Sang Il Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| | - Yeon Kyung Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| | - Hong Sung Kim
- Department of Biomedical Laboratory Science, Korea Nazarene University, Cheonan, Republic of Korea
| | - Keun Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Republic of Korea
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea,
| |
Collapse
|
5
|
Kaneto N, Yokoyama S, Hayakawa Y, Kato S, Sakurai H, Saiki I. RAC1 inhibition as a therapeutic target for gefitinib-resistant non-small-cell lung cancer. Cancer Sci 2014; 105:788-94. [PMID: 24750242 PMCID: PMC4317907 DOI: 10.1111/cas.12425] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/09/2014] [Accepted: 04/17/2014] [Indexed: 02/06/2023] Open
Abstract
Although epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (EGFR-TKI), including gefitinib, provide a significant clinical benefit in non-small-cell lung cancer (NSCLC) patients, the acquisition of drug resistance has been known to limit the efficacy of EGFR-TKI therapy. In this study, we demonstrated the involvement of EGF-EGFR signaling in NSCLC cell migration and the requirement of RAC1 in EGFR-mediated progression of NSCLC. We showed the significant role of RAC1 pathway in the cell migration or lamellipodia formation by using gene silencing of RAC1 or induction of constitutive active RAC1 in EGFR-mutant NSCLC cells. Importantly, the RAC1 inhibition suppressed EGFR-mutant NSCLC cell migration and growth in vitro, and growth in vivo even in the gefitinib-resistant cells. In addition, these suppressions by RAC1 inhibition were mediated through MEK or PI3K independent mechanisms. Collectively, these results open up a new opportunity to control the cancer progression by targeting the RAC1 pathway to overcome the resistance to EGFR-TKI in NSCLC patients.
Collapse
Affiliation(s)
- Naoki Kaneto
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
6
|
Farouk N, Ramadan HE, El-Amir MA. Labeling of EGFR inhibitor to prepare a potential SPECT agent for molecular imaging of breast cancer via iodine-bromine exchange. RADIOCHEMISTRY 2012. [DOI: 10.1134/s1066362212050141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
7
|
Volpi G, Facchinetti F, Moretto N, Civelli M, Patacchini R. Cigarette smoke and α,β-unsaturated aldehydes elicit VEGF release through the p38 MAPK pathway in human airway smooth muscle cells and lung fibroblasts. Br J Pharmacol 2011; 163:649-61. [PMID: 21306579 PMCID: PMC3101625 DOI: 10.1111/j.1476-5381.2011.01253.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 11/09/2010] [Accepted: 01/04/2011] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Vascular endothelial growth factor (VEGF) is an angiogenic factor known to be elevated in the sputum of asymptomatic smokers as well as smokers with bronchitis type of chronic obstructive pulmonary disease. The aim of this study was to investigate whether acute exposure to cigarette smoke extract altered VEGF production in lung parenchymal cells. EXPERIMENTAL APPROACH We exposed human airway smooth muscle cells (ASMC), normal human lung fibroblasts (NHLF) and small airways epithelial cells (SAEC) to aqueous cigarette smoke extract (CSE) in order to investigate the effect of cigarette smoke on VEGF expression and release. KEY RESULTS Vascular endothelial growth factor release was elevated by sub-toxic concentrations of CSE in both ASMC and NHLF, but not in SAEC. CSE-evoked VEGF release was mimicked by its component acrolein at concentrations (10-100 µM) found in CSE, and prevented by the antioxidant and α,β-unsaturated aldehyde scavenger, N-acetylcysteine (NAC). Both CSE and acrolein (30 µM) induced VEGF mRNA expression in ASMC cultures, suggesting an effect at transcriptional level. Crotonaldehyde and 4-hydroxy-2-nonenal, an endogenous α,β-unsaturated aldehyde, stimulated VEGF release, as did H(2)O(2). CSE-evoked VEGF release was accompanied by rapid and lasting phosphorylation of p38 MAPK (mitogen-activated protein kinase), which was abolished by NAC and mimicked by acrolein. Both CSE- and acrolein-evoked VEGF release were blocked by selective inhibition of p38 MAPK signalling. CONCLUSIONS AND IMPLICATIONS α,β-Unsaturated aldehydes and possibly reactive oxygen species contained in cigarette smoke stimulate VEGF expression and release from pulmonary cells through p38 MAPK signalling.
Collapse
Affiliation(s)
- Giorgia Volpi
- Department of Pharmacology, Chiesi Farmaceutici S.p.A., Parma, Italy
| | | | | | | | | |
Collapse
|
8
|
Jang SH, Choi SJ, Oh JH, Chae SW, Nam K, Park JS, Lee HJ. Nonviral gene delivery to human ovarian cancer cells using arginine-grafted PAMAM dendrimer. Drug Dev Ind Pharm 2010; 37:41-6. [PMID: 20950058 DOI: 10.3109/03639045.2010.489563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND A specific and effective strategy is in demand to treat ovarian cancer successfully. Epidermal growth factor receptor (EGFR) is highly expressed in ovarian cancer, and thus EGFR antisense gene therapy can be a potential therapeutic strategy. METHOD L-Arginine-grafted-polyamidoamine dendrimer (PAMAM-Arg) has been reported to be a novel nonviral gene delivery carrier. Therefore, the ability of PAMAM-Arg in transferring a luciferase gene to ovarian carcinoma SK-OV3 cells has been examined, and the cytotoxicity of the cationic polymer has been investigated. In addition, the suppression of cell proliferation has been evaluated by transferring an EGFR antisense gene to SK-OV3 cells using PAMAM-Arg. Polyethyleneimine (PEI) 25K was used as a positive control. RESULTS As a result, in vitro gene transfection efficiency of PAMAM-Arg was enhanced with increasing transfection time and N/P ratios. PAMAM-Arg transferred the luciferase gene into cells more efficiently than PEI. In addition, PAMAM-Arg was minimally toxic to the cells whereas PEI 25K was highly toxic. The polyplexes formed by the EGFR antisense gene and PAMAM-Arg significantly reduced thymidine incorporation into the cells suggesting the suppression of cancer cell proliferation. CONCLUSION These results suggest that a PAMAM-Arg/EGFR antisense gene complex can be used as a safe and efficient therapeutic agent for cancer gene therapy.
Collapse
Affiliation(s)
- Soo Hyun Jang
- Division of Life and Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
9
|
Zhou W, Hur W, McDermott U, Dutt A, Xian W, Picarro SB, Zhang J, Sharma SV, Brugge J, Meyerson M, Settleman J, Gray NS. A structure-guided approach to creating covalent FGFR inhibitors. CHEMISTRY & BIOLOGY 2010; 17:285-95. [PMID: 20338520 PMCID: PMC2920453 DOI: 10.1016/j.chembiol.2010.02.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 01/14/2010] [Accepted: 01/29/2010] [Indexed: 02/08/2023]
Abstract
The fibroblast growth factor receptor tyrosine kinases (FGFR1, 2, 3, and 4) represent promising therapeutic targets in a number of cancers. We have developed the first potent and selective irreversible inhibitor of FGFR1, 2, 3, and 4, which we named FIIN-1 that forms a covalent bond with cysteine 486 located in the P loop of the FGFR1 ATP binding site. We demonstrated that the inhibitor potently inhibits Tel-FGFR1-transformed Ba/F3 cells (EC(50) = 14 nM) as well as numerous FGFR-dependent cancer cell lines. A biotin-derivatized version of the inhibitor, FIIN-1-biotin, was shown to covalently label FGFR1 at Cys486. FIIN-1 is a useful probe of FGFR-dependent cellular phenomena and may provide a starting point of the development of therapeutically relevant irreversible inhibitors of wild-type and drug-resistant forms of FGFR kinases.
Collapse
Affiliation(s)
- Wenjun Zhou
- Department of Cancer Biology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wooyoung Hur
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ultan McDermott
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Amit Dutt
- The Broad Institute, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wa Xian
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Scott B. Picarro
- Department of Cancer Biology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jianming Zhang
- Department of Cancer Biology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sreenath V. Sharma
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Joan Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Matthew Meyerson
- The Broad Institute, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jeffrey Settleman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Correspondence: Nathanael S. Gray ()
| |
Collapse
|
10
|
Enediyne lidamycin enhances the effect of epidermal growth factor receptor tyrosine kinase inhibitor, gefitinib, in epidermoid carcinoma A431 cells and lung carcinoma H460 cells. Anticancer Drugs 2009; 20:41-9. [PMID: 19342999 DOI: 10.1097/cad.0b013e328318292c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gefitinib, a low-molecular-weight epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, is effective in a wide variety of tumor types. Preclinical studies have shown potentiated antitumor efficacies of this agent in combination with chemotherapy or radiotherapy. The antitumor antibiotic lidamycin (LDM) showed extremely potent cytotoxicity in vitro and marked therapeutic effect in vivo. In this report, the cytotoxic and biochemical activity of LDM and gefitinib on human epidermoid carcinoma A431 cells and human large cell lung cancer H460 cells as a single agent or in combination has been evaluated. In the MTT assay, LDM showed much more potent cytotoxicity than gefitinib to both cell lines. A431 cells with a highly EGFR-expressing level were more sensitive to gefitinib than H460 cells, which expressed EGFR at an intermediate level. LDM plus gefitinib showed potentiation of antiproliferative activity and apoptosis induction, which were associated with downregulation of EGFR signaling pathway and nuclear factor-kappa B expression, and the increase of cleaved poly (adenosine diphosphate-ribose) polymerase in the two cell lines, although to a lesser degree in H460 cells. Combined treatment induced G1 phase arrest similar to that of gefitinib alone in A431 cells and intensified G2/M phase accumulation in H460 cells. The above results indicate that LDM potentiates the effects of gefitinib in both gefitinib sensitive and less sensitive cells in association with enhanced inhibition of EGFR-dependent signaling.
Collapse
|
11
|
Mauriz JL, González-Gallego J. Antiangiogenic drugs: current knowledge and new approaches to cancer therapy. J Pharm Sci 2009; 97:4129-54. [PMID: 18200520 DOI: 10.1002/jps.21286] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Angiogenesis--process of new blood-vessel growth from existing vasculature--is an integral part of both normal developmental processes and numerous pathologies such as cancer, ischemic diseases and chronic inflammation. Angiogenesis plays a crucial role facilitating tumour growth and the metastatic process, and it is the result of a dynamic balance between proangiogenic and antiangiogenic factors. The potential to block tumour growth and metastases by angiogenesis inhibition represents an intriguing approach to the cancer treatment. Angiogenesis continues to be a topic of major scientific interest; and there are currently more antiangiogenic drugs in cancer clinical trials than those that fit into any other mechanistic category. Based on preclinical studies, researchers believe that targeting the blood vessels which support tumour growth could help treatment of a broad range of cancers. Angiogenic factors or their receptors, endothelial cell proliferation, matrix metalloproteinases or endothelial cell adhesion, are the main targets of an increasing number of clinical trials approved to test the tolerance and therapeutic efficacy of antiangiogenic agents. Unfortunately, contrary to initial expectations, it has been described that antiangiogenic treatment can cause different toxicities in cancer patients. The purpose of this article is to provide an overview of current attempts to inhibit tumour angiogenesis for cancer therapy.
Collapse
Affiliation(s)
- Jose L Mauriz
- Ciberehd and Institute of Biomedicine, University of León, Campus of Vegazana, s/n, 24071 León, Spain
| | | |
Collapse
|
12
|
Rauh-Adelmann C, Moskow JM, Graham JR, Yen LG, Boucher JI, Murphy CE, Nadler TK, Gordon NF, Radding JA. Quantitative measurement of epidermal growth factor receptor–mitogen-activated protein kinase signal transduction using a nine-plex, peptide-based immunoassay. Anal Biochem 2008; 375:255-64. [DOI: 10.1016/j.ab.2007.12.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 12/28/2007] [Accepted: 12/30/2007] [Indexed: 01/07/2023]
|
13
|
|
14
|
Hare KJ, Hartmann B, Kissow H, Holst JJ, Poulsen SS. The intestinotrophic peptide, glp-2, counteracts intestinal atrophy in mice induced by the epidermal growth factor receptor inhibitor, gefitinib. Clin Cancer Res 2007; 13:5170-5. [PMID: 17785573 DOI: 10.1158/1078-0432.ccr-07-0574] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors have been introduced as antitumor agents in the treatment of cancers overexpressing the receptor. The treatment has gastrointestinal side effects which may decrease patient compliance and limit the efficacy. Glucagon-like peptide-2 (GLP-2) is an intestinal hormone with potent intestinotrophic properties and therapeutic potential in disorders with compromised intestinal capacity. The growth stimulation is highly specific to the gastrointestinal tract, and no effects are observed elsewhere. The aim of this study was to examine whether the inhibition of the EGFR induces intestinal atrophy and if this can be counteracted by treatment with GLP-2. EXPERIMENTAL DESIGN Mice were treated for 10 days with either gefitinib orally, GLP-2 as injections, or a combination of both. After sacrifice, the weight and length of the segments of the gastrointestinal tract were determined, and histologic sections were analyzed by morphometric methods. RESULTS A significant atrophy of the small-intestinal wall was observed after treatment with gefitinib because both intestinal weight and morphometrically estimated villus height and cross-sectional area were decreased. The same parameters were increased by GLP-2 treatment alone, and when GLP-2 was combined with the gefitinib treatment, the parameters remained unchanged. CONCLUSIONS Treatment with an EGFR tyrosine kinase inhibitor in mice results in small-intestinal growth inhibition that can be completely prevented by simultaneous treatment with GLP-2. This suggests that the gastrointestinal side effects elicited by treatment with EGFR tyrosine kinase inhibitors can be circumvented by GLP-2 treatment.
Collapse
Affiliation(s)
- Kristine Juul Hare
- Department of Anatomy, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
15
|
Park HS, Lee HJ, Im JG, Goo JM, Lee CH, Park CM, Chun EJ. Gefitinib-induced pneumonitis in non-small cell lung cancer: radiological and clinical findings in five patients. Clin Imaging 2007; 31:306-12. [PMID: 17825737 DOI: 10.1016/j.clinimag.2007.04.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2007] [Accepted: 04/13/2007] [Indexed: 11/19/2022]
Abstract
PURPOSE The objective of this study was to describe the radiological and clinical features of gefitinib-induced pneumonitis in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS Five patients who suffered dyspnea after gefitinib treatment were selected. Chest radiographs and computed tomography (CT) findings, along with clinical course, were evaluated. RESULTS Patients complained of subacute dyspnea and hypoxia. Three patients improved after discontinuation of gefitinib, while remaining two showed no response. Unilateral or bilateral ground glass opacity was observed on chest radiographs and CT. CONCLUSION Radiological findings of gefitinib-induced pneumonitis were nonspecific, but radiologists should be aware of this adverse reaction, which can appear during the treatment in NSCLC patients.
Collapse
Affiliation(s)
- Hee Sun Park
- Department of Radiology, Seoul National University College of Medicine, Seoul 110-744, South Korea
| | | | | | | | | | | | | |
Collapse
|
16
|
Rosanò L, Di Castro V, Spinella F, Tortora G, Nicotra MR, Natali PG, Bagnato A. Combined targeting of endothelin A receptor and epidermal growth factor receptor in ovarian cancer shows enhanced antitumor activity. Cancer Res 2007; 67:6351-9. [PMID: 17616694 DOI: 10.1158/0008-5472.can-07-0883] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ovarian carcinomas overexpress endothelin A receptors (ET(A)R) and epidermal growth factor (EGF) receptor (EGFR). In these cells, endothelin-1 (ET-1) triggers mitogenic and invasive signaling pathways that are in part mediated by EGFR transactivation. Combined targeting of ET(A)R, by the specific ET(A)R antagonist ZD4054, and of EGFR by the EGFR inhibitor gefitinib (IRESSA), may offer improvements in ovarian carcinoma treatment. In HEY and OVCA 433 ovarian carcinoma cells, ET-1 or EGF induced rapid activation of EGFR, p42/44 mitogen-activated protein kinase (MAPK), and AKT. ZD4054 was able to reduce the ET-1-induced EGFR transactivation. Gefitinib significantly inhibited EGF- and ET-1-induced EGFR phosphorylation, but incompletely reduced the ET-1-induced activation of downstream targets. ZD4054 plus gefitinib resulted in a greater inhibition of EGFR, MAPK, and AKT phosphorylation, indicating the critical role of these interconnected signaling proteins. ZD4054 effectively inhibited cell proliferation, invasiveness, and vascular endothelial growth factor (VEGF) secretion. Concomitantly, ZD4054 enhanced apoptosis and E-cadherin promoter activity and expression. In both cell lines, the drug combination resulted in a significant decrease in cell proliferation (65%), invasion (52%), and VEGF production (50%), accompanied by a 2-fold increase in apoptosis. The coadministration of ZD4054 enhanced the efficacy of gefitinib leading to partial (82%) or complete tumor regression on HEY ovarian carcinoma xenografts. Antitumor effects were paralleled by biochemical and immunohistologic evidence of decreased vascularization, Ki-67, matrix metalloproteinase-2 (MMP-2), VEGF, MAPK and EGFR, and enhanced E-cadherin expression. The cross-signaling between the EGFR/ET(A)R pathways provides a rationale to combine EGFR inhibitors with ET(A)R antagonists, identifying new effective therapeutic opportunities for ovarian cancer.
Collapse
Affiliation(s)
- Laura Rosanò
- Molecular Pathology Laboratory, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
ARMOUR A. Gefitinib in advanced non-small cell lung cancer: Clinical experience in patients of Asian origin. Asia Pac J Clin Oncol 2007. [DOI: 10.1111/j.1743-7563.2007.00090.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
18
|
Fernandes C, Oliveira C, Gano L, Bourkoula A, Pirmettis I, Santos I. Radioiodination of new EGFR inhibitors as potential SPECT agents for molecular imaging of breast cancer. Bioorg Med Chem 2007; 15:3974-80. [PMID: 17449254 DOI: 10.1016/j.bmc.2007.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Revised: 03/29/2007] [Accepted: 04/05/2007] [Indexed: 11/16/2022]
Abstract
In our search for the development of novel SPECT radioligands for EGFR positive tumours, new potentially irreversible tyrosine kinase (TK) inhibitors are being explored. The radioiodination of N-{4-[(3-chloro-4-fluorophenyl) amino]quinazoline-6-yl}-3-bromopropionamide, a novel EGFR-TK inhibitor synthesised in our laboratory, was accomplished via halogen exchange. Purification by RP-HPLC gave [125I]-N-{4-[(3-chloro-4-fluorophenyl)amino]quinazoline-6-yl}-3-iodopropionamide with a radiochemical purity higher than 95% and a high specific activity. In vitro studies indicate that both iodinated quinazoline and its bromo precursor inhibit A431 cell growth and also possess higher potency than the parent quinazoline to inhibit the EGFR autophosphorylation. In vivo stability studies suggest metabolization of the radioiodinated quinazoline indicating a short biological half-life. The in vitro results point out that these quinazoline derivatives could be promising candidates for SPECT imaging of EGFR positive tumours provided that they are selectively modified in order to achieve better in vivo radiochemical stability.
Collapse
Affiliation(s)
- Célia Fernandes
- Departamento de Química, ITN, Estrada Nacional 10, 2686-953, Sacavém Codex, Portugal
| | | | | | | | | | | |
Collapse
|
19
|
Yan Y, Lu Y, Wang M, Vikis H, Yao R, Wang Y, Lubet RA, You M. Effect of an epidermal growth factor receptor inhibitor in mouse models of lung cancer. Mol Cancer Res 2007; 4:971-81. [PMID: 17189387 DOI: 10.1158/1541-7786.mcr-06-0086] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gefitinib (Iressa, ZD1839) is a potent high-affinity competitive tyrosine kinase inhibitor aimed primarily at epidermal growth factor receptor (EGFR). Inhibitors in this class have recently been approved for clinical use in the treatment of advanced non-small cell lung cancer as monotherapy following failure of chemotherapy. We examined the efficacy of gefitinib on lung tumorigenesis in mouse models using both postinitiation and progression protocols. Gefitinib was given at a dose of 200 mg/kg body weight (i.g.) beginning either 2 or 12 weeks following carcinogen initiation. In the postinitiation protocol, gefitinib significantly inhibited both tumor multiplicity (approximately 70%) and tumor load (approximately 90%) in A/J or p53-mutant mice (P < 0.0001). Interestingly, gefitinib was also highly effective against lung carcinogenesis in the progression protocol when individual animals already have multiple preinvasive lesions in the lung. Gefitinib exhibited approximately 60% inhibition of tumor multiplicity and approximately 80% inhibition of tumor load when compared with control mice (both P < 0.0001). These data show that gefitinib is a potent chemopreventive agent in both wild-type and p53-mutant mice and that a delayed administration was still highly effective. Analyses of mutations in the EGFR and K-ras genes in lung tumors from either control or treatment groups showed no mutations in EGFR and consistent mutation in K-ras. Using an oligonucleotide array on control and gefitinib-treated lesions showed that gefitinib treatment failed to alter the activity or the expression level of EGFR. In contrast, gefitinib treatment significantly altered the expression of a series of genes involved in cell cycle, cell proliferation, cell transformation, angiogenesis, DNA synthesis, cell migration, immune responses, and apoptosis. Thus, gefitinib showed highly promising chemopreventive and chemotherapeutic activity in this mouse model of lung carcinogenesis.
Collapse
Affiliation(s)
- Ying Yan
- Department of Surgery, The Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Ueda SI, Basaki Y, Yoshie M, Ogawa K, Sakisaka S, Kuwano M, Ono M. PTEN/Akt signaling through epidermal growth factor receptor is prerequisite for angiogenesis by hepatocellular carcinoma cells that is susceptible to inhibition by gefitinib. Cancer Res 2006; 66:5346-53. [PMID: 16707461 DOI: 10.1158/0008-5472.can-05-3684] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common tumor-related causes of death worldwide for which there is still no satisfactory treatment. We previously reported the antiangiogenic effect of gefitinib, a selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that has been used successfully to treat lung cancer. In this study, we investigated the effects of gefitinib on tumor-induced angiogenesis by using HCC cell lines (HCC3, CBO12C3, and AD3) in vitro as well as in vivo. Oral administration of gefitinib inhibited angiogenesis induced by HCC3 and CBO12C3, but not by AD3 in the mouse dorsal air sac model. Production of both vascular endothelial growth factor (VEGF) and chemokine C-X-C motif ligand 1 (CXCL1) by EGF-stimulated HCC was more markedly inhibited by gefitinib in HCC3 and CBO12C3 cells than in AD3 cells. EGF stimulated the phosphorylation of EGFR, Akt, and extracellular signal-regulated kinase 1/2 (ERK1/2) in HCC3 and CBO12C3 cells, whereas EGF stimulated phosphorylation of EGFR and ERK1/2, but not Akt in AD3 cells. In fact, Akt was constitutively activated in the absence of EGF in AD3 cells. Gefitinib inhibited Akt phosphorylation in all three cell lines, but it was about five times less effective in AD3 cells. The concentration of PTEN in AD3 cells was about a half that in HCC3 and CBO12C3 cells. Transfection of HCC3 cells with PTEN small interfering RNA reduced their sensitivity to gefitinib in terms of its inhibitory effect on both Akt phosphorylation and the production of VEGF and CXCL1. In conclusion, effect of gefitinib on HCC-induced angiogenesis depends on its inhibition of the production of angiogenic factors, probably involving a PTEN/Akt signaling pathway.
Collapse
Affiliation(s)
- Shu-Ichi Ueda
- Department of Medical Biochemistry, Graduate School of Medical Science and Station-II for Collaborative Research, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Sui G, Bonde P, Dhara S, Broor A, Wang J, Marti G, Feldmann G, Duncan M, Montgomery E, Maitra A, Harmon JW. Epidermal Growth Factor Receptor and Hedgehog Signaling Pathways Are Active in Esophageal Cancer Cells From Rat Reflux Model. J Surg Res 2006; 134:1-9. [PMID: 16488438 DOI: 10.1016/j.jss.2005.12.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2004] [Revised: 12/07/2005] [Accepted: 12/27/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Advancements in experimental therapeutics for esophageal cancers have been hampered by the lack of a reliable preclinical model that recapitulates the biology of human cancer, including in vivo growth in an animal model. METHODS Bilious reflux was induced by esophago-jejunostomy in Sprague-Dawley rats. Nine of 12 (75%) Sprague-Dawley rats developed squamous or adenosquamous cancers, and three cell lines were created by in vitro propagation of freshly resected tumors, JA and JB lines from one cancer, and the AMY cell line from another. We subsequently tested the ability of these cell lines to propagate long-term in vitro and form xenografts in vivo, both hallmarks of transformed cells. In addition, we determined the effects of small molecule inhibitors of two important oncogenic pathways-the epidermal growth factor receptor (EGFR) and Hedgehog (Hh) signaling pathways, in vitro, as a "proof of principle" of using these unique cell lines for developing targeted therapies for esophageal cancer. Mechanism-based growth inhibition was assessed by down-regulation of activated downstream targets of EGFR in the case of Iressa, and by Hh luciferase reporter activity with cyclopamine. RESULTS JA, JB, and AMY cell lines were able to grow continuously in vitro and consistently form xenografts in vivo in athymic mice, both subcutaneously, as well as in the "orthotopic" location at the gastroesophageal serosal junction (n = 2 mice per line, six of six engrafted). By histology, the tumors grow in vivo as well-differentiated keratinizing squamous cell carcinomas. JB cells had the highest expression of EGFR protein and also the most profound response to Iressa (gefitinib), an EGFR inhibitor (IC50 < 1 microm). Growth inhibition by Iressa was mirrored functionally by down-regulation of activated targets of the EGFR pathway, phospho-ERK1/2 and phospho-MEK levels. AMY cells expressed approximately 900-fold elevation of the Hh ligand, Indian Hh (Ihh), compared with normal esophageal epithelium, whereas expression of another Hh ligand, Sonic Hh (Shh), was not detected. On treatment with the specific Hh small molecule inhibitor cyclopamine, AMY cells demonstrated growth inhibition, which was accompanied by significant down-regulation of endogenous Hh luciferase reporter activity at 24 h and increased apoptosis in treated cells. CONCLUSIONS We have established a model of esophageal carcinogenesis, capable of long-term in vitro and in vivo passage, and demonstrated therapeutic potential of targeting the EGFR and Hh pathways in the cell lines created from the rodent cancers. These unique cell lines should provide a platform for rapid preclinical validation of novel therapeutics for esophageal cancers.
Collapse
Affiliation(s)
- Guoping Sui
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lee TL, Yeh J, Van Waes C, Chen Z. Epigenetic modification of SOCS-1 differentially regulates STAT3 activation in response to interleukin-6 receptor and epidermal growth factor receptor signaling through JAK and/or MEK in head and neck squamous cell carcinomas. Mol Cancer Ther 2006; 5:8-19. [PMID: 16432158 DOI: 10.1158/1535-7163.mct-05-0069] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) has been reported to be activated by interleukin-6 receptor (IL-6R) or epidermal growth factor receptor (EGFR) in head and neck squamous cell carcinomas (HNSCC), which may have important implications for responsiveness to therapeutics targeted at EGFR, IL-6R, or intermediary kinases. Suppressor of cytokine signaling-1 (SOCS-1) has been implicated recently in the negative regulation of IL-6R/Janus-activated kinase (JAK)-mediated activation of STAT3, suggesting that SOCS-1 could affect alternative activation of STAT3 by EGFR, IL-6R, and associated kinases. We investigated whether epigenetic modification of SOCS-1 affects STAT3 activation in response to IL-6R-, EGFR-, JAK-, or mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK)-mediated signal activation. STAT3 was predominantly activated by IL-6R via Jak1/Jak2 in HNSCC lines UMSCC-9 and UMSCC-38 in association with transcriptional silencing of SOCS-1 by hypermethylation. In UMSCC-11A cells with unmethylated SOCS-1, STAT3 activation was regulated by both EGFR and IL-6R via a JAK-independent pathway involving MEK. Pharmacologic inhibitors of JAK and MEK and expression of SOCS-1 following demethylation or transient transfection inhibited STAT3 activation and cell proliferation and induced cell apoptosis in corresponding cell lines. Hypermethylation of SOCS-1 was found in about one-third of human HNSCC tissues, making it a potentially relevant marker for STAT-targeted therapy in HNSCC patients. We conclude that SOCS-1 methylation status can differentially affect STAT3 activation by IL-6R and EGFR through JAK or MEK in different HNSCC and response to pharmacologic antagonists. Identifying the potential factors and the regulatory pathways in STAT3 activation has important implications for the development and selection of molecularly targeted therapy in HNSCC.
Collapse
Affiliation(s)
- Tin Lap Lee
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, 10/5D55, MSC-1419, Bethesda, MD 20892-1419, USA
| | | | | | | |
Collapse
|
23
|
Siegel-Lakhai WS, Beijnen JH, Schellens JHM. Current knowledge and future directions of the selective epidermal growth factor receptor inhibitors erlotinib (Tarceva) and gefitinib (Iressa). Oncologist 2006; 10:579-89. [PMID: 16177282 DOI: 10.1634/theoncologist.10-8-579] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Gefitinib (Iressa); AstraZeneca Pharmaceuticals, Wilmington, DE, http://www.astrazeneca-us.com) and erlotinib (Tarceva); OSI Pharmaceuticals, Inc., Melville, NY, http://www.osip.com) are so-called small molecules that selectively inhibit epidermal growth factor receptor (EGFR) tyrosine kinase activity. Both drugs received registration approval by the U.S. Food and Drug Administration (FDA) for the second- and third-line treatment of non-small cell lung cancer (NSCLC), but the failure of gefitinib to show a survival advantage over placebo has resulted in a discussion about the registration of gefitinib. Recently published results have revealed that mutations in the tyrosine kinase domain of EGFR are strongly associated with increased gefitinib and erlotinib sensitivity in patients with advanced NSCLC. Here, we present the current knowledge and the future directions of the EGFR tyrosine kinase inhibitors gefitinib and erlotinib.
Collapse
|
24
|
Van Cutsem E, Labianca R, Cognetti F, Tabernero J. Targeted therapies for patients with advanced colorectal cancer: focus on cetuximab. Target Oncol 2006. [DOI: 10.1007/s11523-005-0001-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
25
|
Jung HS, Byun GW, Lee KE, Mun YC, Nam SH, Kwon JM, Lee SN, Im SA, Seong CM, Lee SN. Gefitinib trial in a fanconi's anemia patient with multiple squamous cell carcinomas and hepatocellular carcinoma. Cancer Res Treat 2005; 37:370-3. [PMID: 19956375 DOI: 10.4143/crt.2005.37.6.370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Accepted: 10/26/2005] [Indexed: 11/21/2022] Open
Abstract
FA (Fanconi's Anemia) is an autosomal recessive disorder that is characterized by pancytopenia with bone marrow hypoplasia, diverse congenital abnormalities and an increased predisposition towards malignancy. The mainstay of the treatment for these cancers has been surgery, because of the hypersensitive reactions of FA patients to DNA cross- linking agents or radiation. Therefore, there has been no effective therapy for advanced squamous cell carcinoma. We report here on a patient suffering from advanced multiple squamous cell carcinoma and hepatocellular carcinoma along with an FA, and this patient was treated with gefitinib.
Collapse
Affiliation(s)
- Hae Sun Jung
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mimori K, Yamashita K, Ohta M, Yoshinaga K, Ishikawa K, Ishii H, Utsunomiya T, Barnard GF, Inoue H, Mori M. Coexpression of matrix metalloproteinase-7 (MMP-7) and epidermal growth factor (EGF) receptor in colorectal cancer: an EGF receptor tyrosine kinase inhibitor is effective against MMP-7-expressing cancer cells. Clin Cancer Res 2005; 10:8243-9. [PMID: 15623600 DOI: 10.1158/1078-0432.ccr-04-0849] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE Matrix metalloproteinase-7 (MMP-7) plays an important role in carcinoma invasion and metastasis of cancer. Recent studies focus on diverse roles of MMP-7, other than as a protease, during cancer progression. MMP-7 activates the epidermal growth factor (EGF) receptor by releasing an EGF ligand, tumor growth factor (TGF)-alpha. EXPERIMENTAL DESIGN We examined expression of MMP-7 and EGF receptor in an immunohistochemical study of 40 colorectal cancer (CRC) cases. To determine the relationship between the EGF receptor and MMP-7, with a potential curative application, we compared the antitumor activity of the EGF receptor tyrosine kinase inhibitor (gefitinib) between MMP-7 transfectant, KYSE150 and HT29, and control cells. RESULTS We found a statistically significant correlation (P = 0.04) between MMP-7 and activated (phosphorylated) EGF receptor expression, both being positive in six (15%) cases. Gefitinib reduced the cell number ratio more for MMP-7 transfectant than mock cells, and the proportion of apoptotic cells was 1.5 times higher in MMP-7 transfectant than mock cells by annexin/propidium iodide staining. This was mediated by activation of a TGF-beta signal as confirmed by the abundant expression of TGF-beta protein, the cytoplasmic to nuclear translocation of Smad4 protein by the administration of gefitinib, and the quantitative assay of the plasminogen activator inhibitor-1 promoter/luciferase construction. CONCLUSIONS We propose that there are some cancers with up-regulated MMP-7 expression that leads to the activation of apoptotic activity of TGF-beta, which is susceptible to treatment with EGF receptor tyrosine kinase inhibitor.
Collapse
Affiliation(s)
- Koshi Mimori
- Department of Surgery, Medical Institute of Bioregulation, Kyushu University, Beppu, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Araujo RP, Petricoin EF, Liotta LA. A mathematical model of combination therapy using the EGFR signaling network. Biosystems 2005; 80:57-69. [PMID: 15740835 DOI: 10.1016/j.biosystems.2004.10.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Revised: 10/12/2004] [Accepted: 10/13/2004] [Indexed: 10/26/2022]
Abstract
An increasing awareness of the significance of abnormal signal transduction in tumors and the concomitant development of target-based drugs to selectively modulate aberrantly-activated signaling pathways has given rise to a variety of promising new strategies in cancer treatment. This paper uses mathematical modeling to investigate a novel type of combination therapy in which multiple nodes in a signaling cascade are targeted simultaneously with selective inhibitors, pursuing the hypothesis that such an approach may induce the desired signal attenuation with lower doses of the necessary agents than when one node is targeted in isolation. A mathematical model is presented which builds upon previous theoretical work on EGFR signaling, simulating the effect of administering multiple kinase inhibitors in various combinations. The model demonstrates that attenuation of biochemical signals is significantly enhanced when multiple upstream processes are inhibited, in comparison with the inhibition of a single upstream process. Moreover, this enhanced attenuation is most pronounced in signals downstream of serially-connected target points. In addition, the inhibition of serially-connected processes appears to have a supra-additive (synergistic) effect on the attenuation of downstream signals, owing to the highly non-linear relationships between network parameters and signals.
Collapse
Affiliation(s)
- R P Araujo
- FDA-NCI Clinical Proteomics Program, Laboratory of Pathology, Center for Cancer Research, NCI/NIH, 8800 Rockville Pike, Building 29A, HFM 710, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
28
|
von Minckwitz G, Jonat W, Fasching P, du Bois A, Kleeberg U, Lück HJ, Kettner E, Hilfrich J, Eiermann W, Torode J, Schneeweiss A. A multicentre phase II study on gefitinib in taxane- and anthracycline-pretreated metastatic breast cancer. Breast Cancer Res Treat 2005; 89:165-72. [PMID: 15692759 DOI: 10.1007/s10549-004-1720-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is considered to be a viable drug target in a variety of solid tumors. The clinical benefit and safety of the EGFR tyrosine kinase inhibitor gefitinib ('Iressa')1 was evaluated in this Phase II, multicentre study of patients with taxane and anthracycline pretreated, metastatic breast cancer. METHODS Gefitinib (500 mg/day) was given to 58 patients until disease progression. Primary endpoint was the clinical response rate to the study treatment. RESULTS One patient (1.7%) had objective partial tumor response of her liver and pleural metastasis. Fifty-seven patients (98.3%) were non-responders with 52 patients (89.7%) having progressive disease and five patients (8.6%) were not evaluable. Two patients reported a significant improvement in pain at metastatic sites (1 liver, 1 bone). The median time to progression was 61 days (95% CI : 54-82 days) and the proportion of patients alive and progression free at 6 months at trial closure was 1.8% (95% CI : 0.0-5.2%). The median survival time was 357 days (95% CI : 257-441 days). Fifty-four patients (93.1%) discontinued study medication prematurely due to disease progression and three (5.2%) due to adverse events (diarrhea, pruritus, peripheral edema). CONCLUSIONS Gefitinib monotherapy at 500 mg daily did not appear to be efficacious in the treatment of heavily pretreated metastatic breast cancer patients. It was well tolerated and the side effect profile was as expected from current knowledge of the drug. There was no correlation between EGFR expression and response in this study.
Collapse
Affiliation(s)
- Gunter von Minckwitz
- German Breast Group/Universitäts-Frauenklinik Frankfurt and Klinikum der Goethe-Universitaet, Zentrum für Frauenheilkunde and Geburtshilfe, Neu-Isenburg/Frankfurt, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Camirand A, Zakikhani M, Young F, Pollak M. Inhibition of insulin-like growth factor-1 receptor signaling enhances growth-inhibitory and proapoptotic effects of gefitinib (Iressa) in human breast cancer cells. Breast Cancer Res 2005; 7:R570-9. [PMID: 15987464 PMCID: PMC1175059 DOI: 10.1186/bcr1028] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2004] [Revised: 02/23/2005] [Accepted: 03/18/2005] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Gefitinib (Iressa, ZD 1839, AstraZeneca) blocks the tyrosine kinase activity of the epidermal growth factor receptor (EGFR) and inhibits proliferation of several human cancer cell types including breast cancer. Phase II clinical trials with gefitinib monotherapy showed an objective response of 9 to 19% in non-small-cell lung cancer patients and less than 10% for breast cancer, and phase III results have indicated no benefit of gefitinib in combination with chemotherapy over chemotherapy alone. In order to improve the antineoplastic activity of gefitinib, we investigated the effects of blocking the signalling of the insulin-like growth factor 1 receptor (IGF-1R), a tyrosine kinase with a crucial role in malignancy that is coexpressed with EGFR in most human primary breast carcinomas. METHODS AG1024 (an inhibitor of IGF-1R) was used with gefitinib for treatment of MDA468, MDA231, SK-BR-3, and MCF-7 breast cancer lines, which express similar levels of IGF-1R but varying levels of EGFR. Proliferation assays, apoptosis induction studies, and Western blot analyses were conducted with cells treated with AG1024 and gefitinib as single agents and in combination. RESULTS Gefitinib and AG1024 reduced proliferation in all lines when used as single agents, and when used in combination revealed an additive-to-synergistic effect on cell growth inhibition. Flow cytometry measurements of cells stained with annexin V-propidium iodide and cells stained for caspase-3 activation indicated that adding an IGF-1R-targeting strategy to gefitinib results in higher levels of apoptosis than are achieved with gefitinib alone. Gefitinib either reduced or completely inhibited p42/p44 Erk kinase phosphorylation, depending on the cell line, while Akt phosphorylation was reduced by a combination of the two agents. Overexpression of IGF-1R in SK-BR-3 cells was sufficient to cause a marked enhancement in gefitinib resistance. CONCLUSION These results indicate that IGF-1R signaling reduces the antiproliferative effects of gefitinib in several breast cancer cell lines, and that the addition of an anti-IGF-1R strategy to gefitinib treatment may be more effective than a single-agent approach.
Collapse
Affiliation(s)
- Anne Camirand
- Lady Davis Institute for Medical Research and Department of Oncology, McGill University, Montréal, QC, Canada
| | - Mahvash Zakikhani
- Lady Davis Institute for Medical Research and Department of Oncology, McGill University, Montréal, QC, Canada
- Biology Department, Faculty of Science, Az-Zahra University, Vanak, Tehran, Iran
| | - Fiona Young
- Lady Davis Institute for Medical Research and Department of Oncology, McGill University, Montréal, QC, Canada
| | - Michael Pollak
- Lady Davis Institute for Medical Research and Department of Oncology, McGill University, Montréal, QC, Canada
| |
Collapse
|
30
|
Reck M, Gatzemeier U. Gefitinib (‘Iressa’): a new therapy for advanced non-small-cell lung cancer. Respir Med 2005; 99:298-307. [PMID: 15733505 DOI: 10.1016/j.rmed.2004.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2004] [Accepted: 08/10/2004] [Indexed: 11/18/2022]
Abstract
Patients with advanced non-small-cell lung cancer (NSCLC) have a poor prognosis and suffer many disease-related symptoms. Treatments should therefore aim to palliate these symptoms as well as improve overall quality of life. Gefitinib ("Iressa") is a novel epidermal growth factor receptor tyrosine kinase inhibitor that targets cell signaling involved in tumor growth and differentiation. In Phase II trials in recurrent NSCLC, gefitinib provided objective responses and symptom relief, and was generally well tolerated. Now approved for the treatment of advanced NSCLC in over 25 countries, including Japan and the USA, gefitinib has demonstrated significant clinical benefit in a disease area with limited treatment options.
Collapse
Affiliation(s)
- Martin Reck
- Department of Thoracic Oncology, Hospital Grosshansdorf, Woehrendamm 80, Grosshansdorf 22927, Germany.
| | | |
Collapse
|
31
|
Vlachtsis K, Nikolaou A, Markou K, Fountzilas G, Daniilidis I. Clinical and molecular prognostic factors in operable laryngeal cancer. Eur Arch Otorhinolaryngol 2005; 262:890-8. [PMID: 15739081 DOI: 10.1007/s00405-005-0916-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Accepted: 12/10/2004] [Indexed: 01/05/2023]
Abstract
Many factors affect the prognosis in operable laryngeal squamous cell carcinoma (LSCC). Many clinical factors have been implicated in tumor recurrence and poor survival of the patients. The aim of the present study is to investigate the demographic, clinical and histological characteristics as prognostic factors. Moreover, our aim is to analyze the role of modern molecular biomarkers in the prognosis of patients with LSCC. One hundred patients with operable laryngeal carcinoma underwent surgery as primary treatment between April 1999 and April 2002. Ninety-four of them were men and 6 women, with a median age of 62 years (39-77). All demographic data of the patients were recorded. Staging of the tumor revealed 20 cases with T2 cancer, 46 cases with T3 and 34 cases with T4, while N classification included 91 patients with N0 tumor, 3 with N1 and 6 with N2. Among the 100 cases, 47 were located in the glottis, 46 in the supraglottic region and 7 were transglottic. Histology grading revealed 35 cases of grade G1, 50 cases of G2 and 15 cases of G3. Postoperatively, all patients were followed regularly for the possibility of tumor relapse, with a median follow-up period of 40.2 months (4.8-58.4). During the operation, a tissue specimen was collected from the tumor. The specimens were used for RNA and DNA extraction. Isolated RNA was used to investigate the expression of wt-p53, bcl-2, VEGF and EGFR by the reverse transcriptase PCR method (RT-PCR) using specific primers, while genomic DNA was used for the detection of EBV and HPV (16/18 subtypes) by the consensus primer-mediated polymerase chain reaction method (PCR). All data such as tumor recurrence and survival were recorded. Statistical analysis was performed using the SPSS and STATA statistical packages in order to investigate the role of all clinical and molecular factors and their combinations as significant prognostic markers. The tumor recurrence rate was 31%, while the tumor associated death rate was 27% and total death rate 30%. Univariate analysis for overall survival showed significance for the T stage, TNM stage and site of the tumor. Univariate analysis for the time to progression showed significance for the T stage, N stage, TNM stage, site of the tumor and tumors simultaneously positive for EGFR and VEGF, while EGFR expression was borderline insignificant. Multivariate analysis revealed TNM stage as the only significant factor for overall survival, and TNM stage, site of the tumor and EGFR expression as significant factors for time to progression. The molecular biomarkers EGFR and VEGF have a prognostic significance in laryngeal cancer in addition to the established clinical prognostic factors such as the stage and site of the tumor. These markers, apart from their role in carcinogenesis, seem to play an important role in tumor relapse.
Collapse
Affiliation(s)
- Konstantinos Vlachtsis
- Department of Otorhinolaryngology Head and Neck Surgery, AHEPA Hospital, Aristotle University of Thessaloniki, Greece.
| | | | | | | | | |
Collapse
|
32
|
Mimeault M, Jouy N, Depreux P, Hénichart JP. Synergistic antiproliferative and apoptotic effects induced by mixed epidermal growth factor receptor inhibitor ZD1839 and nitric oxide donor in human prostatic cancer cell lines. Prostate 2005; 62:187-99. [PMID: 15389789 DOI: 10.1002/pros.20138] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND The specific inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase, ZD1839 induces potent antitumoral effects on several advanced cancer types. The present study was undertaken to determine whether the combination of ZD1839 with an agent donating nitric oxide (NO(*)), sodium nitroprusside (SNP) results in a synergy of anticarcinogenic responses on metastatic prostate cancer (PC) cells. METHODS The antiproliferative and apoptotic/necrotic effects of ZD1839 and SNP alone or in combination were estimated on EGF- and serum-stimulated LNCaP, DU145, and PC3 cells by MTT growth tests, trypan blue dye exclusion method, and flow cytometric analyses. Moreover, the cellular ceramide levels were evaluated by the diacylglycerol kinase enzymatic method and the amounts of cytosolic cytochrome c by ELISA assays. RESULTS ZD1839 and SNP alone or in combination at lower concentrations induced an inhibition of EGF- and serum-stimulated growth of LNCaP, DU145, and PC3 concomitant with an arrest in the G1 phase of cellular cycle. Interestingly, the mixed ZD1839 and SNP also caused a more substantial apoptotic/necrotic death of these PC cells as compared to drugs alone. Moreover, we have observed that an inhibition of acidic sphingomyelinase, hydrogen peroxide (H(2)O(2)) accumulation and caspase cascades results in a significant reduction of apoptotic/necrotic death induced by mixed ZD1839 and SNP in EGF-stimulated PC3 cells. In addition, the combined ZD1839 plus SNP also induced a higher cellular ceramide and reactive oxygen species (ROS) production, mitochondrial transmembrane potential decrease, and cytochrome c amount released into cytosol as compared to drugs alone. CONCLUSIONS The simultaneous use of EGFR inhibitor and compound releasing NO(*) might lead to a synergy in the ceramide and ROS production which might cause cellular membrane damages resulting in a massive apoptotic/necrotic death of metastatic PC cells.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | |
Collapse
|
33
|
Nicholson RI, Hutcheson IR, Britton D, Knowlden JM, Jones HE, Harper ME, Hiscox SE, Barrow D, Gee JMW. Growth factor signalling networks in breast cancer and resistance to endocrine agents: new therapeutic strategies. J Steroid Biochem Mol Biol 2005; 93:257-62. [PMID: 15860268 DOI: 10.1016/j.jsbmb.2004.12.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent evidence demonstrates that growth factor networks are highly interactive with the estrogen receptor (ER) in the control of breast cancer growth and development. As such, tumor responses to anti-hormones are likely to be a composite of the ER and growth factor inhibitory activity of these agents, with alterations/aberrations in growth factor signalling providing a mechanism for the development of anti-hormone resistance. In this light, the current article focuses on illustrating the relationship between growth factor signalling and anti-hormone failure in our in-house tumor models of breast cancer and describes how we are now beginning to successfully target their actions to improve the effects of anti-hormonal drugs and to block aggressive disease progression.
Collapse
Affiliation(s)
- R I Nicholson
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3XF, Wales, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Schiff BA, McMurphy AB, Jasser SA, Younes MN, Doan D, Yigitbasi OG, Kim S, Zhou G, Mandal M, Bekele BN, Holsinger FC, Sherman SI, Yeung SC, El-Naggar AK, Myers JN. Epidermal Growth Factor Receptor (EGFR) Is Overexpressed in Anaplastic Thyroid Cancer, and the EGFR Inhibitor Gefitinib Inhibits the Growth of Anaplastic Thyroid Cancer. Clin Cancer Res 2004; 10:8594-602. [PMID: 15623643 DOI: 10.1158/1078-0432.ccr-04-0690] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE No effective treatment options currently are available to patients with anaplastic thyroid cancer (ATC), resulting in high mortality rates. Epidermal growth factor (EGF) has been shown to play a role in the pathogenesis of many types of cancer, and its receptor (EGFR) provides an attractive target for molecular therapy. EXPERIMENTAL DESIGN The expression of EGFR was determined in ATC in vitro and in vivo and in human tissue arrays of ATC. We assessed the potential of the EGFR inhibitor gefitinib ("Iressa," ZD1839) to inhibit EGFR activation in vitro and in vivo, inhibit ATC cellular proliferation, induce apoptosis, and reduce the growth of ATC cells in vivo when administered alone and in combination with paclitaxel. RESULTS EGFR was overexpressed in ATC cell lines in vitro and in vivo and in human ATC specimens. Activation of EGFR by EGF was blocked by the addition of gefitinib. In vitro studies showed that gefitinib greatly inhibited cellular proliferation and induced apoptosis in ATC cell lines and slowed tumor growth in a nude mouse model of thyroid carcinoma cells injected subcutaneously. CONCLUSIONS ATC cells consistently overexpress EGFR, rendering this receptor a potential target for molecular therapy. Gefitinib effectively blocks activation of EGFR by EGF, inhibits ATC cellular proliferation, and induces apoptosis in vitro. Our in vivo results show that gefitinib has significant antitumor activity against ATC in a subcutaneous nude mouse tumor model and therefore is a potential candidate for human clinical trials.
Collapse
Affiliation(s)
- Bradley A Schiff
- Department of Head and Neck Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Knight LA, Di Nicolantonio F, Whitehouse P, Mercer S, Sharma S, Glaysher S, Johnson P, Cree IA. The in vitro effect of gefitinib ('Iressa') alone and in combination with cytotoxic chemotherapy on human solid tumours. BMC Cancer 2004; 4:83. [PMID: 15560844 PMCID: PMC535559 DOI: 10.1186/1471-2407-4-83] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Accepted: 11/23/2004] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Activation of the epidermal growth factor receptor (EGFR) triggers downstream signaling pathways that regulate many cellular processes involved in tumour survival and growth. Gefitinib ('Iressa') is an orally active tyrosine kinase inhibitor (TKI) targeted to the ATP-binding domain of EGFR (HER1; erbB1). METHODS In this study we have used a standardised ATP-based tumour chemosensitivity assay (ATP-TCA) to measure the activity of gefitinib alone or in combination with different cytotoxic drugs (cisplatin, gemcitabine, oxaliplatin and treosulfan) against a variety of solid tumours (n = 86), including breast, colorectal, oesophageal and ovarian cancer, carcinoma of unknown primary site, cutaneous and uveal melanoma, non-small cell lung cancer (NSCLC) and sarcoma. The IC50 and IC90 were calculated for each single agent or combination. To allow comparison between samples the IndexSUM was calculated based on the percentage tumour growth inhibition (TGI) at each test drug concentration (TDC). Gefitinib was tested at concentrations ranging from 0.0625-2 microM (TDC = 0.446 microg/ml). This study represents the first use of a TKI in the assay. RESULTS There was heterogeneity in the degree of TGI observed when tumours were tested against single agent gefitinib. 7% (6/86) of tumours exhibited considerable inhibition, but most showed a more modest response resulting in a low TGI. The median IC50 value for single agent gefitinib in all tumours tested was 3.98 microM. Interestingly, gefitinib had both positive and negative effects when used in combination with different cytotoxics. In 59% (45/76) of tumours tested, the addition of gefitinib appeared to potentiate the effect of the cytotoxic agent or combination (of these, 11% (5/45) had a >50% decrease in their IndexSUM). In 38% of tumours (29/76), the TGI was decreased when the combination of gefitinib + cytotoxic was used in comparison to the cytotoxic alone. In the remaining 3% (2/76) there was no change observed. CONCLUSION The in vitro model suggests that gefitinib may have differential effects in response to concomitant cytotoxic chemotherapy with the agents tested during this study. The mechanism involved may relate to the effect of TKIs on growth rate versus their effect on the ability of the cell to survive the stimulus to apoptosis produced by chemotherapy.
Collapse
Affiliation(s)
- Louise A Knight
- Translational Oncology Research Centre, Department of Histopathology, Queen Alexandra Hospital, Portsmouth, UK
| | - Federica Di Nicolantonio
- Translational Oncology Research Centre, Department of Histopathology, Queen Alexandra Hospital, Portsmouth, UK
| | | | | | - Sanjay Sharma
- Translational Oncology Research Centre, Department of Histopathology, Queen Alexandra Hospital, Portsmouth, UK
| | - Sharon Glaysher
- Translational Oncology Research Centre, Department of Histopathology, Queen Alexandra Hospital, Portsmouth, UK
| | - Penny Johnson
- Translational Oncology Research Centre, Department of Histopathology, Queen Alexandra Hospital, Portsmouth, UK
| | - Ian A Cree
- Translational Oncology Research Centre, Department of Histopathology, Queen Alexandra Hospital, Portsmouth, UK
| |
Collapse
|
37
|
Sumitomo M, Asano T, Asakuma J, Asano T, Horiguchi A, Hayakawa M. ZD1839 modulates paclitaxel response in renal cancer by blocking paclitaxel-induced activation of the epidermal growth factor receptor-extracellular signal-regulated kinase pathway. Clin Cancer Res 2004; 10:794-801. [PMID: 14760103 DOI: 10.1158/1078-0432.ccr-0948-03] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We evaluated the antitumor activity of ZD1839, a selective epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor, in combination with paclitaxel in human renal cell carcinomas (RCCs). EXPERIMENTAL DESIGN Eight human RCC lines and the surgical specimens obtained from 10 RCC patients were used. The protein expression was detected by Western blotting, immunohistochemistry and/or flow cytometry. Apoptosis was evaluated by flow cytometry and fragmented DNA ELISA. SKRC-49 tumor xenografts in athymic nude mice were treated with ZD1839 and/or paclitaxel, and tumor volume was determined RESULTS EGFR protein was expressed and phosphorylated in eight RCC lines and EGFR expression was markedly increased in RCC specimens compared with adjacent normal renal tissues. Treatment of SKRC-49 with 1 micro M ZD1839 resulted in a marked decrease in the phosphorylation of EGFR but not of HER-2. Treatment of SKRC-49 with ZD1839 in combination with 5 nM paclitaxel resulted in a significant increase in apoptotic cell number compared with paclitaxel alone, whereas ZD1839 alone failed to induce apoptosis. Although administration of ZD1839 or paclitaxel resulted in a transient growth inhibition in SKRC-49 xenografts, significant tumor regrowth delay was observed when paclitaxel was combined with ZD1839. Paclitaxel phosphorylated extracellular signal-regulated kinase through EGFR activation predominantly in cancer cells. ZD1839 promoted paclitaxel-induced Bcl-2 down-regulation resulting in promoting apoptosis by blocking paclitaxel-induced activation of the EGFR-extracellular signal-regulated kinase antiapoptotic pathway independent of Akt activity in SKRC-49. CONCLUSIONS Our findings support the idea that the significant clinical benefit is obtained from ZD1839 in combination with paclitaxel for the treatment of RCC.
Collapse
Affiliation(s)
- Makoto Sumitomo
- Department of Urology, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | | | | | | | | | | |
Collapse
|
38
|
Nicholson RI, Hutcheson IR, Knowlden JM, Jones HE, Harper ME, Jordan N, Hiscox SE, Barrow D, Gee JMW. Nonendocrine Pathways and Endocrine Resistance. Clin Cancer Res 2004; 10:346S-54S. [PMID: 14734490 DOI: 10.1158/1078-0432.ccr-031206] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
An increasing body of evidence demonstrates that growth factor networks are highly interactive with estrogen receptor signaling in the control of breast cancer growth. As such, tumor responses to antiestrogens are likely to be a composite of the estrogen receptor and growth factor-inhibitory activity of these agents, with alterations/aberrations in growth factor signaling providing a mechanism for the development of antiestrogen resistance. In this light, the current article focuses on illustrating the relationship between growth factor signaling and antiestrogen failure in our in-house tumor models of breast cancer and describing how we are now beginning to successfully target growth factor activity to improve the effects of antiestrogen drugs and to block aggressive disease progression.
Collapse
Affiliation(s)
- Robert I Nicholson
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff, Wales, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Blackledge G. Growth factor receptor tyrosine kinase inhibitors; clinical development and potential for prostate cancer therapy. J Urol 2003; 170:S77-83; discussion S83. [PMID: 14610415 DOI: 10.1097/01.ju.0000095022.80033.d3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE The development of effective, novel, targeted cancer therapies with minimal side effects has long been a goal in cancer research. A key group of targets identified for drug development consists of the receptor tyrosine kinases, which have pivotal roles in the growth factor signaling that is subverted in carcinogenesis and in the host processes, such as angiogenesis, involved in tumor progression. MATERIALS AND METHODS A literature review of the role of receptor tyrosine kinases in human malignancies is followed by a discussion of the potential use of inhibitors of receptor tyrosine kinases as anticancer therapy, focusing on the epidermal growth factor receptor tyrosine kinase inhibitor gefitinib (Iressa, ZD1839, AstraZeneca, Macclesfield, United Kingdom). RESULTS Several small molecule inhibitors that are specific to individual receptor tyrosine kinases have been developed and a number of these potential anticancer agents are progressing through clinical trials. Various surrogate end points are being assessed to demonstrate the activity of these inhibitors against their targets. Results from studies of gefitinib alone and with the antiandrogen bicalutamide in both hormone dependent and independent prostate tumor xenografts suggested that gefitinib may have potential as monotherapy and combination therapy in the treatment of both forms of the disease. Gefitinib is currently undergoing further preclinical and clinical evaluation for the treatment of prostate cancer. CONCLUSIONS A number of tyrosine kinase inhibitors, including gefitinib, are progressing through clinical development and are beginning to provide new treatment options for a range of malignancies.
Collapse
|
40
|
Espinosa E, Zamora P, Feliu J, González Barón M. Classification of anticancer drugs—a new system based on therapeutic targets. Cancer Treat Rev 2003; 29:515-23. [PMID: 14585261 DOI: 10.1016/s0305-7372(03)00116-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The arrival of a great number of new antineoplastic agents has made it necessary to reclassify all of them. Anticancer drugs may act at different levels: cancer cells, endothelium, extracellular matrix, the immune system or host cells. The tumour cell can be targeted at the DNA, RNA or protein level. Most classical chemotherapeutic agents interact with tumour DNA, whereas monoclonal antibodies and small molecules are directed against proteins. The endothelium and extracellular matrix may be affected also by specific antibodies and small molecules.
Collapse
|
41
|
Gee JMW, Harper ME, Hutcheson IR, Madden TA, Barrow D, Knowlden JM, McClelland RA, Jordan N, Wakeling AE, Nicholson RI. The antiepidermal growth factor receptor agent gefitinib (ZD1839/Iressa) improves antihormone response and prevents development of resistance in breast cancer in vitro. Endocrinology 2003; 144:5105-17. [PMID: 12960029 DOI: 10.1210/en.2003-0705] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although many estrogen receptor-positive breast cancers initially respond to antihormones, responses are commonly incomplete with resistance ultimately emerging. Delineation of signaling mechanisms underlying these phenomena would allow development of therapies to improve antihormone response and compromise resistance. This in vitro investigation in MCF-7 breast cancer cells examines whether epidermal growth factor receptor (EGFR) signaling limits antiproliferative and proapoptotic activity of antihormones and ultimately supports development of resistance. It addresses whether the anti-EGFR agent gefitinib (ZD1839/Iressa; TKI: 1 mum) combined with the antihormones 4-hydroxytamoxifen (TAM: 0.1 mum) or fulvestrant (Faslodex; 0.1 mum) enhances growth inhibition and prevents resistance. TAM significantly suppressed MCF-7 growth over wk 2-5, reducing proliferation detected by immunocytochemistry and fluorescence-activated cell sorter cell cycle analysis. A modest apoptotic increase was observed by fluorescence-activated cell sorter and fluorescence microscopy, with incomplete bcl-2 suppression. EGFR induction occurred during TAM response, as measured by immunocytochemistry and Western blotting, with EGFR-positive, highly proliferative resistant growth subsequently emerging. Although TKI alone was ineffective on growth, TAM plus TKI cotreatment exhibited superior antigrowth activity vs. TAM, with no viable cells by wk 12. Cotreatment was more effective in inhibiting proliferation, promoting apoptosis, and eliminating bcl-2. Cotreatment blocked EGFR induction, markedly depleted ERK1/2 MAPK and protein kinase B phosphorylation, and prevented emergence of EGFR-positive resistance. Faslodex plus TKI cotreatment was also a superior antitumor strategy. Thus, increased EGFR evolves during treatment with antihormones, limiting their efficacy and promoting resistance. Gefitinib addition to antihormonal therapy could prove more effective in treating estrogen receptor-positive breast cancer and may combat development of resistance.
Collapse
Affiliation(s)
- J M W Gee
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3XF, Wales, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mahtouk K, Jourdan M, De Vos J, Hertogh C, Fiol G, Jourdan E, Rossi JF, Klein B. An inhibitor of the EGF receptor family blocks myeloma cell growth factor activity of HB-EGF and potentiates dexamethasone or anti-IL-6 antibody-induced apoptosis. Blood 2003; 103:1829-37. [PMID: 14576062 PMCID: PMC2386161 DOI: 10.1182/blood-2003-05-1510] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We previously found that some myeloma cell lines express the heparin-binding epidermal growth factor-like growth factor (HB-EGF) gene. As the proteoglycan syndecan-1 is an HB-EGF coreceptor as well as a hallmark of plasma cell differentiation and a marker of myeloma cells, we studied the role of HB-EGF on myeloma cell growth. The HB-EGF gene was expressed by bone marrow mononuclear cells in 8 of 8 patients with myeloma, particularly by monocytes and stromal cells, but not by purified primary myeloma cells. Six of 9 myeloma cell lines and 9 of 9 purified primary myeloma cells expressed ErbB1 or ErbB4 genes coding for HB-EGF receptor. In the presence of a low interleukin-6 (IL-6) concentration, HB-EGF stimulated the proliferation of the 6 ErbB1+ or ErbB4+ cell lines, through the phosphatidylinositol 3-kinase/AKT (PI-3K/AKT) pathway. A pan-ErbB inhibitor blocked the myeloma cell growth factor activity and the signaling induced by HB-EGF. This inhibitor induced apoptosis of patients'myeloma cells cultured with their tumor environment. It also increased patients' myeloma cell apoptosis induced by an anti-IL-6 antibody or dexamethasone. The ErbB inhibitor had no effect on the interaction between multiple myeloma cells and stromal cells. It was not toxic for nonmyeloma cells present in patients' bone marrow cultures or for the growth of hematopoietic progenitors. Altogether, these data identify ErbB receptors as putative therapeutic targets in multiple myeloma.
Collapse
Affiliation(s)
- Karène Mahtouk
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - Michel Jourdan
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - John De Vos
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - Catherine Hertogh
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - Geneviève Fiol
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - Eric Jourdan
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - Jean-François Rossi
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
| | - Bernard Klein
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- Unité de thérapie cellulaire
CHRU MontpellierFR
- * Correspondence should be adressed to: Bernard Klein
| |
Collapse
|
43
|
Yang SF, Wang SL, Chai CY, Su YC, Fu OY, Chen CY. Intra-Abdominal Desmoplastic Small Round Cell Tumor with Elevated Serum CA 125: A Case Report. Kaohsiung J Med Sci 2003; 19:531-6. [PMID: 14620681 DOI: 10.1016/s1607-551x(09)70503-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Desmoplastic small round cell tumor (DSRCT) is a rare and highly aggressive tumor usually involving the peritoneum. It occurs more commonly in young males and is characterized by distinctive clinical, histologic, and immunophenotypic features. The histogenesis of DSRCT remains unknown. Coexpression of epithelial, mesenchymal, and neural antigens in the same cell provides evidence that DSRCT may arise from a primitive pluripotent stem cell with divergent differentiation. Recently, according to cytogenetic studies, some authors have proposed that the divergent differentiation of DSRCT may be the result of the fusion of Ewing's sarcoma gene and Wilms' tumor suppressor gene. Clinically, an elevated serum CA 125 concentration is found in some patients with DSRCT. We present the case of a 29-year-old man with diffuse intra-abdominal DSRCT and elevated serum CA 125 concentration and briefly review the relevant literature.
Collapse
Affiliation(s)
- Sheau-Fang Yang
- Department of Pathology, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | |
Collapse
|
44
|
Averbuch SD. The Impact of Gefitinib on Epidermal Growth Factor Receptor Signaling Pathways in Cancer. Clin Lung Cancer 2003; 5 Suppl 1:S5-S10. [PMID: 14641988 DOI: 10.3816/clc.2003.s.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ErbB family of receptor tyrosine kinases, of which the epidermal growth factor receptor (EGFR) is the prototype, is associated with the formation and malignant progression of most of the common solid tumors. These molecules play a key role in a complex network of signal transduction pathways that function in normal development as well as in neoplastic transformation. The EGFR and other family members are therefore promising targets for new anticancer therapies. In normal tissues, EGFR-tyrosine kinase (TK) activity is strictly controlled. However, in tumor cells, there are multiple mechanisms that can lead to increased or inappropriate EGFR-TK activity, including altered expression of EGFR, its ligand, or interacting molecules; decreased deactivation through phosphatases or downregulation; or mutation of the EGFR protein. Novel therapeutic approaches aimed at inhibiting increased EGFR-TK activity include antibodies that block the extracellular ligand-binding site, antibody or ligand fusion proteins that specifically target toxins to the tumor cells, or small-molecule TK inhibitors (TKIs) that act intracellularly to block downstream signal transduction from EGFR. Studies have shown that such blockade can lead to reduced cellular proliferation, inhibition of survival signals, and inhibition of tumor metastasis and angiogenesis. Additionally, some agents, including EGFR antibodies and TKIs such as gefitinib have been demonstrated to be effective against various human solid tumors in preclinical models and have shown activity in advanced non-small-cell lung cancer and other solid tumors.
Collapse
Affiliation(s)
- Steven D Averbuch
- Clinical Research, Oncology, AstraZeneca Pharmaceuticals, Wilmington, DE, USA.
| |
Collapse
|
45
|
Abstract
As investigators at the bench define the oncogenic pathways involved in ovarian cancer growth and invasion, clinical scientists will develop therapies to target these important pathways. The approach to the treatment of ovarian cancer will change to focus on strategies designed to eliminate minimal residual disease after primary therapy and in this way cure the disease. Concepts of consolidation after standard adjuvant chemotherapy and maintenance regimens will start to be evaluated for clinical efficacy. In addition, the focus on recurrent disease may change from repeated cytotoxic approaches to regimens chosen to prevent invasion or the development of drug resistance, changing the approach to ovarian cancer recurrence to a treatment plan of managing a chronic, but not imminently terminal, disease.
Collapse
Affiliation(s)
- Mary L Disis
- Department of Oncology, University of Washington, Box 356527, Seattle, WA 98195-6527, USA.
| | | |
Collapse
|
46
|
Abstract
OBJECTIVES To review malignant cell characteristics, which serve as the basis for the development of molecular, targeted cancer therapies. To provide an introduction and overview of new targeted agents, including monoclonal antibodies, enzyme inhibitors, antiangiogenic agents, gene therapy, and vaccines. DATA SOURCES Published scientific papers, review articles, and book chapters. CONCLUSION Through new understandings and theories of how cancer cells survive, thrive, and metastasize, researchers have created new targeted therapies for cancer treatment to minimize the harmful systemic effects of traditional therapy. IMPLICATIONS FOR NURSING PRACTICE As with any new therapeutic modality, scientific rationale and mechanism of action must be appreciated by health care staff to build a solid foundation for patient education and to provide astute management of acute and latent effects.
Collapse
Affiliation(s)
- Robin Gemmill
- Departments of Surgical Oncology and Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | | |
Collapse
|
47
|
Herbst RS, LoRusso PM, Purdom M, Ward D. Dermatologic Side Effects Associated with Gefitinib Therapy: Clinical Experience and Management. Clin Lung Cancer 2003; 4:366-9. [PMID: 14599302 DOI: 10.3816/clc.2003.n.016] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Gefitinib (ZD1839, Iressa ), a selective inhibitor of the epidermal growth factor receptor-tyrosine kinase, is currently in clinical trials to treat a variety of solid tumors. Similar side effects were seen in numerous clinical trials of gefitinib, including recent phase II trials (Iressa Dose Evaluation in Advanced Lung Cancer [IDEAL]-1 and IDEAL-2) in patients with advanced, previously treated non-small-cell lung cancer (NSCLC). The most frequent drug-related adverse effects reported in these trials were diarrhea, dry skin, acneiform rash, and nausea and vomiting. Recently, IDEAL-2 investigators and oncology nurses participated in a panel discussion on the management of dermatologic adverse effects associated with gefitinib treatment. These dermatologic effects were related to the mechanism of action of gefitinib and were not caused by a drug-related allergic reaction. In IDEAL-2, investigators managed the symptoms of dry skin and rash with a variety of treatments, including topical clindamycin, antibiotics, and corticosteroids. Most patients had a high level of tolerance for the dermatologic effects associated with gefitinib, which were milder than toxicities associated with chemotherapy. The severity of skin effects cycled over time during treatment but typically subsided after several weeks of gefitinib treatment. In general, dermatologic effects of gefitinib were easily managed, reversible, and well tolerated by these patients with advanced NSCLC.
Collapse
Affiliation(s)
- Roy S Herbst
- Department of Thoracic/Head and Neck Medical Oncology, M. D. Anderson Cancer Center, Houston, TX 77030-4009, USA.
| | | | | | | |
Collapse
|
48
|
Mimeault M, Pommery N, Hénichart JP. New advances on prostate carcinogenesis and therapies: involvement of EGF-EGFR transduction system. Growth Factors 2003; 21:1-14. [PMID: 12795332 DOI: 10.1080/0897719031000094921] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The prostate cancers (PCs) are among the major causes of death because therapeutic treatments are not effective against advanced and metastatic forms of this cellular hyperproliferative disorder. In fact, although androgen-deprivation therapies permit to cure localized PC forms, the metastatic PC cells have acquired multiple functional features that confer to them resistance to ionizing radiations and anticarcinogenic drugs currently used in therapy. The present review describes last advances on molecular mechanisms that might be responsible for sustained growth and survival of PC cells. In particular, emphasis is on intracellular signaling cascades which are involved in the mitogenic and antiapoptotic effects of epidermal growth factor EGF-EGFR system. Of therapeutic interest, recent advances and prospects for development of new treatments against incurable forms of metastatic PC forms are also discussed.
Collapse
Affiliation(s)
- Murielle Mimeault
- Institut de Chimie Pharmaceutique Albert Lespagnol, Faculté de Pharmacie, 3 Rue du Professeur Laguesse, BP83, 59006 Lille, Cédex, France.
| | | | | |
Collapse
|
49
|
Tímár J, Ladányi A, Peták I, Jeney A, Kopper L. Molecular pathology of tumor metastasis III. Target array and combinatorial therapies. Pathol Oncol Res 2003; 9:49-72. [PMID: 12704448 DOI: 10.1007/bf03033715] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2003] [Accepted: 03/22/2003] [Indexed: 12/23/2022]
Abstract
Therapy of tumor progression and the metastatic disease is the biggest challenge of clinical oncology. Discovery of the diverse molecular pathways behind this complex disease outlined an approach to better treatment strategies. The development of combined cytotoxic treatment protocols has produced promising results but no breakthrough in the clinical management of metastatic disease. The multiple - specific and non-specific pathways and cellular targets of tumor progression are outlined in this review. Such an approach, individually designed for various cancer types, may have a better chance to treat or even cure cancer patients with progressive disease.
Collapse
Affiliation(s)
- József Tímár
- National Institute of Oncology, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
50
|
Abstract
There is a need for new, selective anticancer agents that differentiate between malignant and nonmalignant cells. The benefits of such agents would include a higher therapeutic index and lower toxicity than conventional therapies. Although expressed in nonmalignant cells, the epidermal growth factor receptor (EGFR) is highly expressed in a variety of tumors, and its expression correlates with poor response to treatment, disease progression, and poor survival. Evidence for a role for the EGFR in the inhibition and pathogenesis of various cancers has led to the rational design and development of agents that selectively target this receptor. Activation of the EGFR signaling pathway in cancer cells has been linked with increased cell proliferation, angiogenesis, and metastasis, and decreased apoptosis. Preclinical data show that anti-EGFR therapies can inhibit these effects in vitro and in vivo. In addition, preclinical data confirm that many such agents have the potential to increase the effectiveness of current cytotoxic agents. Following accelerated drug development programs, phase III trials are now under way for a number of EGFR-targeted therapies, including the monoclonal antibody IMC-C225 and the EGFR-tyrosine kinase inhibitors ZD1839 (Iressa) and OSI-774. Thus, the rationale for EGFR-targeted approaches to cancer treatment is apparent and now well established, and there is increasing evidence that they may represent a significant contribution to cancer therapy.
Collapse
Affiliation(s)
- José Baselga
- Medical Oncology Service, Hospital General Universitari Vall d'Hebron, Barcelona, Spain.
| |
Collapse
|