101
|
Horn L, Sandler A. Epidermal growth factor receptor inhibitors and antiangiogenic agents for the treatment of non-small cell lung cancer. Clin Cancer Res 2009; 15:5040-8. [PMID: 19671872 PMCID: PMC3528182 DOI: 10.1158/1078-0432.ccr-09-0520] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Non-small cell lung cancer (NSCLC) is a major global health problem and represents the leading cause of cancer-related deaths worldwide. The majority of patients with NSCLC are diagnosed with advanced-stage disease, and the prognosis for such patients is poor. The currently approved cytotoxic chemotherapy is associated with substantial limitations in both efficacy and safety. The availability of agents targeted against the epidermal growth factor receptor (EGFR), as well as the antiangiogenic agent bevacizumab, have provided some clinical benefit. Nonetheless, the efficacy of these agents is also inadequate, and resistance has emerged as a clinical problem. Numerous novel targeted therapies are now in clinical development and may have potential for overcoming the limitations associated with currently available agents. In this article we review clinical data for molecular-targeted therapies in NSCLC, with emphasis on EGFR inhibitors and antiangiogenic agents.
Collapse
Affiliation(s)
- Leora Horn
- Vanderbilt Ingram Cancer Centre, Division of Hematology/Oncology, Nashville, Tennessee, USA
| | | |
Collapse
|
102
|
Mahalingam D, Mita A, Mita MM, Nawrocki ST, Giles FJ. Targeted therapy for advanced non-small cell lung cancers: historical perspective, current practices, and future development. Curr Probl Cancer 2009; 33:73-111. [PMID: 19409299 DOI: 10.1016/j.currproblcancer.2009.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Devalingam Mahalingam
- Institute of Drug Development, Division of Cancer Research and Therapy Center, University of Texas Health Science Center, San Antonio, Texas, USA
| | | | | | | | | |
Collapse
|
103
|
Abstract
Lung cancer is the leading cause of cancer-related mortality in the United States. Patients with advanced disease have a median survival of approximately 10 months when treated with standard platinum-based therapy. Improvements in our understanding of cancer biology have led to the development of novel agents that more precisely affect the target of interest, allowing for a more rational approach to clinical trial design. Angiogenesis, the growth of new vessels from pre-existing vessels, is a fundamental step in tumor growth and progression. Inhibition of tumor-related angiogenesis has become an attractive target for anticancer therapy. A large randomized trial demonstrated an improvement in overall survival when bevacizumab, a monoclonal antibody against vascular endothelial growth factor (VEGF), was combined with chemotherapy in patients with advanced non-small cell lung cancer (NSCLC). Small molecule inhibitors targeting both the VEGF receptor (VEGFR) and the tyrosine kinase receptor have also shown promise when combined with standard chemotherapy, but their role in the treatment of patients with NSCLC remains to be determined. This paper reviews clinical trials that have incorporated antiangiogenic agents in the treatment of patients with NSCLC.
Collapse
|
104
|
Sano D, Choi S, Milas ZL, Zhou G, Galer CE, Su YW, Gule M, Zhao M, Zhu Z, Myers JN. The effect of combination anti-endothelial growth factor receptor and anti-vascular endothelial growth factor receptor 2 targeted therapy on lymph node metastasis: a study in an orthotopic nude mouse model of squamous cell carcinoma of the oral tongue. ACTA ACUST UNITED AC 2009; 135:411-20. [PMID: 19380367 DOI: 10.1001/archoto.2009.14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
OBJECTIVE To evaluate the therapeutic effect of treatment with a combination of the monoclonal antibodies to the vascular endothelial growth factor receptor (DC101) and the epidermal growth factor receptor (cetuximab) in an orthotopic nude mouse model of metastatic squamous cell carcinoma of the oral tongue (SCCOT). DESIGN In vivo study. SETTING A translational research laboratory at a comprehensive cancer center. SUBJECTS Male athymic nude mice aged 8 to 12 weeks. INTERVENTION To develop orthotopic nude mouse models of SCCOT, OSC-19 cells or luciferase (Luc)-expressing OSC-19-Luc and JMAR-Luc cells were injected into the tongues of nude mice. Animals were randomly divided into 4 groups: DC101 alone, cetuximab alone, DC101 plus cetuximab, or placebo, and all treatments were administered twice per week for 4 weeks. The in vivo antitumor activity was monitored noninvasively by bioluminescence imaging. Tumors were resected at necropsy, and immunohistochemical and immunofluorescent staining were performed. MAIN OUTCOME MEASURES Tumor size, bioluminescence, animal survival, and percentage of animals with lymph node metastasis. RESULTS At the conclusion of the treatment period, the mean tumor volumes in the cetuximab alone and the DC101 plus cetuximab groups had decreased significantly compared with those that received the placebo control (68% [P = .002] and 84% [P < .001], respectively). Significant effects of the treatment were also observed in bioluminescence imaging. Mice treated with DC101 plus cetuximab also lived longer and had a lower incidence of neck lymph node metastases compared with the control group (P = .003). CONCLUSIONS Treatment with DC101 plus cetuximab inhibited the growth of SCCOT and decreased the incidence of the neck lymph node metastases in vivo. These results suggest that this combination treatment may be an effective strategy against metastatic SCCOT and warrants further preclinical trials.
Collapse
Affiliation(s)
- Daisuke Sano
- Department of Head and Neck Surgery, Unit 441, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030-4009, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Aliu SO, Wilmes LJ, Moasser MM, Hann BC, Li KL, Wang D, Hylton NM. MRI methods for evaluating the effects of tyrosine kinase inhibitor administration used to enhance chemotherapy efficiency in a breast tumor xenograft model. J Magn Reson Imaging 2009; 29:1071-9. [PMID: 19388114 DOI: 10.1002/jmri.21737] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
PURPOSE To evaluate whether quantitative MRI parameters are sensitive to the effects of the tyrosine kinase inhibitor gefitinib and can discriminate between two different treatment protocols. MATERIALS AND METHODS Untreated mice with BT474 breast tumor xenografts were characterized in a preliminary study. Subsequently, tumor volume, apparent diffusion coefficient (ADC), transendothelial permeability (K(ps)), and fractional plasma volume (fPV) were measured in three groups of mice receiving: 1) control vehicle for 10 days, or gefitinib as 2) a single daily dose for 10 days or 3) a 2-day pulsed dose. RESULTS Gefitinib treatment resulted in significant tumor growth inhibition (pulsed: 439 +/- 93; daily: 404 +/- 53; control: 891 +/- 174 mm(3), P < 0.050) and lower cell density (pulsed: 0.15 +/- 0.01, daily: 0.17 +/- 0.01, control: 0.24 +/- 0.01, P < 0.050) after 9 days. Tumor ADC increased in treated groups but decreased in controls (P > 0.050). Tumor K(ps) decreased with pulsed treatment but rebounded afterwards and increased with daily treatment (P > 0.050). Tumor fPV increased in both treated groups, decreasing afterwards with pulsed treatment (P > 0.050). CONCLUSION Quantitative MRI can provide a sensitive measure of gefitinib-induced tumor changes, potentially distinguish between treatment regimens, and may be useful for determining optimal treatment scheduling for enhancing chemotherapy delivery.
Collapse
Affiliation(s)
- S O Aliu
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California 94107, USA.
| | | | | | | | | | | | | |
Collapse
|
106
|
Horn L, Sandler AB. Emerging data with antiangiogenic therapies in early and advanced non-small-cell lung cancer. Clin Lung Cancer 2009; 10 Suppl 1:S7-16. [PMID: 19362948 PMCID: PMC3528174 DOI: 10.3816/clc.2009.s.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lung cancer is the leading cause of cancer-related mortality in the United States. Patients treated with adjuvant chemotherapy have a 5-year survival rate of 25% to 70% depending on stage, whereas those with advanced disease have a median survival of approximately 8 months when treated with standard platinum-based therapy. Improvements in our understanding of cancer biology have led to the development of novel agents that more precisely affect the target of interest, allowing for a more rational approach to clinical trial design. Angiogenesis, the growth of new vessels from preexisting vessels, is a fundamental step in tumor growth and progression. Inhibition of tumor-related angiogenesis has become an attractive target for anticancer therapy. Bevacizumab, a monoclonal antibody against vascular endothelial growth factor (VEGF), is the most studied antiangiogenic agent in patients with non-small-cell lung cancer (NSCLC). There was an improvement in overall survival when bevacizumab was combined with paclitaxel and carboplatin in patients with advanced NSCLC that was not seen when bevacizumab was combined with cisplatin and gemcitabine. Studies with bevacizumab in the adjuvant and advanced setting are ongoing in patients with NSCLC. Small-molecule inhibitors targeting the VEGF receptor and the tyrosine kinase receptor have also shown promise when combined with standard chemotherapy, but their role in the treatment of patients with NSCLC remains to be determined. This article reviews clinical trials that have incorporated antiangiogenic agents in the treatment of patients with NSCLC.
Collapse
Affiliation(s)
- Leora Horn
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | | |
Collapse
|
107
|
Mahalingam D, Giles F. Challenges in developing targeted therapy for pancreatic adenocarcinoma. Expert Opin Ther Targets 2009; 12:1389-401. [PMID: 18851695 DOI: 10.1517/14728222.12.11.1389] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic adenocarcinoma is a leading cause of cancer deaths in the US. Gemcitabine-based chemotherapy remains the cornerstone treatment for advanced pancreatic cancers. Research into the molecular pathogenesis of pancreatic cancers has allowed scientists to understand the complex heterogeneous signals associated with them. Targeting these pathways with chemical inhibitors could improve patient outcome. OBJECTIVE To describe the molecular heterogeneity typical of pancreatic cancers and to discuss targeted therapies in development, and the challenges facing these agents. METHODS We reviewed Pub Med. literature, clinical trial database (clinicaltrials.gov), American Society of Clinical Oncology (ASCO) and American Association of Cancer Research (AACR) websites. CONCLUSIONS Molecular pathogenesis of pancreatic cancer involves multiple pathways and defined mutations. This molecular heterogeneity is a major reason for failure of targeted therapy. Targeting multiple oncogenic pathways using novel targeted therapies could improve patient survival.
Collapse
Affiliation(s)
- Devalingam Mahalingam
- Institute of Drug Development, Division of Hematology and Medical Oncology, University of Texas Health Science Centre, San Antonio, Texas 78229, USA
| | | |
Collapse
|
108
|
Flores AI, Bedoya F, Grau M, de Salamanca RE, Vegh I. In vivo effect of an luteinizing hormone-releasing hormone analog on vascular endothelial growth factor and epidermal growth factor receptor expression in mammary tumors. J Carcinog 2009; 8:11. [PMID: 19491505 PMCID: PMC2699606 DOI: 10.4103/1477-3163.51852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 04/02/2009] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The hypothalamic luteinizing hormone-releasing hormone (LHRH) is well known for its role in the control of pituitary gonadotropin secretion and it has demonstrated a direct antiproliferative effect on some cancer cell lines of LHRH and its synthetic analogs. The study was designed to assess whether administration of the LHRH analog (goserelin) has any effect on the expression of the vascular endothelial growth factor (VEGF) and the epidermal growth factor receptor (EGFR) in rats with N-nitroso-N-methylurea (NMU)-induced-mammary tumors " in vivo". MATERIALS AND METHODS The animals with tumors were assessed after acute or chronic treatment with goserelin, and in all the animals VEGF and EGFR expression was examined both in plasma and tumor homogenates by enzyme immunoassay. RESULTS The basal plasma values of VEGF were lower in the healthy control group than in rats with NMU-induced tumors ( P = 0.025). Following acute treatment with goserelin, VEGF expression in plasma increased above basal levels after 60 min ( P = 0.05) and dropped during chronic treatment. Likewise, in the tumor homogenate the mean VEGF expression was higher at 60 min post-goserelin administration than the basal levels, although VEGF expression then diminished at 90 min. Plasma EGFR expression was higher in rats with NMU-induced tumors than in healthy controls ( P Conclusions: The results allow us to conclude that goserelin may exert a short-term stimulatory effect on the release of VEGF, as well as a long-term inhibitory effect on VEGF but not EGFR expression.
Collapse
Affiliation(s)
- Ana Isabel Flores
- Centro de Investigación, Hospital Universitario 12 de Octubre, Av. Córdoba s/n, CP28041, Madrid, Spain.
| | | | | | | | | |
Collapse
|
109
|
Döme B, Magyar M. [Tumor vasculature as a therapeutic target in non-small cell lung cancer]. Magy Onkol 2008; 52:247-59. [PMID: 18845495 DOI: 10.1556/monkol.52.2008.3.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite developments in conventional (chemo)radiotherapy and surgery, survival of non-small cell lung cancer (NSCLC) patients remains poor. Treatments with targeted molecular drugs offer novel therapeutic strategies. Bevacizumab, a recombinant anti-vascular endothelial growth factor (VEGF) antibody, is the antiangiogenic drug at the most advanced stage of development in the therapy of NSCLC. However, a number of questions and future challenges relating to the use of bevacizumab in NSCLC remain. Furthermore, novel agents targeting the pre-existing NSCLC vasculature (i.e. vascular disrupting agents, VDAs) or multiple tyrosine kinase inhibitors have emerged as unique drug classes delivering promising results in several preclinical and clinical studies. Herein, we review the most recent data using these novel targeted agents either alone or in combination with chemotherapy in NSCLC.
Collapse
Affiliation(s)
- Balázs Döme
- Országos Korányi TBC és Pulmonológiai Intézet IV. Tüdogyógyászati Osztály 1529 Budapest Piheno u. 1. Országos Korányi TBC és Pulmonológiai Intézet Tumorbiológiai Osztály Budapest.
| | | |
Collapse
|
110
|
Vincenzi B, Schiavon G, Silletta M, Santini D, Tonini G. The biological properties of cetuximab. Crit Rev Oncol Hematol 2008; 68:93-106. [DOI: 10.1016/j.critrevonc.2008.07.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 07/07/2008] [Accepted: 07/11/2008] [Indexed: 11/25/2022] Open
|
111
|
Si FC. Inhibitory effect of Qigesan and its separated formula on angiogenesis of esophageal carcinoma Eca109 cell-transplanted nude mouse tumor. Shijie Huaren Xiaohua Zazhi 2008; 16:3139-3145. [DOI: 10.11569/wcjd.v16.i28.3139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the effects and mechanism of Qigesan and its separated formula on tumor angiogenesis.
METHODS: Exponentially growing Eca109 human esophageal carcinoma cells (1 × 106 cells/mouse) were injected subcutaneously into the flank of 6- week-old female BALB/c nude mice. On the next day after inoculation, the decoctions (40 folds of adult dosage) of Qigesan (W) and its separated formula (blood-activating group P and resolving phlegm group R) were administered by oral gavage once a day for 60 days. Microvessel density (MVD) and VEGF expression in the transplanted tumor was detected using immunohistochemtry, and the protein expressions of EGFR, PDGFR, VEGF and phospholipase C-γ1 (PLC-γ1) were determined using Western blot.
RESULTS: Microvessel density (MVD) in the transplanted tumor of all treatment groups were markedly decreased compared with that in control group (36.43 ± 4.16, 40.29 ± 2.87 42.43 ± 3.04 vessels/mm2vs 48.57 ± 7.45 vessels/mm2, P < 0.05 or 0.01) MVD and VEGF had statistically positive correlation by SPASS13.0 (r = 0.712, P = 0.0005). The protein expressions of EGFR, PDGFR, VEGF and phospholipase C-γ1 (PLC-γ1) were obviously decreased compared with that in control group. Inhibitory effects of Qigesan and its separated formula on MVD and those protein expressions were the best in group W and second in group P.
CONCLUSION: Qigesan and its separated formula can inhibit tumor angiogenesis, which is related to the inhibition of protein expressions of EGFR, PDGFR, VEGF and PLC-γ1.
Collapse
|
112
|
Choi S, Sano D, Cheung M, Zhao M, Jasser SA, Ryan AJ, Mao L, Chen WT, El-Naggar AK, Myers JN. Vandetanib Inhibits Growth of Adenoid Cystic Carcinoma in an Orthotopic Nude Mouse Model. Clin Cancer Res 2008; 14:5081-9. [DOI: 10.1158/1078-0432.ccr-08-0245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
113
|
Bianco R, Rosa R, Damiano V, Daniele G, Gelardi T, Garofalo S, Tarallo V, De Falco S, Melisi D, Benelli R, Albini A, Ryan A, Ciardiello F, Tortora G. Vascular endothelial growth factor receptor-1 contributes to resistance to anti-epidermal growth factor receptor drugs in human cancer cells. Clin Cancer Res 2008; 14:5069-80. [PMID: 18694994 DOI: 10.1158/1078-0432.ccr-07-4905] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The resistance to selective EGFR inhibitors involves the activation of alternative signaling pathways, and Akt activation and VEGF induction have been described in EGFR inhibitor-resistant tumors. Combined inhibition of EGFR and other signaling proteins has become a successful therapeutic approach, stimulating the search for further determinants of resistance as basis for novel therapeutic strategies. EXPERIMENTAL DESIGN We established human cancer cell lines with various degrees of EGFR expression and sensitivity to EGFR inhibitors and analyzed signal transducers under the control of EGFR-dependent and EGFR-independent pathways. RESULTS Multitargeted inhibitor vandetanib (ZD6474) inhibited the growth and the phosphorylation of Akt and its effector p70S6 kinase in both wild-type and EGFR inhibitor-resistant human colon, prostate, and breast cancer cells. We found that the resistant cell lines exhibit, as common feature, VEGFR-1/Flt-1 overexpression, increased secretion of VEGF and placental growth factor, and augmented migration capabilities and that vandetanib is able to antagonize them. Accordingly, a new kinase assay revealed that in addition to VEGF receptor (VEGFR)-2, RET, and EGFR, vandetanib efficiently inhibits also VEGFR-1. The contribution of VEGFR-1 to the resistant phenotype was further supported by the demonstration that VEGFR-1 silencing in resistant cells restored sensitivity to anti-EGFR drugs and impaired migration capabilities, whereas exogenous VEGFR-1 overexpression in wild-type cells conferred resistance to these agents. CONCLUSIONS This study shows that VEGFR-1 contributes to anti-EGFR drug resistance in different human cancer cells. Moreover, vandetanib inhibits VEGFR-1 activation, cell proliferation, and migration, suggesting its potential utility in patients resistant to EGFR inhibitors.
Collapse
Affiliation(s)
- Roberto Bianco
- Cattedra di Oncologia Medica, Dipartimento di Endocrinologia e Oncologia Molecolare e Clinica, Università di Napoli Federico II, Via S. Pansini 5, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Tortora G, Ciardiello F, Gasparini G. Combined targeting of EGFR-dependent and VEGF-dependent pathways: rationale, preclinical studies and clinical applications. ACTA ACUST UNITED AC 2008; 5:521-30. [PMID: 18594498 DOI: 10.1038/ncponc1161] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 12/10/2007] [Indexed: 11/09/2022]
Abstract
Cellular heterogeneity, redundancy of molecular pathways and effects of the microenvironment contribute to the survival, motility and metastasis of cells in solid tumors. It is unlikely that tumors are entirely dependent on only one abnormally activated signaling pathway; consequently, treatment with an agent that interferes with a single target may be insufficient. Combined blockade of functionally linked and relevant multiple targets has become an attractive therapeutic strategy. The EGFR and ERBB2 (HER2) pathways and VEGF-dependent angiogenesis have a pivotal role in cancer pathogenesis and progression. Robust experimental evidence has shown that these pathways are functionally linked and has demonstrated a suggested role for VEGF in the acquired resistance to anti-ERBB drugs when these receptors are pharmacologically blocked. Combined inhibition of ERBB and VEGF signaling interferes with a molecular feedback loop responsible for acquired resistance to anti-ERBB agents and promotes apoptosis while ablating tumor-induced angiogenesis. To this aim, either two agents highly selective against VEGF and ERBB respectively, or, alternatively, a single multitargeted agent, can be used. Preclinical studies have proven the efficacy of both these approaches and early clinical studies have provided encouraging results. This Review discusses the experimental rationale for, preclinical studies of and clinical trials on combined blockade of ERBB and VEGF signaling.
Collapse
Affiliation(s)
- Giampaolo Tortora
- Clinical Unit and Laboratories of Molecular Therapy at the University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
115
|
Liver regeneration and tumor stimulation--a review of cytokine and angiogenic factors. J Gastrointest Surg 2008; 12:966-80. [PMID: 18181006 DOI: 10.1007/s11605-007-0459-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 12/06/2007] [Indexed: 01/31/2023]
Abstract
Liver resection for metastatic (colorectal carcinoma) tumors is often followed by a significant incidence of tumor recurrence. Cellular and molecular changes resulting from hepatectomy and the subsequent liver regeneration process may influence the kinetics of tumor growth and contribute to recurrence. Clinical and experimental evidence suggests that factors involved in liver regeneration may also stimulate the growth of occult tumors and the reactivation of dormant micrometastases. An understanding of the underlying changes may enable alternative strategies to minimize tumor recurrence and improve patient survival after hepatectomy.
Collapse
|
116
|
Gupta P, Emdad L, Lebedeva IV, Sarkar D, Dent P, Curiel DT, Settleman J, Fisher PB. Targeted combinatorial therapy of non-small cell lung carcinoma using a GST-fusion protein of full-length or truncated MDA-7/IL-24 with Tarceva. J Cell Physiol 2008; 215:827-36. [PMID: 18270968 DOI: 10.1002/jcp.21369] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), a cytokine belonging to the IL-10 family, displays cancer-specific apoptosis-inducing properties when delivered by a replication-incompetent adenovirus (Ad.mda-7) or as a GST-tagged recombinant protein (GST-MDA-7). Previous studies demonstrated that an adenovirus expressing M4, a truncated version of MDA-7/IL-24 containing amino acid residues 104-206, also induced similar cancer-specific apoptosis. We generated recombinant GST-M4 proteins and examined the potency of GST-MDA-7 and GST-M4 on a panel of epidermal growth factor receptor (EGFR) wild type and mutant non-small cell lung carcinoma (NSCLC) cells either as a single agent or in combination with a reversible EGFR inhibitor, Tarceva. The combination of either GST-MDA-7 or GST-M4 ( approximately 0.1 microM) and Tarceva (10 microM), at sub-optimal apoptosis-inducing concentrations synergistically enhanced growth inhibition and apoptosis induction over that observed with either agent alone. The combination treatment also augmented inhibition of EGFR signaling, analyzed by phosphorylation of EGFR and its downstream effectors AKT and ERK1/2, over that with single-agent therapy. Tarceva enhanced GST-MDA-7 and GST-M4 toxicity in cells expressing mutated EGFR proteins that are resistant to the inhibitory effects of Tarceva. In total, these data suggest that combined treatment of NSCLC cells with an EGFR inhibitor can augment the efficacy of GST-MDA-7 and GST-M4 and that the EGFR inhibitor Tarceva may mediate this combinatorial effect by inhibiting multiple tyrosine kinases in addition to the EGFR. This approach highlights a potential new combinatorial strategy, which may prove beneficial for NSCLC patients with acquired resistance to EGFR inhibitors.
Collapse
Affiliation(s)
- Pankaj Gupta
- Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University, College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
117
|
Dougherty U, Sehdev A, Cerda S, Mustafi R, Little N, Yuan W, Jagadeeswaran S, Chumsangsri A, Delgado J, Tretiakova M, Joseph L, Hart J, Cohen EEW, Aluri L, Fichera A, Bissonnette M. Epidermal growth factor receptor controls flat dysplastic aberrant crypt foci development and colon cancer progression in the rat azoxymethane model. Clin Cancer Res 2008; 14:2253-2262. [PMID: 18413814 DOI: 10.1158/1078-0432.ccr-07-4926] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Colonic carcinogenesis deranges growth-regulating epidermal growth factor receptors (EGFR). We previously showed that EGFR signals were up-regulated in human aberrant crypt foci (ACF), putative colon cancer precursors. The azoxymethane model of colon cancer recapitulates many aspects of human colonic tumors. Recent studies indicate that flat dysplastic ACF with increased beta-catenin are tumor precursors in this model. We asked, therefore, if EGFR signals are required for flat dysplastic ACF development and cancer progression. EXPERIMENTAL DESIGN Rats received azoxymethane or saline, and standard chow or chow supplemented with gefitinib, an EGFR inhibitor, for 44 weeks. EGFR signals were quantified in normal colon, flat ACF, and tumors by computerized analysis of immunostains and Western blots. K-ras mutations were assessed by PCR and mRNA for egfr ligands by quantitative real-time PCR. RESULTS EGFR inhibition with gefitinib decreased the incidence of flat dysplastic ACF from 66% to 36% and tumors from 71% to 22% (P < 0.05). This inhibitor also reduced the overexpressions of cyclin D1 and Cox-2 in flat ACF. Furthermore, in flat ACF, EGFR blockade decreased the up-regulation of c-Jun, FosB, phosphorylated active signal transducers and activators of transcription 3, and CCAAT/enhancer binding protein-beta, potential regulators of cyclin D1 and Cox-2. In colonic tumors, EGFR blockade significantly decreased angiogenesis, proliferation, and progression while also increasing apoptosis (P < 0.05). Gefitinib also inhibited the activations of extracellular signal-regulated kinase, Src, and AKT pathways in tumors. CONCLUSIONS We have shown for the first time that EGFR promotes the development of flat dysplastic ACF and the progression of malignant colonic tumors. Furthermore, we have mechanistically identified several transcription factors and their targets as EGFR effectors in colonic carcinogenesis.
Collapse
Affiliation(s)
- Urszula Dougherty
- Department of Medicine, University of Chicago Hospitals and Clinics, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Gangjee A, Namjoshi OA, Yu J, Ihnat MA, Thorpe JE, Warnke LA. Design, synthesis and biological evaluation of substituted pyrrolo[2,3-d]pyrimidines as multiple receptor tyrosine kinase inhibitors and antiangiogenic agents. Bioorg Med Chem 2008; 16:5514-28. [PMID: 18467105 DOI: 10.1016/j.bmc.2008.04.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 04/04/2008] [Accepted: 04/04/2008] [Indexed: 01/05/2023]
Abstract
Direct and indirect involvement of receptor tyrosine kinases (RTKs) in tumor growth and metastasis makes them ideal targets for anticancer therapy. A paradigm shift from inhibition of single RTK to inhibition of multiple RTKs has been recently demonstrated. We designed and synthesized eight N(4)-phenylsubstituted-6-(2-phenylethylsubstituted)-7H-pyrrolo[2,3-d]pyrimidine-2,4-diamines as homologated series of our previously published RTK inhibitors. We reasoned that increased flexibility of the side chain, which determines potency and selectivity, would improve the spectrum of RTK inhibition. These compounds were synthesized using a bis-electrophilic cyclization to afford substituted pyrrolo[2,3-d]pyrimidines followed by chlorination and substitution at the 4-position with various anilines. Five additional compounds of this series were previously reported by Gangjee et al.(1) with activities against IGFR only. Their synthesis, characterization and biological activities against a variety of other RTKs are reported in this study for the first time. The biological evaluation, in whole cell assays, showed several analogs had remarkable inhibitory activity against epithelial growth factor receptor (EGFR), vascular endothelial growth factor receptor-1 (VEGFR-1), platelet-derived growth factor receptor-beta (PDGFR-beta), the growth of A431 cells in culture, and in the chicken embryo chorioallantoic membrane (CAM) angiogenesis assay. The inhibitory data against the RTKs in this study demonstrate that variation of the 6-ethylaryl substituents as well as the N(4)-phenyl substituents of these analogs does indeed control both the potency and specificity of inhibitory activity against RTKs. In addition, homologation of the chain length of the 6-substituent from a methylene to an ethyl increases the spectrum of RTK inhibition. New multi-RTK inhibitors (8, 12) and potent inhibitors of angiogenesis (15, 19) were identified with the best compound, N(4)-(3-trifluromethylphenyl)-6-(2-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine (15), with an IC(50) value of 30nM in the CAM angiogenesis inhibition assay.
Collapse
Affiliation(s)
- Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA.
| | | | | | | | | | | |
Collapse
|
119
|
Moasser MM, Wilmes LJ, Wong CH, Aliu S, Li KL, Wang D, Hom YK, Hann B, Hylton NM. Improved tumor vascular function following high-dose epidermal growth factor receptor tyrosine kinase inhibitor therapy. J Magn Reson Imaging 2008; 26:1618-25. [PMID: 17968965 DOI: 10.1002/jmri.21196] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To determine if inhibitors of the human growth factor receptor (HER) family can be used to enhance tumor vascular permeability and perfusion and optimize the efficacy of cytotoxic chemotherapeutics. Poor tumor vascular function limits the delivery and efficacy of cancer chemotherapeutics and HER family tyrosine kinases mediate tumor-endothelial signaling in both of these compartments. MATERIALS AND METHODS BT474 human breast cancer tumors were established in mice and the biologic effects of the HER tyrosine kinase inhibitor (TKI) gefitinib on tumor vascular function was determined by dynamic contrast-enhanced MRI (DCE-MRI), and on tumor vascular architecture and perfusion by immunofluorescence microscopy. RESULTS A brief dose of gefitinib enhances the antitumor activity of paclitaxel in vivo but not in cell culture, suggesting that its chemoenhancing activity involves the in vivo microenvironment. A brief high dose of gefitinib induces a decrease in endothelial transfer constant (Kps) and a concomitant increase in tumor fractional plasma volume (fPV). These changes are accompanied by a rapid reduction in tumor volume, likely due to decreased tumor edema, and modestly improved tumor vascular architecture and perfusion on microscopy. CONCLUSION These data suggest that HER family TKIs have the potential to optimize the tumor microenvironment for delivery of cytotoxic chemotherapeutics.
Collapse
Affiliation(s)
- Mark M Moasser
- Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Abstract
PURPOSE OF REVIEW The use of targeted therapies, particularly those against the key mediator of angiogenesis, vascular endothelial growth factor, has the potential to improve outcomes for nonsmall cell lung cancer (NSCLC) patients. This review summarizes recent findings on targeting mediators of angiogenesis, treatment options and molecular targets in development. RECENT FINDINGS Bevacizumab, a recombinant humanized monoclonal antivascular endothelial growth factor antibody, in a phase III study, showed significantly improved overall and progression-free survival when used in combination with standard first-line chemotherapy in patients with advanced NSCLC and was generally well tolerated. Adverse events, including tumor-related bleeding, have been noted in some patients, predominantly those with squamous cell histology or centrally located tumors. SUMMARY Several small-molecule vascular endothelial growth factor receptor tyrosine kinase inhibitors have also shown promise in phase I and II trials in NSCLC. This review summarizes the most important findings on angiogenesis inhibitors in NSCLC and discusses the potential for the use of these novel agents in different settings.
Collapse
|
121
|
SPECT imaging with 99mTc-labeled EGFR-specific nanobody for in vivo monitoring of EGFR expression. Mol Imaging Biol 2008; 10:167-75. [PMID: 18297364 DOI: 10.1007/s11307-008-0133-8] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 11/30/2007] [Accepted: 12/14/2007] [Indexed: 01/19/2023]
Abstract
PURPOSE Overexpression of the epidermal growth factor receptor (EGFR) occurs with high incidence in various carcinomas. The oncogenic expression of the receptor has been exploited for immunoglobulin-based diagnostics and therapeutics. We describe the use of a llama single-domain antibody fragment, termed Nanobody, for the in vivo radioimmunodetection of EGFR overexpressing tumors using single photon emission computed tomography (SPECT) in mice. METHODS Fluorescence-activated cell sorting (FACS) analysis was performed to evaluate the specificity and selectivity of 8B6 Nanobody to bind EGFR on EGFR overexpressing cells. The Nanobody was then labeled with (99m)Tc via its C-terminal histidine tail. Uptake in normal organs and tissues was assessed by ex vivo analysis. In vivo tumor targeting of (99m)Tc-8B6 Nanobody was evaluated via pinhole SPECT in mice bearing xenografts of tumor cells with either high (A431) or moderate (DU145) overexpression of EGFR. RESULTS FACS analysis indicated that the 8B6 Nanobody only recognizes cells overexpressing EGFR. In vivo blood clearance of (99m)Tc-8B6 Nanobody is relatively fast (half-life, 1.5 h) and mainly via the kidneys. At 3 h postinjection, total kidney accumulation is high (46.6+/-0.9%IA) compared to total liver uptake (18.9+/-0.6%IA). Pinhole SPECT imaging of mice bearing A431 xenografts showed higher average tumor uptake (5.2+/-0.5%IA/cm(3)) of (99m)Tc-8B6 Nanobody compared to DU145 xenografts (1.8+/-0.3%IA/cm(3), p<0.001). CONCLUSION The EGFR-binding Nanobody investigated in this study shows high specificity and selectivity towards EGFR overexpressing cells. Pinhole SPECT analysis with (99m)Tc-8B6 Nanobody enabled in vivo discrimination between tumors with high and moderate EGFR overexpression. The favorable biodistribution further corroborates the suitability of Nanobodies for in vivo tumor imaging.
Collapse
|
122
|
Loupakis F, Vasile E, Santini D, Masi G, Falcone A, Graziano F. EGF-receptor targeting with monoclonal antibodies in colorectal carcinomas: rationale for a pharmacogenomic approach. Pharmacogenomics 2008; 9:55-69. [DOI: 10.2217/14622416.9.1.55] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Monoclonal antibodies directed against the EGF-receptor (EGFR) have recently been approved for the treatment of metastatic colorectal cancer (CRC) patients with EGFR-positive tumors at immunohistochemistry (IHC). Surprisingly, data demonstrate a lack of correlation between the tumor’s EGFR expression at IHC and outcome. Indeed, as pointed out from small experiences, patients with EGFR-IHC-negative metastatic CRC have the same chance as EGFR-IHC-positive patients to benefit from an anti-EGFR monoclonal antibody, underlying the importance of different, more reliable, selection criteria. In particular, the identification of such predictive factors is important as these agents are expensive, have side effects and are really only effective in a minority of patients. Several potential clinical and biological predictive markers of activity and/or efficacy for such agents have been evaluated in retrospective series with promising results. Moving from clinical data suggesting that there could be a subpopulation of CRC patients that are more liable to benefit from anti-EGFR monoclonal antibodies, here we review major studies on determinants of outcome in this field.
Collapse
Affiliation(s)
- Fotios Loupakis
- Azienda USL 6 Livorno, Division of Medical Oncology, Viale Alfieri 36, 57100 Livorno, Italy
| | - Enrico Vasile
- Azienda USL 6 Livorno, Division of Medical Oncology, Viale Alfieri 36, 57100 Livorno, Italy
| | - Daniele Santini
- University Campus Biomedico, Medical Oncology, Via Emilio Longoni, 47 00155, Rome
| | - Gianluca Masi
- Azienda USL 6 Livorno, Division of Medical Oncology, Viale Alfieri 36, 57100 Livorno, Italy
| | - Alfredo Falcone
- Azienda USL 6 Livorno, Division of Medical Oncology, Viale Alfieri 36, 57100 Livorno, Italy
- University of Pisa, Department of Oncology, Via Roma, 55 56100, Pisa, Italy
| | - Francesco Graziano
- Azienda Ospedale San Salvatore, Deparment of Onco-hematology, Medical Oncology Unit, Pesaro, Italy
| |
Collapse
|
123
|
Melo MACD, Violet C. Quimioterapia neoadjuvante torna ressecável carcinoma de cabeça de pâncreas. Rev Col Bras Cir 2007. [DOI: 10.1590/s0100-69912007000400014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
124
|
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
|
125
|
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: 337] [Impact Index Per Article: 18.7] [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
|
126
|
Hammarsten P, Rudolfsson SH, Henriksson R, Wikström P, Bergh A. Inhibition of the epidermal growth factor receptor enhances castration-induced prostate involution and reduces testosterone-stimulated prostate growth in adult rats. Prostate 2007; 67:573-81. [PMID: 17252557 DOI: 10.1002/pros.20529] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) mediates regulatory signals in the normal prostate, but the functional importance of this is unclear. METHODS Adult male rats were castrated, or castrated + treated with gefitinib (Iressa, ZD1839), an EGFR tyrosine kinase inhibitor, for 3 days. Seven-day castrated rats were treated with testosterone, or testosterone + gefitinib, for 3 days. RESULTS Both castration alone and testosterone treatment in castrated animals increased the mRNA and protein levels of EGFR and phospho-EGFR in the ventral prostate. Inhibition of EGFR during castration and during testosterone-stimulated prostate growth resulted in a decrease in total epithelial weight, epithelial cell proliferation, endothelial cell proliferation, and increased epithelial cell apoptosis. CONCLUSIONS This study suggests that increased EGFR signaling during castration mediates stimulatory effects balancing castration-induced prostate regression, and that EGFR signaling is a necessary component in testosterone-stimulated prostate growth.
Collapse
Affiliation(s)
- Peter Hammarsten
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | | | | | | |
Collapse
|
127
|
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]
|
128
|
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: 139] [Impact Index Per Article: 7.0] [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
|
129
|
de Gramont A, Van Cutsem E. Investigating the potential of bevacizumab in other indications: metastatic renal cell, non-small cell lung, pancreatic and breast cancer. Oncology 2005; 69 Suppl 3:46-56. [PMID: 16301835 DOI: 10.1159/000088483] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bevacizumab (Avastin) has unprecedented survival benefit in patients with metastatic colorectal cancer. Trials are already in progress to investigate the potential of bevacizumab in indications including metastatic renal cell cancer (RCC), non-small cell lung cancer (NSCLC), pancreatic cancer, breast and ovarian cancer. Bevacizumab offers the potential to increase survival without substantially altering the toxicity profile in these tumor types. Bevacizumab has shown activity in patients with refractory metastatic RCC, where progression-free survival (PFS) was significantly longer in patients treated with bevacizumab (10 mg/kg every 2 weeks) than those treated with placebo (hazard ratio=2.55, p<0.001). In addition, combining bevacizumab with erlotinib (Tarceva) has shown a median time to progression of more than 11 months. In NSCLC, a phase II trial revealed that adding bevacizumab to chemotherapy increased therapeutic benefit compared with chemotherapy alone. Adverse events were mild and easily managed, but six patients receiving bevacizumab developed severe hemoptysis. Entry criteria for NSCLC trials have been adjusted to exclude patients with squamous cell histology to try to avoid this issue. Adding bevacizumab (10 mg/kg) to the current standard of care, gemcitabine, in stage IV pancreatic cancer has also shown promising efficacy. Partial responses were seen in 19% of patients, with a further 48% having stable disease. Several ongoing clinical trials are also studying bevacizumab with various chemotherapy and radiotherapy regimens. Bevacizumab combined with carboplatin (Paraplatin)/paclitaxel (Taxol) was further examined in a phase III randomized trial that accrued 878 patients with advanced non-squamous cell NSCLC. Patients given chemotherapy (paclitaxel and carboplatin) plus bevacizumab had a higher response rate, longer PFS and an increase in survival compared with patients on chemotherapy alone. Both regimens were generally well tolerated. Bevacizumab has also shown activity in patients with metastatic breast cancer. In 715 patients, a significant, 2-fold increase in response rate was observed in patients receiving bevacizumab plus paclitaxel compared with paclitaxel alone. Median PFS was also significantly increased (p<0.001). Bevacizumab has the potential to provide significant efficacy benefits for patients with metastatic RCC, NSCLC, pancreatic cancer, and other tumor types when used first line in combination with standard therapy.
Collapse
|