51
|
Heymach JV, Johnson BE, Prager D, Csada E, Roubec J, Pesek M, Spásová I, Belani CP, Bodrogi I, Gadgeel S, Kennedy SJ, Hou J, Herbst RS. Randomized, Placebo-Controlled Phase II Study of Vandetanib Plus Docetaxel in Previously Treated Non–Small-Cell Lung Cancer. J Clin Oncol 2007; 25:4270-7. [PMID: 17878479 DOI: 10.1200/jco.2006.10.5122] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Vandetanib is a once-daily oral inhibitor of vascular endothelial growth factor receptor-2 and epidermal growth factor receptor kinase activity. The activity of vandetanib plus docetaxel was assessed in patients with previously treated non–small-cell lung cancer (NSCLC). Patients and Methods This two-part study comprised an open-label run-in phase and a double-blind randomized phase. Eligible patients had locally advanced or metastatic (stage IIIB/IV) NSCLC after failure of first-line platinum-based chemotherapy. The primary objective of the randomized phase was to prolong progression-free survival (PFS) in patients receiving vandetanib (100 or 300 mg/d) plus docetaxel (75 mg/m2 intravenous infusion every 21 days) versus placebo plus docetaxel. The study was designed to have more than 75% power to detect 50% prolongation at a one-sided significance level of P < .20. Secondary objectives included objective response rate, overall survival, safety and tolerability. Results In the randomized phase (n = 127), median PFS was 18.7 weeks for vandetanib 100 mg plus docetaxel (n = 42; hazard ratio v docetaxel = 0.64; one-sided P = .037); 17.0 weeks for vandetanib 300 mg plus docetaxel (n = 44; hazard ratio v docetaxel = 0.83; one-sided P = .231); and 12 weeks for docetaxel (n = 41). There was no statistically significant difference in overall survival among the three treatment arms. Common adverse events included diarrhea, rash, and asymptomatic prolongation of corrected QT (QTC) interval. Conclusion The primary objective was achieved, with vandetanib 100 mg plus docetaxel demonstrating a significant prolongation of PFS compared with docetaxel in relation to the prespecified significance level. On the basis of these encouraging data, phase III evaluation of vandetanib 100 mg plus docetaxel in second-line NSCLC has been initiated.
Collapse
|
52
|
Abdollahi A, Schwager C, Kleeff J, Esposito I, Domhan S, Peschke P, Hauser K, Hahnfeldt P, Hlatky L, Debus J, Peters JM, Friess H, Folkman J, Huber PE. Transcriptional network governing the angiogenic switch in human pancreatic cancer. Proc Natl Acad Sci U S A 2007; 104:12890-5. [PMID: 17652168 PMCID: PMC1931565 DOI: 10.1073/pnas.0705505104] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A shift of the angiogenic balance to the proangiogenic state, termed the "angiogenic switch," is a hallmark of cancer progression. Here we devise a strategy for identifying genetic participants of the angiogenic switch based on inverse regulation of genes in human endothelial cells in response to key endogenous pro- and antiangiogenic proteins. This approach reveals a global network pattern for vascular homeostasis connecting known angiogenesis-related genes with previously unknown signaling components. We also demonstrate that the angiogenic switch is governed by simultaneous regulations of multiple genes organized as transcriptional circuitries. In pancreatic cancer patients, we validate the transcriptome-derived switch of the identified "angiogenic network:" The angiogenic state in chronic pancreatitis specimens is intermediate between the normal (angiogenesis off) and neoplastic (angiogenesis on) condition, suggesting that aberrant proangiogenic environment contributes to the increased cancer risk in patients with chronic pancreatitis. In knockout experiments in mice, we show that the targeted removal of a hub node (peroxisome proliferative-activated receptor delta) of the angiogenic network markedly impairs angiogenesis and tumor growth. Further, in tumor patients, we show that peroxisome proliferative-activated receptor delta expression levels are correlated with advanced pathological tumor stage, increased risk for tumor recurrence, and distant metastasis. Our results therefore also may contribute to the rational design of antiangiogenic cancer agents; whereas "narrow" targeted cancer drugs may fail to shift the robust angiogenic regulatory network toward antiangiogenesis, the network may be more vulnerable to multiple or broad-spectrum inhibitors or to the targeted removal of the identified angiogenic "hub" nodes.
Collapse
Affiliation(s)
- Amir Abdollahi
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
- To whom correspondence may be addressed. E-mail: , , or
| | - Christian Schwager
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
| | - Jörg Kleeff
- Departments of General Surgery, University of Heidelberg Medical School and European Pancreas Center, Heidelberg 69120, Germany
| | - Irene Esposito
- Institute of Pathology, University of Heidelberg Medical School, Heidelberg 69120, Germany
| | - Sophie Domhan
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
| | - Peter Peschke
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
| | - Kai Hauser
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
- Department of Mathematics, University of California, Berkeley, CA 94720-3840
| | - Philip Hahnfeldt
- Center of Cancer Systems Biology, Department of Medicine, Caritas St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135-2997
- **Children's Hospital Boston, Vascular Biology Program and Harvard Medical School, Department of Surgery, Karp Family Research Laboratories, Boston, MA 02115
| | - Lynn Hlatky
- Center of Cancer Systems Biology, Department of Medicine, Caritas St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135-2997
| | - Jürgen Debus
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
| | - Jeffrey M. Peters
- Department of Veterinary Science and Center for Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, PA 16802; and
| | - Helmut Friess
- Departments of General Surgery, University of Heidelberg Medical School and European Pancreas Center, Heidelberg 69120, Germany
| | - Judah Folkman
- **Children's Hospital Boston, Vascular Biology Program and Harvard Medical School, Department of Surgery, Karp Family Research Laboratories, Boston, MA 02115
- To whom correspondence may be addressed. E-mail: , , or
| | - Peter E. Huber
- *Department of Radiation Oncology, German Cancer Research Center (DKFZ) and University of Heidelberg Medical School, Heidelberg 69120, Germany
- To whom correspondence may be addressed. E-mail: , , or
| |
Collapse
|
53
|
Damiano V, Caputo R, Garofalo S, Bianco R, Rosa R, Merola G, Gelardi T, Racioppi L, Fontanini G, De Placido S, Kandimalla ER, Agrawal S, Ciardiello F, Tortora G. TLR9 agonist acts by different mechanisms synergizing with bevacizumab in sensitive and cetuximab-resistant colon cancer xenografts. Proc Natl Acad Sci U S A 2007; 104:12468-73. [PMID: 17636117 PMCID: PMC1920540 DOI: 10.1073/pnas.0705226104] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synthetic agonists of Toll-like receptor 9 (TLR9), a class of agents that induce specific immune response, exhibit antitumor activity and are currently being investigated in cancer patients. Intriguingly, their mechanisms of action on tumor growth and angiogenesis are still incompletely understood. We recently discovered that a synthetic agonist of TLR9, immune modulatory oligonucleotide (IMO), acts by impairing epidermal growth factor receptor (EGFR) signaling and potently synergizes with anti-EGFR antibody cetuximab in GEO human colon cancer xenografts, whereas it is ineffective in VEGF-overexpressing cetuximab-resistant GEO cetuximab-resistant (GEO-CR) tumors. VEGF is activated by EGFR, and its overexpression causes resistance to EGFR inhibitors. Therefore, we used IMO and the anti-VEGF antibody bevacizumab as tools to study IMO's role on EGFR and angiogenesis and to explore its therapeutic potential in GEO, LS174T, and GEO-CR cancer xenografts. We found that IMO enhances the antibody-dependent cell-mediated cytotoxicity (ADCC) activity of cetuximab, that bevacizumab has no ADCC, and IMO is unable to enhance it. Nevertheless, the IMO-plus-bevacizumab combination synergistically inhibits the growth of GEO and LS174T as well as of GEO-CR tumors, preceded by inhibition of signaling protein expression, microvessel formation, and human, but not murine, VEGF secretion. Moreover, IMO inhibited the growth, adhesion, migration, and capillary formation of VEGF-stimulated endothelial cells. The antitumor activity was irrespective of the TLR9 expression on tumor cells. These studies demonstrate that synthetic agonists of TLR9 interfere with growth and angiogenesis also by EGFR- and ADCC-independent mechanisms affecting endothelial cell functions and provide a strong rationale to combine IMO with bevacizumab and EGFR inhibitory drugs in colon cancer patients.
Collapse
Affiliation(s)
- Vincenzo Damiano
- Departments of *Endocrinologia e Oncologia Molecolare e Clinica and
| | - Rosa Caputo
- Departments of *Endocrinologia e Oncologia Molecolare e Clinica and
| | - Sonia Garofalo
- Departments of *Endocrinologia e Oncologia Molecolare e Clinica and
| | | | - Roberta Rosa
- Departments of *Endocrinologia e Oncologia Molecolare e Clinica and
| | - Gerardina Merola
- Biologia e Patologia Cellulare e Molecolare, Università di Napoli Federico II, 80131 Napoli, Italy
| | - Teresa Gelardi
- Departments of *Endocrinologia e Oncologia Molecolare e Clinica and
| | - Luigi Racioppi
- Biologia e Patologia Cellulare e Molecolare, Università di Napoli Federico II, 80131 Napoli, Italy
| | | | | | | | | | - Fortunato Ciardiello
- Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale, Seconda Università di Napoli, 80131 Napoli, Italy
- Oncotech, 80131 Napoli, Italy; and
| | - Giampaolo Tortora
- Oncotech, 80131 Napoli, Italy; and
- **To whom correspondence should be addressed. E-mail:
| |
Collapse
|
54
|
Abstract
The long-term prognosis for patients with advanced non-small cell lung cancer (NSCLC) remains poor despite the availability of several cytotoxic chemotherapy regimens. The use of targeted therapies, particularly those against the key mediator of angiogenesis vascular endothelial growth factor (VEGF), has the potential to improve outcomes for NSCLC patients. Bevacizumab, a recombinant humanized monoclonal anti-VEGF antibody, is the most clinically advanced antiangiogenic agent in NSCLC. In a phase III study, bevacizumab showed significantly improved overall and progression-free survival when used in combination with standard first-line chemotherapy in patients with advanced NSCLC. Bevacizumab was generally well tolerated in patients with NSCLC; however, tumor-related bleeding adverse events have been noted in some patients, predominantly those with squamous cell histology or centrally located tumors. Several small-molecule VEGF receptor tyrosine kinase inhibitors have also shown promise in phase I and II trials in NSCLC. This review summarizes the most important findings of angiogenesis inhibitors in NSCLC and discusses the potential for the use of these novel agents in different settings of NSCLC.
Collapse
Affiliation(s)
- Giuseppe Giaccone
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
55
|
|
56
|
Tabernero J. The role of VEGF and EGFR inhibition: implications for combining anti-VEGF and anti-EGFR agents. Mol Cancer Res 2007; 5:203-20. [PMID: 17374728 DOI: 10.1158/1541-7786.mcr-06-0404] [Citation(s) in RCA: 340] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple cellular pathways influence the growth and metastatic potential of tumors. This creates heterogeneity, redundancy, and the potential for tumors to bypass signaling pathway blockade, resulting in primary or acquired resistance. Combining therapies that inhibit different signaling pathways has the potential to be more effective than inhibition of a single pathway and to overcome tumor resistance. Vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) inhibitors have become key therapies in several tumor types. Close relationships between these factors exist: VEGF signaling is up-regulated by EGFR expression and, conversely, VEGF up-regulation independent of EGFR signaling seems to contribute to resistance to EGFR inhibition. Therefore, inhibition of both pathways could improve antitumor efficacy and overcome resistance to EGFR inhibition. Preclinical studies have shown that VEGF and EGFR inhibitors can have additive effects and that combined inhibition is effective in EGFR inhibitor-resistant cell lines. Clinical trials have also produced promising data: combining the anti-VEGF monoclonal antibody bevacizumab with the anti-EGFR antibody cetuximab or the EGFR tyrosine kinase inhibitor erlotinib increases benefit compared with either of these anti-EGFR agents alone or combined with chemotherapy. The potential of this novel approach to anticancer therapy will be elucidated by large, ongoing clinical trials.
Collapse
Affiliation(s)
- Josep Tabernero
- Medical Oncology Service, Vall d'Hebron University Hospital, P. Vall d'Hebron, 119-129, 08035 Barcelona, Spain.
| |
Collapse
|
57
|
Jimeno A, Kulesza P, Wheelhouse J, Chan A, Zhang X, Kincaid E, Chen R, Clark DP, Forastiere A, Hidalgo M. Dual EGFR and mTOR targeting in squamous cell carcinoma models, and development of early markers of efficacy. Br J Cancer 2007; 96:952-9. [PMID: 17342092 PMCID: PMC2360107 DOI: 10.1038/sj.bjc.6603656] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a validated target in squamous cell carcinoma (SCC) of the head and neck. Most patients, however, do not respond or develop resistance to this agent. Mammalian target of rapamycin (mTOR) is involved in the pathogenesis of SCC of the head and neck (SCCHN). This study aimed to determine if targeting mTOR in combination with EGFR is effective in SCC, and to develop early pharmacodynamic markers of efficacy. Two SCC cell lines, one resistant (HEP2) and one of intermediate susceptibility (Detroit 562) to EGFR inhibitors, were xenografted in vivo and treated with an mTOR inhibitor (temsirolimus), an EGFR inhibitor (erlotinib) or a combination of both. Temsirolimus exerted superior growth arrest in both cell lines than erlotinib. The combined treatment resulted in synergistic antitumor effects in the Detroit 562 cell line. Immunohistochemical assessment of pharmacodynamic effects in fine-needle aspiration (FNA) biopsies early after treatment using phospho MAPK, Phospho-P70 and Ki67 as end points demonstrated pathway abrogation in the Detroit 562 tumours treated with the combination, the only group where regressions were seen. In conclusion, an mTOR inhibitor showed antitumor activity in EGFR-resistant SCC cell lines. Marked antitumor effects were associated with dual pathway inhibition, which were detected by early FNA biopsies.
Collapse
Affiliation(s)
- A Jimeno
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - P Kulesza
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - J Wheelhouse
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - A Chan
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - X Zhang
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - E Kincaid
- Department of Pathology, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - R Chen
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - D P Clark
- Department of Pathology, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - A Forastiere
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
| | - M Hidalgo
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 1M89, Baltimore, MD 21231-1000, USA
- E-mail:
| |
Collapse
|
58
|
Pasqualetti G, Danesi R, Del Tacca M, Bocci G. Vascular endothelial growth factor pharmacogenetics: a new perspective for anti-angiogenic therapy. Pharmacogenomics 2007; 8:49-66. [PMID: 17187509 DOI: 10.2217/14622416.8.1.49] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The pharmacogenetic approach to anti-angiogenic therapy should be considered a possible strategy for many pathological conditions with high incidence in Western countries, including solid tumors, age-related macular degeneration or endometriosis. While pharmacogenetic studies are building stronger foundations for the systematic investigations of phenotype–genotype relationships in many research and clinical fields of medicine, pharmacogenetic data regarding anti-angiogenic drugs are still lacking. Here we review preclinical and clinical genetic studies on angiogenic determinants such as vascular endothelial growth factor and vascular endothelial growth factor receptor-2. We suggest that pharmacogenetic profiling of patients who are candidates for the currently available anti-angiogenic agents targeting vascular endothelial growth factor and vascular endothelial growth factor receptor-2 may aid the selection of patients on the basis of their likelihood of responding to the drugs or suffering from toxicity.
Collapse
Affiliation(s)
- Giuseppe Pasqualetti
- University of Pisa, Division of Pharmacology and Chemotherapy, Department of Internal Medicine, Via Roma, 55, I-56126 Pisa, Italy
| | | | | | | |
Collapse
|
59
|
|
60
|
Learn PA, Krishnegowda N, Talamantez J, Kahlenberg MS. Compensatory increases in Her-2/neu activation in response to EGFR tyrosine kinase inhibition in colon cancer cell lines. J Surg Res 2006; 136:227-31. [PMID: 17059835 DOI: 10.1016/j.jss.2006.07.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 06/09/2006] [Accepted: 07/31/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND Tyrosine kinase receptors of the ErbB family have become promising targets for anti-neoplastic drugs, but mechanisms of resistance are incompletely understood. To investigate such pathways, we applied a small-molecule, selective EGFR inhibitor, OSI-774, to three well-characterized colon cancer cell lines and studied the alterations of expression and activation of receptors in the erbB family. METHODS MTT assays were performed to determine the IC(50)s of GEO, FET, and HCT 116 human colorectal cancer cell lines treated with OSI-774. Plated cells were then exposed to either DMSO control or 7 microm of OSI-774 for treatment durations of 1, 3, 5, 7, 10, 14, 28, and 56 days. Cell lysates were evaluated by Western blotting, evaluating both total and phosphorylated levels of EGFR, Her-2/neu, and erbB-3. RESULTS IC(50) values for GEO, FET, and HCT 116 cell lines exposed to OSI-774 were 12.0, 16.0, and greater than 100 microm, respectively. In all treated cell lines, OSI-774 diminished EGFR activation but did not affect total expression compared with controls. In contrast, Her-2/neu activation was increased in all cell lines. These changes in EGFR and Her-2/neu were identified within 24 h but peaked later in the treatment cycle. ErbB-3 expression and activation did not follow a consistent pattern between cell lines. CONCLUSIONS Inhibition of EGFR led to increased activation of Her-2/neu. This result suggests a possible mechanism by which cells might escape the proapoptotic signals resulting from EGFR blockade. Our findings suggest concurrent inhibition of multiple members of the erbB family may yield stronger apoptotic responses than single receptor blockade alone.
Collapse
Affiliation(s)
- Peter A Learn
- Department of Surgery, Wilford Hall Medical Center, San Antonio, Texas, USA
| | | | | | | |
Collapse
|
61
|
Fischer C, Schneider M, Carmeliet P. Principles and therapeutic implications of angiogenesis, vasculogenesis and arteriogenesis. Handb Exp Pharmacol 2006:157-212. [PMID: 16999228 DOI: 10.1007/3-540-36028-x_6] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The vasculature is the first organ to arise during development. Blood vessels run through virtually every organ in the body (except the avascular cornea and the cartilage), assuring metabolic homeostasis by supplying oxygen and nutrients and removing waste products. Not surprisingly therefore, vessels are critical for organ growth in the embryo and for repair of wounded tissue in the adult. Notably, however, an imbalance in angiogenesis (the growth of blood vessels) contributes to the pathogenesis of numerous malignant, inflammatory, ischaemic, infectious and immune disorders. During the last two decades, an explosive interest in angiogenesis research has generated the necessary insights to develop the first clinically approved anti-angiogenic agents for cancer and blindness. This novel treatment is likely to change the face of medicine in the next decade, as over 500 million people worldwide are estimated to benefit from pro- or anti-angiogenesis treatment. In this following chapter, we discuss general key angiogenic mechanisms in health and disease, and highlight recent developments and perspectives of anti-angiogenic therapeutic strategies.
Collapse
Affiliation(s)
- C Fischer
- Centre for Transgene Technology and Gene Therapy, Flanders Interuniversity Institute for Biotechnology, KULeuven, Campus Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | | | | |
Collapse
|
62
|
Jimeno A, Hidalgo M. Multitargeted therapy: Can promiscuity be praised in an era of political correctness? Crit Rev Oncol Hematol 2006; 59:150-8. [PMID: 16843676 DOI: 10.1016/j.critrevonc.2006.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Accepted: 01/17/2006] [Indexed: 01/02/2023] Open
Abstract
The rapidly expanding knowledge of the pathogenesis of cancer at the molecular level is providing new targets for drug discovery and development. However, cancer is a complex disease characterized by multiple genetic and molecular alterations affecting cell proliferation, survival, differentiation and invasion among others. Many of these alterations represent potential targets for the development of new anticancer therapeutics. Because of the enormous biological diversity of cancer, it is unlikely that attacking only one of these targets will eliminate a malignant cell. Rather, strategic combination of agents targeted against the most critical of those alterations will be needed. Another approach that is rendering promising clinical results is the use of more unspecific agents that inhibit or modulate several relevant targets simultaneously. A deep biologic understanding of the relative relevance of each target in different cancer types will be key to efficiently direct those drugs to diseases more likely to benefit from its particular modulation profile.
Collapse
Affiliation(s)
- Antonio Jimeno
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD 21231, USA
| | | |
Collapse
|
63
|
Srinivasan D, Plattner R. Activation of Abl tyrosine kinases promotes invasion of aggressive breast cancer cells. Cancer Res 2006; 66:5648-55. [PMID: 16740702 DOI: 10.1158/0008-5472.can-06-0734] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Abl family of nonreceptor tyrosine kinases consists of two related proteins, c-Abl and Abl-related gene (Arg). Activated forms of the Abl kinases (BCR-Abl, Tel-Abl, and Tel-Arg) induce the development of human leukemia; it is not known, however, whether Abl kinases are activated in solid tumors or whether they contribute to tumor development or progression. Previously, we showed that Abl kinases are activated downstream of growth factor receptors, Src family kinases, and phospholipase Cgamma1 (PLCgamma1) in fibroblasts and influence growth factor-mediated proliferation, membrane ruffling, and migration. Growth factor receptors, Src kinases, and PLCgamma1 are deregulated in many solid tumors and drive tumor invasion and metastasis. In this study, we found that Abl kinases are constitutively activated, in highly invasive breast cancer cell lines, downstream of deregulated ErbB receptors and Src kinases. Furthermore, activation of Abl kinases promotes breast cancer cell invasion, as treatment of cells with the Abl kinase inhibitor, STI571, or silencing c-Abl and Arg expression with RNA interference dramatically inhibits Matrigel invasion. This is the first evidence that (a) Abl kinases are deregulated and activated in a nonhematopoietic cancer, (b) activation of Abl kinases in breast cancer cells occurs via a novel mechanism, and (c) constitutive activation of Abl kinases promotes invasion of breast cancer cells. These data suggest that pharmacologic inhibitors targeted against Abl kinases could potentially be useful in preventing breast cancer progression in tumors harboring activated Abl kinases.
Collapse
Affiliation(s)
- Divyamani Srinivasan
- Department of Molecular and Biomedical Pharmacology, University of Kentucky School of Medicine, Lexington, Kentucky, USA
| | | |
Collapse
|
64
|
Hwang LH. Gene therapy strategies for hepatocellular carcinoma. J Biomed Sci 2006; 13:453-68. [PMID: 16633742 DOI: 10.1007/s11373-006-9085-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 03/23/2006] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent cancers worldwide. Effective therapy to this cancer is currently lacking, creating an urgent need for new therapeutic strategies for HCC. Gene therapy approach that relies on the transduction of cells with genetic materials, such as apoptotic genes, suicide genes, genes coding for antiangiogenic factors or immunomodulatory molecules, small interfering RNA (siRNA), or oncolytic viral vectors, may provide a promising strategy. The aforementioned strategies have been largely evaluated in the animal models with HCC or liver metastasis. Due to the diversity of vectors and therapeutic genes, being used alone or in combination, gene therapy approach may generate great beneficial effects to control the growth of tumors within the liver.
Collapse
Affiliation(s)
- Lih-Hwa Hwang
- Hepatitis Research Center, National Taiwan University Hospital and Graduate Institute of Microbiology, National Taiwan University College of Medicine, 7 Chung-Shan S. Road, Taipei 10016, Taiwan, R.O.C.
| |
Collapse
|
65
|
Scartozzi M, Pierantoni C, Berardi R, Squadroni M, Cascinu S. Anti-EGFR strategies as an incremental step for the treatment of colorectal cancer patients: moving from scientific evidence to clinical practice. Expert Opin Ther Targets 2006; 10:281-287. [PMID: 16548776 DOI: 10.1517/14728222.10.2.281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The epidermal growth factor receptor (EGFR) is a 170,000 Da transmembrane glycoprotein involved in signalling pathways affecting cellular growth, differentiation and proliferation. An abnormal overexpression of the EGFR has been described in many human tumours and implicated in the development and prognosis of malignancies, thus representing not only a possible prognostic marker, but primarily a rational molecular target for a new class of anticancer agents. Several clinical trials have been reported with the use of EGFR-targeted monoclonal antibodies and tyrosine kinase inhibitors, mainly in combination with chemotherapy for advanced colorectal cancer patients. Taken together, results available so far suggest that anti-EGFR treatment strategies represent an incremental step for the the treatment of colorectal cencer patients with a manageable and acceptable toxicity profile. Nevertheless, many critical issues are yet unresolved, such as the optimal chemotherapy regimen to combine with anti-EGFR treatment and the most adequate patients setting. Moreover, the biological selection of colorectal tumours most likely to benefit from this treatment approach is still to be defined.
Collapse
Affiliation(s)
- Mario Scartozzi
- Azienda Ospedaliera Ospedali Riuniti-Universita, Clinica di Oncologia Medica, Politecnica delle Marche, via Conca 60020, Ancona, Italy
| | | | | | | | | |
Collapse
|
66
|
du Manoir JM, Francia G, Man S, Mossoba M, Medin JA, Viloria-Petit A, Hicklin DJ, Emmenegger U, Kerbel RS. Strategies for Delaying or Treating In vivo Acquired Resistance to Trastuzumab in Human Breast Cancer Xenografts. Clin Cancer Res 2006; 12:904-16. [PMID: 16467105 DOI: 10.1158/1078-0432.ccr-05-1109] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Acquired resistance to trastuzumab (Herceptin) is common in patients whose breast cancers show an initial response to the drug. The basis of this acquired resistance is unknown, hampering strategies to delay or treat such acquired resistance, due in part to the relative lack of appropriate in vivo tumorigenic models. EXPERIMENTAL DESIGN We derived an erbB-2-positive variant called 231-H2N, obtained by gene transfection from the highly tumorigenic erbB-2/HER2-negative human breast cancer cell line, MDA-MB-231. Unlike MDA-MB-231, the 231-H2N variants was sensitive to trastuzumab both in vitro and especially in vivo, thus allowing selection of variant resistant to drug treatment in the latter situation after showing an initial response. RESULTS The growth of established orthotopic tumors in severe combined immunodeficient mice was blocked for 1 month by trastuzumab, after which rapid growth resumed. These relapsing tumors were found to maintain resistance to trastuzumab, both in vitro and in vivo. We evaluated various therapeutic strategies for two purposes: (a) to delay such tumor relapses or (b) to treat acquired trastuzumab resistance once it has occurred. With respect to the former, a daily oral low-dose metronomic cyclophosphamide regimen was found to be particularly effective. With respect to the latter, an anti-epidermal growth factor receptor antibody (cetuximab) was effective as was the anti-vascular endothelial growth factor (anti-VEGF) antibody bevacizumab, which was likely related to elevated levels of VEGF detected in trastuzumab-resistant tumors. CONCLUSIONS Our results provide a possible additional rationale for combined biological therapy using drugs that target both erbB-2/HER2 and VEGF and also suggest the potential value of combining less toxic metronomic chemotherapy regimens not only with targeted antiangiogenic agents but also with other types of drug such as trastuzumab.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Breast Neoplasms/drug therapy
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cyclophosphamide/administration & dosage
- Cyclophosphamide/therapeutic use
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Drug Resistance, Neoplasm/immunology
- Female
- Gene Expression Regulation
- Humans
- In Vitro Techniques
- Mice
- RNA, Messenger/genetics
- Receptor, ErbB-2/analysis
- Sensitivity and Specificity
- Time Factors
- Trastuzumab
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jeanne M du Manoir
- Molecular and Cellular Biology Research, Sunnybrook and Women's College Health Sciences Centre, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Reardon DA, Quinn JA, Vredenburgh JJ, Gururangan S, Friedman AH, Desjardins A, Sathornsumetee S, Herndon JE, Dowell JM, McLendon RE, Provenzale JM, Sampson JH, Smith RP, Swaisland AJ, Ochs JS, Lyons P, Tourt-Uhlig S, Bigner DD, Friedman HS, Rich JN. Phase 1 Trial of Gefitinib Plus Sirolimus in Adults with Recurrent Malignant Glioma. Clin Cancer Res 2006; 12:860-8. [PMID: 16467100 DOI: 10.1158/1078-0432.ccr-05-2215] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of gefitinib, a receptor tyrosine kinase inhibitor of the epidermal growth factor receptor, plus sirolimus, an inhibitor of the mammalian target of rapamycin, among patients with recurrent malignant glioma. PATIENTS AND METHODS Gefitinib and sirolimus were administered on a continuous daily dosing schedule at dose levels that were escalated in successive cohorts of malignant glioma patients at any recurrence who were stratified based on concurrent use of CYP3A-inducing anticonvulsants [enzyme-inducing antiepileptic drugs, (EIAED)]. Pharmacokinetic and archival tumor biomarker data were also assessed. RESULTS Thirty-four patients with progressive disease after prior radiation therapy and chemotherapy were enrolled, including 29 (85%) with glioblastoma multiforme and 5 (15%) with anaplastic glioma. The MTD was 500 mg of gefitinib plus 5 mg of sirolimus for patients not on EIAEDs and 1,000 mg of gefitinib plus 10 mg of sirolimus for patients on EIAEDs. DLTs included mucositis, diarrhea, rash, thrombocytopenia, and hypertriglyceridemia. Gefitinib exposure was not affected by sirolimus administration but was significantly lowered by concurrent EIAED use. Two patients (6%) achieved a partial radiographic response, and 13 patients (38%) achieved stable disease. CONCLUSION We show that gefitinib plus sirolimus can be safely coadministered on a continuous, daily dosing schedule, and established the recommended dose level of these agents in combination for future phase 2 clinical trials.
Collapse
|
68
|
Macarulla T, Valverde C, Ramos FJ, Casado E, Martinelli E, Tabernero J, Cervantes A. Emerging strategies in the treatment of advanced esophageal, gastroesophageal junction, and gastric cancer: the introduction of targeted therapies. Target Oncol 2006. [DOI: 10.1007/s11523-005-0002-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
69
|
van Cruijsen H, Giaccone G, Hoekman K. Epidermal growth factor receptor and angiogenesis: Opportunities for combined anticancer strategies. Int J Cancer 2005; 117:883-8. [PMID: 16152621 DOI: 10.1002/ijc.21479] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor-induced angiogenesis is essential for malignant growth. This mini review focuses on the role of vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF) and their receptors in this process, and the rationale to combine inhibitors of these growth factors as anticancer therapy. Concomitantly, targeting the VEGF(R) and the EGF(R) signaling pathway may circumvent the problem of acquired resistance to EGFR inhibitors. By targeting both pathways, the antiangiogenic effect may be more pronounced, which may lead to greater antitumor activity. Preliminary efficacy data from clinical trials encourage further exploration of this combined anticancer strategy.
Collapse
Affiliation(s)
- Hester van Cruijsen
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
70
|
Milkiewicz M, Ispanovic E, Doyle JL, Haas TL. Regulators of angiogenesis and strategies for their therapeutic manipulation. Int J Biochem Cell Biol 2005; 38:333-57. [PMID: 16309946 DOI: 10.1016/j.biocel.2005.10.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 10/05/2005] [Accepted: 10/11/2005] [Indexed: 12/19/2022]
Abstract
Angiogenesis provides a mechanism by which delivery of oxygen and nutrients is adapted to compliment changes in tissue mass or metabolic activity. However, maladaptive angiogenesis is integral to the process of several diseases common in Western countries, including tumor growth, vascular insufficiency, diabetic retinopathy and rheumatoid arthritis. Understanding the process of capillary growth, including the identification and functional analyses of key pro- and anti-angiogenic factors, provides knowledge that can be applied to improve/reverse these pathological states. Initially, angiogenesis research focused predominantly on vascular endothelial growth factor (VEGF) as a main player in the angiogenesis cascade. It is apparent now that participation of multiple angiogenic factors and signal pathways is critical to enable effective growth and maturation of nascent capillaries. The purpose of this review is to focus on recent progress in identifying angiogenesis signaling pathways that show promise as targets for successful induction or inhibition of capillary growth. The strategies applied to achieve these contradictory tasks are discussed within the framework of our existing fundamental knowledge of angiogenesis signaling cascades, with an emphasis on comparing the employment of distinctive tactics in modulation of these pathways. Innovative developments that are presented include: (1) inducing a pleiotropic response via activation or inhibition of angiogenic transcription factors; (2) modulation of nitric oxide tissue concentration; (3) manipulating the kallikrein-kinin system; (4) use of endothelial progenitor cells as a means to either directly contribute to capillary growth or to be used as a vehicle to deliver "suicide genes" to tumor tissue.
Collapse
Affiliation(s)
- Malgorzata Milkiewicz
- School of Kinesiology and Health Sciences, York University, Toronto, Ont. M3J 1P3, Canada
| | | | | | | |
Collapse
|
71
|
Jimeno A, Hidalgo M. Blockade of epidermal growth factor receptor (EGFR) activity. Crit Rev Oncol Hematol 2005; 53:179-92. [PMID: 15718144 DOI: 10.1016/j.critrevonc.2004.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2004] [Indexed: 01/02/2023] Open
Abstract
The rapidly expanding knowledge of the pathogenesis of cancer at the molecular level is providing new targets for drug discovery and development. The key role that EGFR plays in the intracellular transduction of environmental variations and the maintenance of cellular homeostasis explains the dependence that many tumor types have on this pathway, and the pivotal role that it plays in the development of malignant features such as uncontrolled proliferation, augmented invasion, and the ability to escape apoptosis. An enormous body of knowledge has been gathered in the past 20 years that has enabled the development of rationally designed EGFR-targeted therapies, and the results of their clinical evaluation are now becoming available. The lack of positive results of some of these trials has highlighted the need for a robust preclinical knowledge in order to efficiently select patients for therapy, and have prompted the implementation of novel trial designs with rational endpoints.
Collapse
Affiliation(s)
- Antonio Jimeno
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Bunting-Blaustein Cancer Research Building, Room 1M88, 1650 Orleans Street, Baltimore, MD 21231-1000, USA
| | | |
Collapse
|
72
|
Förster Y, Meye A, Albrecht S, Schwenzer B. Tissue factor and tumor: clinical and laboratory aspects. Clin Chim Acta 2005; 364:12-21. [PMID: 16139825 DOI: 10.1016/j.cca.2005.05.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 05/13/2005] [Accepted: 05/16/2005] [Indexed: 11/22/2022]
Abstract
This review summarizes data demonstrating the role of TF in tumor development, metastasis and angiogenesis. TF is a transmembrane protein that is expressed constitutively in some kinds of extravascular cells and transiently in intravascular cells after stimulation with cytokines and growth factors. Originally TF was considered to have a function in the initiation of coagulation. In the last years it became evident that TF plays a role in physiological and pathological processes outside the hemostasis. Up-regulation of TF expression appears to be characteristic of tumor tissue. In a variety of human tumors it was shown by immunohistochemistry, that TF can be expressed in malignant cells as well as in tumor-infiltrating macrophages or endothelial cells. Such abnormal TF expression contributes to the angiogenic process by a shift in the balance between endogenous proangiogenic and antiangiogenic factors. Observations of a significant correlation between elevated TF expression with increased microvessel density and VEGF expression underline the TF involvement in tumor angiogenesis. Furthermore, TF expression influences also metastasis. The effect of TF on metastasis may result from its angiogenic effect, but also from the production of growth factors or adhesion proteins.
Collapse
Affiliation(s)
- Yvonne Förster
- Institute of Biochemistry, Technical University Dresden, Bergstrasse 66 D-01069 Dresden, Germany
| | | | | | | |
Collapse
|
73
|
Chou TY, Chiu CH, Li LH, Hsiao CY, Tzen CY, Chang KT, Chen YM, Perng RP, Tsai SF, Tsai CM. Mutation in the tyrosine kinase domain of epidermal growth factor receptor is a predictive and prognostic factor for gefitinib treatment in patients with non-small cell lung cancer. Clin Cancer Res 2005; 11:3750-7. [PMID: 15897572 DOI: 10.1158/1078-0432.ccr-04-1981] [Citation(s) in RCA: 258] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE Mutations in epidermal growth factor receptor (EGFR) can be used to predict the tumor response of patients receiving gefitinib for non-small cell lung cancer (NSCLC). We investigated the association between mutations in EGFR tyrosine kinase domain and tumor response and survival in gefitinib-treated NSCLC patients. EXPERIMENTAL DESIGN EGFR mutations in exons 18 to 21 were analyzed by DNA sequencing of paraffin-embedded tumor tissues from gefitinib-treated NSCLC patients. The results were correlated with clinical variables. RESULTS EGFR mutations were found in 61.1% (33 of 54) of cases; response rate and disease control rate were 56.8% and 68.5%, respectively. There was no significant difference in mutation rates between adenocarcinoma (29 of 43) and nonadenocarcinoma (4 of 11; P = 0.085). However, all four nonadenocarcinomas with EGFR mutations had no response to gefitinib. Presence of EGFR mutations was the only independent predictor for disease control (P = 0.003) and tumor response (P = 0.017) in multivariate analysis; positive predictive values were 87.9% and 70.8% and negative predictive values were 61.9% and 69.2%, respectively. In comparison with patients whose tumor was negative for EGFR mutations, patients with EGFR mutations had better progression-free survival (median, 7.6 versus 1.7 months; P = 0.011) and overall survival (median, 14.7 versus 4.7 months; P = 0.046). CONCLUSIONS Mutations in EGFR tyrosine kinase correlate with treatment response and survival in gefitinib-treated NSCLC patients and can be used as a predictive and prognostic factor. Thus, analysis of EGFR tyrosine kinase mutations in lung adenocarcinoma is of clinical significance, as it can permit the customization of treatment with EGFR tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Teh-Ying Chou
- Department of Pathology and Chest Department, Taipei Veterans General Hospital, School of Medicine and Institute of Genetics and Genome Research Center, National Yang-Ming University, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Rogers SJ, Harrington KJ, Rhys-Evans P, O-Charoenrat P, Eccles SA. Biological significance of c-erbB family oncogenes in head and neck cancer. Cancer Metastasis Rev 2005; 24:47-69. [PMID: 15785872 DOI: 10.1007/s10555-005-5047-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) tends to run an aggressive course and the prognosis has remained virtually unchanged in recent decades. The development of novel therapeutic strategies to improve patient outcome centres on the biology of the disease, namely the pivotal c-erbB family of growth factor receptors. c-erbB1 (or epidermal growth factor receptor, EGFR), is key to the pathogenesis of SCCHN and plays a central role in a complex network of downstream integrated signalling pathways. EGFR overexpression, detected in up to 90% of SCCHN, correlates with an increased risk of locoregional tumour relapse following primary therapy and relative resistance to treatment. The biological sequelae of erbB receptor activation are not simply cell proliferation, but also inhibition of apoptosis, enhanced migration, invasion, angiogenesis and metastasis: the 'hallmarks of cancer' [1]. As EGFR overexpression is associated with a poor clinical outcome in SCCHN, this receptor is attractive as a therapeutic target and the successful development of targeted therapies represents a paradigm shift in the medical approach to head and neck cancer. However, the extensive cross talk between signalling pathways, the multiple molecular aberrations and genetic plasticity in SCCHN all contribute to inherent and acquired resistance to both conventional and novel therapies. Understanding the cancer cell biology, in particular the significance of co-expression of c-erbB (and other) receptors, and the cell survival stimuli from (for example) activation of the phosphoinositide 3-kinase (PI3-kinase) cascade is fundamental to overcome current limitations in biologically targeted therapies.
Collapse
Affiliation(s)
- Susanne J Rogers
- Tumour Biology and Metastasis Team, CRUK Centre for Cancer Therapeutics, McElwain Laboratories, Institute for Cancer Research, Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | | | | | | |
Collapse
|
75
|
Kumar P, Benedict R, Urzua F, Fischbach C, Mooney D, Polverini P. Combination treatment significantly enhances the efficacy of antitumor therapy by preferentially targeting angiogenesis. J Transl Med 2005; 85:756-67. [PMID: 15864318 DOI: 10.1038/labinvest.3700272] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Radiotherapy is one of the most widely used cancer treatments, but it is often unsuccessful due to the development of radioresistance by tumor cells and endothelial cells (ECs) lining the tumor blood vessels. We have previously shown that ECs are protected against ionizing irradiation primarily via the activation of the phosphoinositide 3-kinase (PI3 K)-Akt-Bcl-2 survival pathway. Here we report that combination treatment with low doses of PI3 K inhibitor (LY294002), cisplatin and gamma-irradiation resulted in significantly higher (61%) EC death as compared to each agent used alone (17, 17 and 11%, respectively). This combination treatment was equally effective in inducing tumor cell death (72%). Combination treatment also significantly inhibited EC tube formation in Matrigel (75%) as compared to each of the agents used alone (8, 8 and 18% for LY294002, cisplatin and gamma-irradiation, respectively). In our in vivo severe combined immunodeficient mouse model of human tumor growth and angiogenesis, combination treatment with low doses of LY294002, cisplatin and irradiation significantly inhibited the growth of human oral squamous carcinoma (OSCC-3) as well as prostate cancer (LnCap). The combination therapy was also very effective in inhibiting tumor angiogenesis where it showed a greater than 90% decrease in neovascularization. In contrast, combination treatment showed only a 29% inhibition of physiological angiogenesis. Taken together, these results suggest a potentially novel strategy to overcome the resistance in ECs lining tumor blood vessels, thereby enhancing the effectiveness of the radiation and chemotherapy. Moreover, this strategy of using a combination of low doses of PI3K/Akt inhibitor, cisplatin and radiation has the potential of significantly decreasing untoward side effects associated with the maximum tolerated doses of radiation and chemotherapy while maintaining their therapeutic efficacy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Apoptosis
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/radiotherapy
- Caspase 3
- Caspases/metabolism
- Cell Division
- Cell Line, Tumor
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/physiology
- Humans
- Mice
- Mice, SCID
- Mouth Neoplasms/blood supply
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/pathology
- Mouth Neoplasms/radiotherapy
- Neovascularization, Pathologic/pathology
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Pawan Kumar
- Department of Biological and Material Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
76
|
Graepler F, Verbeek B, Graeter T, Smirnow I, Kong HL, Schuppan D, Bauer M, Vonthein R, Gregor M, Lauer UM. Combined endostatin/sFlt-1 antiangiogenic gene therapy is highly effective in a rat model of HCC. Hepatology 2005; 41:879-86. [PMID: 15739185 DOI: 10.1002/hep.20613] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hepatocellular carcinoma (HCC) is regarded as a suitable target for antiangiogenic strategies. However, antiangiogenic agents aimed at single targets can be neutralized by upregulation of other proangiogenic factors. Therefore, combined approaches addressing at least two angiogenic targets should be more effective. Employing an appropriate rat hepatoma model, we examined the effects of sFlt-1 (soluble vascular endothelial growth factor [VEGF] receptor 1 as an indirect inhibitor of angiogenesis) and endostatin (a direct inhibitor of angiogenesis) in both single-agent as well as combined approaches under in vitro and in vivo conditions. Similar to human HCC, rat Morris hepatoma (MH) cells secreted high levels of VEGF, but no endogenous sFlt-1. Parental MH or MHES(r) cells, stably expressing rat endostatin, were adenovirally transduced either with AdsFlt-1 (encoding sFlt-1) or control vector Adnull (containing no transgene), followed by subcutaneous inoculation into syngeneic ACI rats. Compared with MH/Adnull cells, expressing no antiangiogenic factors at all, tumor weights were reduced fourfold in the MHES(r)/Adnull group, 19-fold in the MH/AdsFlt-1-group, and 77-fold in the MHES(r)/AdsFlt-1 combination therapy group. Analysis of variance did not show a significant interaction between the effects of the two factors ES(r) and sFlt-1; their effects multiplied. In conclusion, combined expression of sFlt-1 and endostatin effectively suppresses HCC growth under in vivo conditions. Supplementary material for this article can be found on the HEPATOLOGY website (http://interscience.wiley.com/jpages/0270-9139/suppmat/index.html).
Collapse
Affiliation(s)
- Florian Graepler
- Department of Internal Medicine, University Clinic Tübingen, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Abstract
Epithelial growth factor receptor (EGFR) appears as a key element in carcinogenesis. It displays high levels of expression in some tumor types. Its activation induces cell proliferation, angiogenesis, cell mobility and inhibition of apoptosis. EGFR inhibitors such as monoclonal antibodies or small molecules tyrosine kinase inhibitors have been developed. For EGFR, the diversity of the activation means (amplification, mutation, enhanced transcription, ligands...) leads to technical caveats. Immunohistochemistry appears to be the most appropriate test for clinical use, but standardized assays and scoring systems are mandatory. Pathologists are involved in the selection of patients for a monoclonal antibody based targeted treatment, Erbitux, and numerous standardization efforts are provided. No consensus has been reached, to date, for a scoring system. Whether the EGFR status has to be tested for the selection of patients is a non answered question. The selection of the "right patient for the right treatment" might be through the evaluation of other putative markers involved in resistance. EGFR testing may be required before targeted treatment. An exciting endpoint might be the functional and dynamic evaluation of EGFR and downstream proteins, for patients, before and during treatment. The exact role of the pathologist still have to be accurately determined.
Collapse
|
78
|
Abstract
Aberrant expression of growth factor receptor systems and dysregulation of the downstream cell signalling molecules have been reported in a wide range of epithelial tumours including head and neck cancer. In some cases, such alterations have been associated with a poor prognosis. In the past 25 years, several antigen specific monoclonal antibodies (mAbs, mouse, chimeric, humanized and human versions), and small molecule kinase inhibitors have been developed that are at different stages of preclinical and clinical developments. Some of these agents (e.g. Herceptin, Iressa, cetuximab, avastin) have already been approved for the treatment of epithelial tumours and may also have potential in the treatment of head and neck cancer patients. This review discusses, the development and potential of these antigen specific agents, in particular the human epidermal growth factor receptor (EGFR) inhibitors, either as a single agent or in combination with other EGFR inhibitors, biological agents (e.g. inhibitors of cycloogenase-2, angiogenesis, insulin like growth factor-I receptor and others), and conventional forms of therapy in the prevention and treatment of head and neck cancer. From preclinical and clinical studies with some of these compounds, it is evident that further detailed studies of biopsies from cancer patients are needed in order to identify markers that can be used not only in the selection of the specific population of cancer patients who would benefit from such antigen specific therapeutic strategies, but also those factors which are responsible for the poor response and the development of a phenotype resistance to such inhibitors. The results of such studies could in turn facilitate the widespread use of such agents in the treatment of a wide range of human cancers including head and neck cancer.
Collapse
Affiliation(s)
- Helmout Modjtahedi
- Division of Oncology, Postgraduate Medical School, University of Surrey, Guildford, Surrey, GU2 7XH, UK.
| |
Collapse
|
79
|
Scartozzi M, Bearzi I, Berardi R, Mandolesi A, Fabris G, Cascinu S. Epidermal growth factor receptor (EGFR) status in primary colorectal tumors does not correlate with EGFR expression in related metastatic sites: implications for treatment with EGFR-targeted monoclonal antibodies. J Clin Oncol 2004; 22:4772-4778. [PMID: 15570078 DOI: 10.1200/jco.2004.00.117] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PURPOSE We hypothesized that the detection of epidermal growth factor receptor (EGFR) expression performed in primary tumors for treatment with EGFR-targeted monoclonal antibodies could not always correlate with EGFR status in metastatic sites, thus making cancer cells in these sites resistant to therapy. The aim of our study was to correlate EGFR expression on primary tumors and related metastases in order to find out whether assessing EGFR status on primary cancer is to be considered an effective tool for planning treatment with EGFR-targeted antibodies. PATIENTS AND METHODS We retrospectively evaluated EGFR immunohistochemistry from primary tumors and related metastatic sites in 99 colorectal cancer patients. The site of primary tumor was colon in 77 patients (78%) and rectum in 22 patients (22%). Metastatic sites analyzed were liver in 84 patients (81%), lung in 13 patients (13%), bone in one patient (1%), and brain in five patients (5%). EGFR status was defined as positive if the percentage of malignant cells stained was > or = 1%. RESULTS EGFR status was positive in 53 primary tumors (53%). In 19 primary tumors expressing EGFR (36%), the corresponding metastatic site was found negative, whereas it was found positive in seven metastases (15%) from EGFR-negative primary cancers. The difference between these two groups of patients (ie, EGFR-positive to EGFR-negative v EGFR-negative to EGFR-positive) was statistically significant (P = .036). CONCLUSION Our results suggest that the detection of the EGFR in primary colorectal cancer could be inadequate for planning therapy with EGFR-targeted monoclonal antibodies in a considerable proportion of both EGFR-positive and -negative primary tumors (36% and 15%, respectively).
Collapse
Affiliation(s)
- Mario Scartozzi
- Clinica di Oncologia Medica, Istituto di Anatomia Patologica, Azienda Ospedaliera Ospedali Riuniti, Ancona, Italy
| | | | | | | | | | | |
Collapse
|