51
|
IGF-1R/epithelial-to-mesenchymal transition (EMT) crosstalk suppresses the erlotinib-sensitizing effect of EGFR exon 19 deletion mutations. Sci Rep 2014; 3:2560. [PMID: 23994953 PMCID: PMC3759044 DOI: 10.1038/srep02560] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 07/31/2013] [Indexed: 02/07/2023] Open
Abstract
Using non-small cell lung carcinoma (NSCLC) cells harboring the erlotinib-sensitizing Epidermal Growth Factor Receptor (EGFR) exon 19 mutation delE746-A750, we developed erlotinib-refractory derivatives in which hyperactive Insulin-like Growth Factor-1 Receptor (IGF-1R) signaling associated with enrichment in epithelial-to-mesenchymal transition (EMT)-related morphological and transcriptional features. We then explored whether an IGF-1R/EMT crosstalk was sufficient to promote erlotinib refractoriness in the absence of second-site EGFR mutations, MET and AXL hyperactivation. Transforming Growth Factor-beta1 (TGFβ1)-induced mesenchymal trans-differentiation was sufficient to impede erlotinib functioning in the presence of drug-sensitive delE746-A750 EGFR mutation. Pharmacological blockade of IGF-1R fully prevented the TGFβ1's ability to activate an EMT protein signature [E-cadherin low/vimentin high]. The sole presence of erlotinib was capable of rapidly activate an IGF-1R-dependent, vimentin-enriched mesenchymal-like phenotype in delE746-A750-mutated epithelial cells. Even if transient, NSCLC cells' intrinsic plasticity to undergo crosstalk between IGF-1R and EMT signaling pathways can sufficiently eliminate the erlotinib-sensitizing effect of highly prevalent EGFR mutations and suggests the urgent need for dual IGF-1R/EMT-targeting strategies to circumvent erlotinib resistance.
Collapse
|
52
|
Reinmuth N, Kloos S, Warth A, Risch A, Muley T, Hoffmann H, Thomas M, Meister M. Insulin-like growth factor 1 pathway mutations and protein expression in resected non-small cell lung cancer. Hum Pathol 2014; 45:1162-8. [PMID: 24745618 DOI: 10.1016/j.humpath.2014.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 02/09/2023]
Abstract
The purpose of this study was to characterize the prevalence of insulin-like growth factor 1 receptor (IGF1R) mutations, single nucleotide polymorphisms (SNP), and protein overexpression in surgically resected non-small cell lung cancers in relation to patient characteristics and prognosis. This retrospective study was conducted on 304 patients with non-small cell lung cancers who underwent curative pulmonary resection (median follow-up for surviving patients, 3.6 years). IGF1R gene alterations (n = 304) and protein expression (n = 181) were evaluated by polymerase chain reaction-based assays and immunohistochemistry, respectively. Membranous and cytoplasmic staining were analyzed separately. In an exploratory analysis, 1 silent mutation in exon 16 and 3 mutations in introns of the IGF1R gene comprising the tyrosine kinase domain were detected. Moreover, evaluating selected IGF1R SNPs, patients with adenocarcinomas and homozygous for the rs8038415 T-allele had a significantly better survival (P = .025) but no different disease-free survival. Regarding expression, membranous but not cytoplasmic IGF1R staining was higher in squamous cell carcinomas versus other histologies (P < .0001) and showed a trend to longer survival (P = .08). No association between SNP variations and protein expression was found. Membranous IGF1R protein expression is higher in squamous cell versus other histologies but does not correlate with prognosis. SNPs and mutations can be detected and may harbor prognostic value. These alterations may be of interest when evaluating the IGF1R as potential therapeutic target and should receive further research.
Collapse
Affiliation(s)
- Niels Reinmuth
- Department of Thoracic Oncology, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany.
| | - Sebastian Kloos
- Department of Thoracic Oncology, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Arne Warth
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany; Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Angela Risch
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany; Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany; Department of Thoracic Surgery, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Hans Hoffmann
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany; Department of Thoracic Surgery, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany
| | - Michael Meister
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany; Department of Translational Research Unit, Thoraxklinik at the University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
53
|
Murakami A, Takahashi F, Nurwidya F, Kobayashi I, Minakata K, Hashimoto M, Nara T, Kato M, Tajima K, Shimada N, Iwakami SI, Moriyama M, Moriyama H, Koizumi F, Takahashi K. Hypoxia increases gefitinib-resistant lung cancer stem cells through the activation of insulin-like growth factor 1 receptor. PLoS One 2014; 9:e86459. [PMID: 24489728 PMCID: PMC3904884 DOI: 10.1371/journal.pone.0086459] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 12/13/2013] [Indexed: 01/10/2023] Open
Abstract
Accumulating evidence indicates that a small population of cancer stem cells (CSCs) is involved in intrinsic resistance to cancer treatment. The hypoxic microenvironment is an important stem cell niche that promotes the persistence of CSCs in tumors. Our aim here was to elucidate the role of hypoxia and CSCs in the resistance to gefitinib in non-small cell lung cancer (NSCLC) with activating epidermal growth factor receptor (EGFR) mutation. NSCLC cell lines, PC9 and HCC827, which express the EGFR exon 19 deletion mutations, were exposed to high concentration of gefitinib under normoxic or hypoxic conditions. Seven days after gefitinib exposure, a small fraction of viable cells were detected, and these were referred to as "gefitinib-resistant persisters" (GRPs). CD133, Oct4, Sox2, Nanog, CXCR4, and ALDH1A1-all genes involved in stemness-were highly expressed in GRPs in PC9 and HCC827 cells, and PC9 GRPs exhibited a high potential for tumorigenicity in vivo. The expression of insulin-like growth factor 1 (IGF1) was also upregulated and IGF1 receptor (IGF1R) was activated on GRPs. Importantly, hypoxic exposure significantly increased sphere formation, reflecting the self-renewal capability, and the population of CD133- and Oct4-positive GRPs. Additionally, hypoxia upregulated IGF1 expression through hypoxia-inducible factor 1α (HIF1α), and markedly promoted the activation of IGF1R on GRPs. Knockdown of IGF1 expression significantly reduced phosphorylated IGF1R-expressing GRPs under hypoxic conditions. Finally, inhibition of HIF1α or IGF1R by specific inhibitors significantly decreased the population of CD133- and Oct4-positive GRPs, which were increased by hypoxia in PC9 and HCC827 cells. Collectively, these findings suggest that hypoxia increased the population of lung CSCs resistant to gefitinib in EGFR mutation-positive NSCLC by activating IGF1R. Targeting the IGF1R pathway may be a promising strategy for overcoming gefitinib resistance in EGFR mutation-positive NSCLC induced by lung CSCs and microenvironment factors such as tumor hypoxia.
Collapse
MESH Headings
- AC133 Antigen
- Animals
- Antigens, CD/metabolism
- Carcinogenesis/drug effects
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Hypoxia/drug effects
- Cell Hypoxia/genetics
- Cell Line, Tumor
- Cell Separation
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- ErbB Receptors/genetics
- Gefitinib
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Knockdown Techniques
- Glycoproteins/metabolism
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice, Inbred NOD
- Mutation/genetics
- Neoplasm Transplantation
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Octamer Transcription Factor-3/metabolism
- Peptides/metabolism
- Quinazolines/pharmacology
- Quinazolines/therapeutic use
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/pathology
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Akiko Murakami
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- * E-mail:
| | - Fariz Nurwidya
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Isao Kobayashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Kunihiko Minakata
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Muneaki Hashimoto
- Department of Molecular and Cellular Parasitology, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Takeshi Nara
- Department of Molecular and Cellular Parasitology, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Motoyasu Kato
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Ken Tajima
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Naoko Shimada
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | | | - Mariko Moriyama
- Pharmaceutical Research and technology institute, Kinki University, School of Medicine, Osaka, Japan
| | - Hiroyuki Moriyama
- Pharmaceutical Research and technology institute, Kinki University, School of Medicine, Osaka, Japan
| | | | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
54
|
Janssen JAMJL, Varewijck AJ. IGF-IR Targeted Therapy: Past, Present and Future. Front Endocrinol (Lausanne) 2014; 5:224. [PMID: 25566194 PMCID: PMC4275034 DOI: 10.3389/fendo.2014.00224] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 12/08/2014] [Indexed: 01/01/2023] Open
Abstract
The IGF-I receptor (IGF-IR) has been studied as an anti-cancer target. However, monotherapy trials with IGF-IR targeted antibodies or with IGF-IR specific tyrosine kinase inhibitors have, overall, been very disappointing in the clinical setting. This review discusses potential reasons why IGF-I R targeted therapy fails to inhibit growth of human cancers. It has become clear that intracellular signaling pathways are highly interconnected and complex instead of being linear and simple. One of the most potent candidates for failure of IGF-IR targeted therapy is the insulin receptor isoform A (IR-A). Activation of the IR-A by insulin-like growth factor-II (IGF-II) bypasses the IGF-IR and its inhibition. Another factor may be that anti-cancer treatment may reduce IGF-IR expression. IGF-IR blocking drugs may also induce hyperglycemia and hyperinsulinemia, which may further stimulate cell growth. In addition, circulating IGF-IRs may reduce therapeutic effects of IGF-IR targeted therapy. Nevertheless, it is still possible that the IGF-IR may be a useful adjuvant or secondary target for the treatment of human cancers. Development of functional inhibitors that affect the IGF-IR and IR-A may be necessary to overcome resistance and to make IGF-IR targeted therapy successful. Drugs that modify alternative downstream effects of the IGF-IR, so called "biasing agonists," should also be considered.
Collapse
Affiliation(s)
- Joseph A. M. J. L. Janssen
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
- *Correspondence: Joseph A. M. J. L. Janssen, Department of Internal Medicine, Erasmus MC, Room D-443, ‘s-Gravendijkwal 230, Rotterdam 3015 CE, Netherlands e-mail:
| | - Aimee J. Varewijck
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
55
|
Kim JS, Kim ES, Liu D, Lee JJ, Solis L, Behrens C, Lippman SM, Hong WK, Wistuba II, Lee HY. Prognostic implications of tumoral expression of insulin like growth factors 1 and 2 in patients with non-small-cell lung cancer. Clin Lung Cancer 2014; 15:213-21. [PMID: 24485233 DOI: 10.1016/j.cllc.2013.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 12/15/2013] [Accepted: 12/23/2013] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The currently available systemic therapies for non-small-cell lung cancer (NSCLC) have limited efficacy. Previous studies indicated an association of elevated insulinlike growth factor (IGF)-1 receptor (IGF-1R) and insulin receptor expression levels with poor survival in patients with NSCLC. To better understand the molecular biomarkers involved in the IGF signaling pathway in NSCLC, the expression levels of IGF-1 and IGF-2 are characterized and evaluated for their association with IGF-1R and phosphorylated IGF-1R (pIGF-1R) expression in NSCLC. MATERIALS AND METHODS A total of 352 patients who underwent NSCLC resection with curative intent were studied. The expression patterns of the IGF-1, IGF-2, IGF-1R, and pIGF-1R proteins were assessed immunohistochemically using tissue microarrays. RESULTS The IGF-1 expression was higher in patients with adenocarcinoma (ADC) than in those with squamous cell carcinoma (SCC), whereas the IGF-2 score was higher in patients with SCC than those with ADC. Likewise, the IGF-1 score was higher in patients with mutated epidermal growth factor receptor (mtEGFR) than in those with wild type EGFR (wtEGFR), whereas the IGF-2 score was higher in patients with wtEGFR than in those with mtEGFR. Patients with low levels of IGF-1 expression had longer overall survival (OS) than those with high IGF-1 expression, and subgroup analyses found a significant difference in OS only in patients with ADC. CONCLUSION The overexpression of IGF-1 predicts poor survival among patients with NSCLC, especially those with ADC. These results might serve as a future guide for clinical trials involving IGF-1R-targeting agents.
Collapse
Affiliation(s)
- Jin-Soo Kim
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Deparment of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Edward S Kim
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Diane Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - J Jack Lee
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luisa Solis
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carmen Behrens
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Scott M Lippman
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Waun Ki Hong
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ignacio I Wistuba
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ho-Young Lee
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; College of Pharmacy, Seoul National University, Seoul, South Korea.
| |
Collapse
|
56
|
Reungwetwattana T, Dy GK. Targeted therapies in development for non-small cell lung cancer. J Carcinog 2013; 12:22. [PMID: 24574860 PMCID: PMC3927069 DOI: 10.4103/1477-3163.123972] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/15/2013] [Indexed: 12/11/2022] Open
Abstract
The iterative discovery in various malignancies during the past decades that a number of aberrant tumorigenic processes and signal transduction pathways are mediated by "druggable" protein kinases has led to a revolutionary change in drug development. In non-small cell lung cancer (NSCLC), the ErbB family of receptors (e.g., EGFR [epidermal growth factor receptor], HER2 [human epidermal growth factor receptor 2]), RAS (rat sarcoma gene), BRAF (v-raf murine sarcoma viral oncogene homolog B1), MAPK (mitogen-activated protein kinase) c-MET (c-mesenchymal-epithelial transition), FGFR (fibroblast growth factor receptor), DDR2 (discoidin domain receptor 2), PIK3CA (phosphatidylinositol-4,5-bisphosphate3-kinase, catalytic subunit alpha)), PTEN (phosphatase and tensin homolog), AKT (protein kinase B), ALK (anaplastic lym phoma kinase), RET (rearranged during transfection), ROS1 (reactive oxygen species 1) and EPH (erythropoietin-producing hepatoma) are key targets of various agents currently in clinical development. These oncogenic targets exert their selective growth advantage through various intercommunicating pathways, such as through RAS/RAF/MEK, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin and SRC-signal transduction and transcription signaling. The recent clinical studies, EGFR tyrosine kinase inhibitors and crizotinib were considered as strongly effective targeted therapies in metastatic NSCLC. Currently, five molecular targeted agents were approved for treatment of advanced NSCLC: Gefitinib, erlotinib and afatinib for positive EGFR mutation, crizotinib for positive echinoderm microtubule-associated protein-like 4 (EML4)-ALK translocation and bevacizumab. Moreover, oncogenic mutant proteins are subject to regulation by protein trafficking pathways, specifically through the heat shock protein 90 system. Drug combinations affecting various nodes in these signaling and intracellular processes are predicted and demonstrated to be synergistic and advantageous in overcoming treatment resistance compared with monotherapy approaches. Understanding the role of the tumor microenvironment in the development and maintenance of the malignant phenotype provided additional therapeutic approaches as well. More recently, improved knowledge on tumor immunology has set the stage for promising immunotherapies in NSCLC. This review will focus on the rationale for the development of targeted therapies in NSCLC and the various strategies employed in preventing or overcoming the inevitable occurrence of treatment resistance.
Collapse
Affiliation(s)
- Thanyanan Reungwetwattana
- Department of Internal Medicine, Division of Medical Oncology, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Grace Kho Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
57
|
Berge EM, Doebele RC. Targeted therapies in non-small cell lung cancer: emerging oncogene targets following the success of epidermal growth factor receptor. Semin Oncol 2013; 41:110-25. [PMID: 24565585 DOI: 10.1053/j.seminoncol.2013.12.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The diagnostic testing, treatment and prognosis of non-small cell lung cancer (NSCLC) has undergone a paradigm shift since the discovery of sensitizing mutations in the epidermal growth factor receptor (EGFR) gene in a subset of NSCLC patients. Several additional oncogenic mutations, including gene fusions and amplifications, have since been discovered, with a number of drugs that target each specific oncogene. This review focuses on oncogenes in NSCLC other than EGFR and their companion "targeted therapies." Particular emphasis is placed on the role of ALK, ROS1, RET, MET, BRAF, and HER2 in NSCLC.
Collapse
Affiliation(s)
- Eamon M Berge
- Department of Medicine, Division of Medical Oncology, University of Colorado, Aurora, CO.
| | - Robert C Doebele
- Department of Medicine, Division of Medical Oncology, University of Colorado, Aurora, CO
| |
Collapse
|
58
|
Shin DH, Lee HJ, Min HY, Choi SP, Lee MS, Lee JW, Johnson FM, Mehta K, Lippman SM, Glisson BS, Lee HY. Combating resistance to anti-IGFR antibody by targeting the integrin β3-Src pathway. J Natl Cancer Inst 2013; 105:1558-70. [PMID: 24092920 PMCID: PMC3797025 DOI: 10.1093/jnci/djt263] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/20/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Several phase II/III trials of anti-insulin-like growth factor 1 receptor (IGF-1R) monoclonal antibodies (mAbs) have shown limited efficacy. The mechanisms of resistance to IGF-1R mAb-based therapies and clinically applicable strategies for overcoming drug resistance are still undefined. METHODS IGF-1R mAb cixutumumab efficacy, alone or in combination with Src inhibitors, was evaluated in 10 human head and neck squamous cell carcinoma (HNSCC) and six non-small cell lung cancer (NSCLC) cell lines in vitro in two- or three-dimensional culture systems and in vivo in cell line- or patient-derived xenograft tumors in athymic nude mice (n = 6-9 per group). Cixutumumab-induced changes in cell signaling and IGF-1 binding to integrin β3 were determined by Western or ligand blotting, immunoprecipitation, immunofluorescence, and cell adhesion analyses and enzyme-linked immunosorbent assay. Data were analyzed by the two-sided Student t test or one-way analysis of variance. RESULTS Integrin β3-Src signaling cascade was activated by IGF-1 in HNSCC and NSCLC cells, when IGF-1 binding to IGF-1R was hampered by cixutumumab, resulting in Akt activation and cixutumumab resistance. Targeting integrin β3 or Src enhanced antitumor activity of cixutumumab in multiple cixutumumab-resistant cell lines and patient-derived tumors in vitro and in vivo. Mean tumor volume of mice cotreated with cixutumumab and integrin β3 siRNA was 133.7 mm(3) (95% confidence interval [CI] = 57.6 to 209.8 mm(3)) compared with those treated with cixutumumab (1472.5 mm(3); 95% CI = 1150.7 to 1794.3 mm(3); P < .001) or integrin β3 siRNA (903.2 mm(3); 95% CI = 636.1 to 1170.3 mm(3); P < .001) alone. CONCLUSIONS Increased Src activation through integrin ανβ3 confers considerable resistance against anti-IGF-1R mAb-based therapies in HNSCC and NSCLC cells. Dual targeting of the IGF-1R pathway and collateral integrin β3-Src signaling module may override this resistance.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/pharmacology
- Blotting, Western
- CSK Tyrosine-Protein Kinase
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Drug Resistance, Neoplasm/drug effects
- Enzyme-Linked Immunosorbent Assay
- Fluorescent Antibody Technique
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/metabolism
- Humans
- Immunoprecipitation
- Integrin beta3/pharmacology
- Lung Neoplasms/drug therapy
- Mice
- Mice, Nude
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, IGF Type 1/immunology
- Signal Transduction/drug effects
- Squamous Cell Carcinoma of Head and Neck
- Xenograft Model Antitumor Assays
- src-Family Kinases/antagonists & inhibitors
Collapse
Affiliation(s)
- Dong Hoon Shin
- Affiliations of authors: Department of Thoracic/Head and Neck Medical Oncology (DHS, SPC, FMJ, SML, BSG) and Department of Experimental Therapeutics (KM), The University of Texas MD Anderson Cancer Center, Houston, TX; College of Pharmacy, Inje University, Gimhae, Gyungnam, Republic of Korea (H-JL); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (H-YM, M-SL, JWL, H-YL)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Conti E, Romiti A, Musumeci MB, Passerini J, Zezza L, Mastromarino V, D'Antonio C, Marchetti P, Paneni F, Autore C, Volpe M. Arterial thrombotic events and acute coronary syndromes with cancer drugs: Are growth factors the missed link? Int J Cardiol 2013; 167:2421-9. [DOI: 10.1016/j.ijcard.2013.01.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
|
60
|
Lee SY, Kang HG, Yoo SS, Kang YR, Choi YY, Lee WK, Choi JE, Jeon HS, Shin KM, Oh IJ, Kim KS, Lee J, Cha SI, Kim CH, Kim YC, Park JY. Polymorphisms in DNA repair and apoptosis-related genes and clinical outcomes of patients with non-small cell lung cancer treated with first-line paclitaxel-cisplatin chemotherapy. Lung Cancer 2013; 82:330-9. [PMID: 23973201 DOI: 10.1016/j.lungcan.2013.07.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 07/19/2013] [Accepted: 07/26/2013] [Indexed: 10/26/2022]
Abstract
This study was conducted to analyze a comprehensive panel of single nucleotide polymorphisms (SNPs) in genes in DNA repair and apoptosis pathways and determine the relationship between polymorphisms and treatment outcomes of patients with non-small cell lung cancer (NSCLC) treated with first-line paclitaxel-cisplatin chemotherapy. Three hundred eighty two patients with NSCLC were enrolled. Seventy-four SNPs in 48 genes (42 SNPs in 27 DNA repair pathway genes and 32 SNPs in 21 apoptotic pathway genes) were genotyped and their associations with chemotherapy response and overall survival (OS) were analyzed. Among SNPs in DNA repair genes, BRCA1 rs799917 was significantly associated with both chemotherapy response and OS. XRCC1 rs25487 exhibited a significant association with chemotherapy response and ERCC2 rs1052555 with OS. Four SNPs in apoptotic genes (TNFRSF1B rs1061624, BCL2 rs2279115, BIRC5 rs9904341, and CASP8 rs3769818) were significantly associated with OS, but not with response to chemotherapy. When the six SNPs which were associated with OS in individual analysis were combined, OS decreased as the number of bad genotypes increased (P(trend) = 2 × 10(-6)). Patients with 3, and 4-6 bad genotypes had significantly worse OS compared with those carrying 0-2 bad genotypes (adjusted hazard ratio [aHR] = 1.54, 95% CI = 1.14-2.08, P = 0.005; aHR = 2.10, 95% CI = 1.55-2.85, P = 2 × 10(-6), respectively). In conclusion, these findings suggest that the six SNPs identified, particularly their combined genotypes, could be used as biomarkers predicting chemotherapy response and survival of NSCLC patients treated with first-line paclitaxel-cisplatin chemotherapy.
Collapse
Affiliation(s)
- Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Chen B, Xiao F, Li B, Xie B, Zhou J, Zheng J, Zhang W. The Role of Epithelial–Mesenchymal Transition and IGF-1R Expression in Prediction of Gefitinib Activity as the Second-Line Treatment for Advanced Nonsmall-Cell Lung Cancer. Cancer Invest 2013; 31:454-60. [DOI: 10.3109/07357907.2013.820315] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
62
|
Liu YJ, Qiang W, Shi J, Lv SQ, Ji MJ, Shi BY. Expression and significance of IGF-1 and IGF-1R in thyroid nodules. Endocrine 2013; 44:158-64. [PMID: 23288662 DOI: 10.1007/s12020-012-9864-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 12/14/2012] [Indexed: 10/27/2022]
Abstract
There are limitations in the diagnosis of thyroid nodules, especially follicular lesions, and their pathogenesis remains unclear. Insulin-like growth factor-1 (IGF-1) has been implicated in tumor cell apoptosis, transformation, invasion, and metastasis; however, its role in thyroid nodules is undetermined. The aim of this study is to investigate the relationship between expression of IGF-1 and thyroid nodule, and evaluate the role of IGF-1 in differential diagnosis and pathogenetic function of benign and malignant thyroid nodules. Sixty-two paraffin-embedded thyroid tissues from patients with thyroid nodules were selected, including 18 follicular adenomas (FA), 17 nodular goiters, 13 papillary thyroid carcinomas (PTC), 2 follicular thyroid carcinomas, and 12 normal controls. IGF-1 and IGF-1R protein and mRNA expression was detected by immunohistochemistry (IHC) and real-time quantitative PCR (qRT-PCR), respectively. Grouping comparisons were employed among the above groups based on the clinical and pathological subtypes. IGF-1 and IGF-1R expression was significantly higher in FA, nodular goiters, and PTC than that in the controls (all P < 0.01). Similarly, IGF-1 mRNA expression was also significantly higher in FA (P < 0.05), nodular goiters (P < 0.01), and PTC (P < 0.01) compared with the controls. IGF-1R mRNA expression was also significantly higher in FA, nodular goiters, and PTC (all P < 0.01) compared with the controls. Compared with FA and nodular goiters, PTC showed significantly higher IGF-1 and IGF-1R protein (P < 0.01, P < 0.05, respectively) and mRNA expression (P < 0.01, P < 0.05, respectively). IGF-1 probably plays an important role in the genesis and development of certain solid cold thyroid nodules, including PTC, nodular goiters, and FA. Detection of IGF-1 and IGF-1R expression in thyroid tissues by IHC or qRT-PCR is hard to distinguish malignant from benign lesions.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/diagnosis
- Adenocarcinoma, Follicular/genetics
- Adenocarcinoma, Follicular/metabolism
- Adult
- Aged
- Aged, 80 and over
- Carcinoma/diagnosis
- Carcinoma/genetics
- Carcinoma/metabolism
- Carcinoma, Papillary
- Female
- Gene Expression
- Goiter, Nodular/diagnosis
- Goiter, Nodular/genetics
- Goiter, Nodular/metabolism
- Humans
- Immunohistochemistry
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Male
- Middle Aged
- Prognosis
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Thyroid Cancer, Papillary
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Thyroid Nodule/diagnosis
- Thyroid Nodule/metabolism
Collapse
Affiliation(s)
- Ying-Jian Liu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Shaanxi Province, Xi'an, 710061, China
| | | | | | | | | | | |
Collapse
|
63
|
Vincent EE, Elder DJE, Curwen J, Kilgour E, Hers I, Tavaré JM. Targeting non-small cell lung cancer cells by dual inhibition of the insulin receptor and the insulin-like growth factor-1 receptor. PLoS One 2013; 8:e66963. [PMID: 23826179 PMCID: PMC3691253 DOI: 10.1371/journal.pone.0066963] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/14/2013] [Indexed: 11/18/2022] Open
Abstract
Phase III trials of the anti-insulin-like growth factor-1 receptor (IGF1R) antibody figitumumab in non-small cell lung cancer (NSCLC) patients have been discontinued owing to lack of survival benefit. We investigated whether inhibition of the highly homologous insulin receptor (IR) in addition to the IGF1R would be more effective than inhibition of the IGF1R alone at preventing the proliferation of NSCLC cells. Signalling through IGF1R and IR in the NSCLC cell lines A549 and Hcc193 was stimulated by a combination of IGF1, IGF2 and insulin. It was inhibited by antibodies that block ligand binding, αIR3 (IGF1R) and IR47-9 (IR), and by the ATP-competitive small molecule tyrosine kinase inhibitors AZ12253801 and NVPAWD742 which inhibit both IGF1R and IR tyrosine kinases. The effect of inhibitors was determined by an anchorage-independent proliferation assay and by analysis of Akt phosphorylation. In Hcc193 cells the reduction in cell proliferation and Akt phosphorylation due to anti-IGF1R antibody was enhanced by antibody-mediated inhibition of the IR whereas in A549 cells, with a relatively low IR:IGF1R expression ratio, it was not. In each cell line proliferation and Akt phosphorylation were more effectively inhibited by AZ12253801 and NVPAWD742 than by combined αIR3 and IR47-9. When the IGF1R alone is inhibited, unencumbered signalling through the IR can contribute to continued NSCLC cell proliferation. We conclude that small molecule inhibitors targeting both the IR and IGF1R more effectively reduce NSCLC cell proliferation in a manner independent of the IR:IGF1R expression ratio, providing a therapeutic rationale for the treatment of this disease.
Collapse
Affiliation(s)
- Emma E. Vincent
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Douglas J. E. Elder
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Jon Curwen
- CIRA Discovery, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Elaine Kilgour
- CIRA Discovery, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Ingeborg Hers
- School of Physiology and Pharmacology, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Jeremy M. Tavaré
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
64
|
Li F, Zhao C, Wang L. Molecular-targeted agents combination therapy for cancer: developments and potentials. Int J Cancer 2013; 134:1257-69. [PMID: 23649791 DOI: 10.1002/ijc.28261] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 04/25/2013] [Indexed: 11/10/2022]
Abstract
Although chemotherapy has advanced into the era of targeted drugs, the antitumor efficacies of current therapies are limited, most likely because of the high degree of cancer clonal heterogeneity, intratumor genetic heterogeneity and cell signal complexity. As shutdown of a single target does not necessarily eradicate the cancer, the use of combinations of molecular-targeted agents (MATs) has been proposed, and some pioneering research has been conducted to examine the efficacy of this strategy. In this article, the clinical and preclinical studies that are underway in an attempt to improve the anticancer efficacy of chemotherapies through combination strategies are summarized. Studies of combining cytotoxic agents with MATs, coinhibiting two or more targets in a single pathway or coinhibiting parallel or compensatory pathways as well as specific combinations will be introduced, and the antitumor potentials of each combination strategy will be evaluated.
Collapse
Affiliation(s)
- Feifei Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China; Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, China; Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | | | | |
Collapse
|
65
|
Tang H, Liao Y, Xu L, Zhang C, Liu Z, Deng Y, Jiang Z, Fu S, Chen Z, Zhou S. Estrogen and insulin-like growth factor 1 synergistically promote the development of lung adenocarcinoma in mice. Int J Cancer 2013; 133:2473-82. [PMID: 23649836 DOI: 10.1002/ijc.28262] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 04/23/2013] [Indexed: 01/30/2023]
Abstract
Estrogen receptor (ER) and insulin-like growth factor-1 receptor (IGF-1R) signaling are implicated in lung cancer progression. Based on their previous findings, the authors sought to investigate whether estrogen and IGF-1 act synergistically to promote lung adenocarcinoma (LADE) development in mice. LADE was induced with urethane in ovariectomized Kunming mice. Tumor-bearing mice were divided into seven groups: 17β-estradiol (E2), E2+fulvestrant (Ful; estrogen inhibitor), IGF-1, IGF-1+AG1024 (IGF-1 inhibitor), E2+IGF-1, E2+IGF-1+Ful+AG1024 and control groups. After 14 weeks, the mice were sacrificed, and then the tumor growth was determined. The expression of ERα/ERβ, IGF-1, IGF-1R and Ki67 was examined using tissue-microarray-immunohistochemistry, and IGF-1, p-ERβ, p-IGF-1R, p-MAPK and p-AKT levels were determined based on Western blot analysis. Fluorescence-quantitative polymerase chain reaction was used to detect the mRNA expression of ERβ, ERβ2 and IGF-1R. Tumors were found in 93.88% (46/49) of urethane-treated mice, and pathologically proven LADE was noted in 75.51% (37/49). In the E2+IGF-1 group, tumor growth was significantly higher than in the E2 group (p < 0.05), the IGF-1 group (p < 0.05) and control group (p < 0.05). Similarly, the expression of ERβ, p-ERβ, ERβ2, IGF-1, IGF-1R, p-IGF-1R, p-MAPK, p-AKT and Ki67 at the protein and/or mRNA levels was markedly higher in the ligand group than in the ligand + inhibitor groups (all p < 0.05). This study demonstrated for the first time that estrogen and IGF-1 act to synergistically promote the development of LADE in mice, and this may be related to the activation of the MAPK and AKT signaling pathways in which ERβ1, ERβ2 and IGF-1R play important roles.
Collapse
Affiliation(s)
- Hexiao Tang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Tsuta K, Mimae T, Nitta H, Yoshida A, Maeshima AM, Asamura H, Grogan TM, Furuta K, Tsuda H. Insulin-like growth factor-1 receptor protein expression and gene copy number alterations in non–small cell lung carcinomas. Hum Pathol 2013; 44:975-82. [DOI: 10.1016/j.humpath.2012.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/22/2012] [Accepted: 09/05/2012] [Indexed: 11/16/2022]
|
67
|
Chen XH, Li ZQ, Peng H, Jin SM, Fu HQ, Zhu TC, Weng XG. Type 1 insulin-like growth factor receptor monoclonal antibody (HX-1162) treatment for liver cancer. Onco Targets Ther 2013; 6:527-30. [PMID: 23700371 PMCID: PMC3660154 DOI: 10.2147/ott.s44162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
One of the most important molecules mediating the proliferation, growth, and metastasis of cancer cells is insulin-like growth factor (IGF), with its receptor IGF-1R. Here, we describe the potential of an IGF-1R monoclonal antibody, HX-1162, on liver cancer apoptosis in vitro and in vivo. We found that HX-1162 could induce the apoptosis of cultured liver cancer cells. Additionally, HX-1162 treatment inhibited the tumor growth after cancer cell grafting and enhanced the cell apoptosis inside the tumor tissue. We conclude that IGF-1R targeting therapy provides a new avenue toward treating liver cancer.
Collapse
Affiliation(s)
- Xue-Hui Chen
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
68
|
Chen HX, Sharon E. IGF-1R as an anti-cancer target--trials and tribulations. CHINESE JOURNAL OF CANCER 2013; 32:242-52. [PMID: 23601239 PMCID: PMC3845553 DOI: 10.5732/cjc.012.10263] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/19/2013] [Accepted: 03/15/2013] [Indexed: 12/23/2022]
Abstract
Type I insulin-like growth factor receptor (IGF-1R) has long been recognized for its role in tumorigenesis and growth, but only recently have the tools for targeting the IGF pathway become available. More than 10 IGF/IGF-1R inhibitors have entered clinical trials, and these belong to three main classes: (1) monoclonal antibodies against IGF-1R, (2) monoclonal antibodies against IGF-1R ligands (IGF-1 and IGF-2), and (3) IGF-1R tyrosine kinase inhibitors. These IGF-1R-targeting agents share common effects on IGF-1R signaling but differ in mechanisms of action, spectrum of target inhibition, and pharmacological features. Clinical activity of IGF-1R inhibitors has been demonstrated with sustained responses in a small number of patients with select tumor types, such as Ewing sarcoma and thymoma. However, many large clinical trials involving patients with adult tumors, including non-small cell lung cancer, breast cancer, and pancreatic cancer, failed to show clinical benefit in the overall patient population. Possible reasons for failure include the complexity of the IGF-1R/insulin receptor system and parallel growth and survival pathways, as well as a lack of patient selection markers. While IGF-1R remains a valid target for selected tumor types, identification of predictive markers and rational combinations will be critical to success in future development.
Collapse
Affiliation(s)
- Helen X Chen
- National Cancer Institute, Bethesda, MD 20892, USA.
| | | |
Collapse
|
69
|
Park MY, Kim DR, Eo EY, Lim HJ, Park JS, Cho YJ, Yoon HI, Lee JH, Lee CT. Genetic blockade of insulin-like growth factor-1 receptor via recombinant adenovirus in lung cancer can be enhanced by the histone deacetylase inhibitor, vorinostat. J Gene Med 2013; 15:115-22. [DOI: 10.1002/jgm.2699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 12/24/2012] [Accepted: 01/29/2013] [Indexed: 11/09/2022] Open
Affiliation(s)
- Mi-Young Park
- Department of Internal Medicine, Respiratory Center; Seoul National University Bundang Hospital; Seongnam; Korea
| | - Dal Rae Kim
- Department of Internal Medicine, Respiratory Center; Seoul National University Bundang Hospital; Seongnam; Korea
| | - Eun Young Eo
- Department of Internal Medicine, Respiratory Center; Seoul National University Bundang Hospital; Seongnam; Korea
| | | | | | | | | | | | | |
Collapse
|
70
|
Jin M, Buck E, Mulvihill MJ. Modulation of insulin-like growth factor-1 receptor and its signaling network for the treatment of cancer: current status and future perspectives. Oncol Rev 2013; 7:e3. [PMID: 25992224 PMCID: PMC4419619 DOI: 10.4081/oncol.2013.e3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/05/2013] [Accepted: 04/15/2013] [Indexed: 12/20/2022] Open
Abstract
Based on over three decades of pre-clinical data, insulin-like growth factor-1 receptor (IGF-1R) signaling has gained recognition as a promoter of tumorogenesis, driving cell survival and proliferation in multiple human cancers. As a result, IGF-1R has been pursued as a target for cancer treatment. Early pioneering efforts targeting IGF-1R focused on highly selective monoclonal antibodies, with multiple agents advancing to clinical trials. However, despite some initial promising results, recent clinical disclosures have been less encouraging. Moreover, recent studies have revealed that IGF-1R participates in a dynamic and complex signaling network, interacting with additional targets and pathways thereof through various crosstalk and compensatory signaling mechanisms. Such mechanisms of bypass signaling help to shed some light on the decreased effectiveness of selective IGF-1R targeted therapies (e.g. monoclonal antibodies) and suggest that targeting multiple nodes within this signaling network might be necessary to produce a more effective therapeutic response. Additionally, such findings have led to the development of small molecule IGF-1R inhibitors which also co-inhibit additional targets such as insulin receptor and epidermal growth factor receptor. Such findings have helped to guide the design rationale of numerous drug combinations that are currently being evaluated in clinical trials.
Collapse
|
71
|
Sechler M, Cizmic AD, Avasarala S, Van Scoyk M, Brzezinski C, Kelley N, Bikkavilli RK, Winn RA. Non-small-cell lung cancer: molecular targeted therapy and personalized medicine - drug resistance, mechanisms, and strategies. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2013; 6:25-36. [PMID: 23690695 PMCID: PMC3656464 DOI: 10.2147/pgpm.s26058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Targeted therapies for cancer bring the hope of specific treatment, providing high efficacy and in some cases lower toxicity than conventional treatment. Although targeted therapeutics have helped immensely in the treatment of several cancers, like chronic myelogenous leukemia, colon cancer, and breast cancer, the benefit of these agents in the treatment of lung cancer remains limited, in part due to the development of drug resistance. In this review, we discuss the mechanisms of drug resistance and the current strategies used to treat lung cancer. A better understanding of these drug-resistance mechanisms could potentially benefit from the development of a more robust personalized medicine approach for the treatment of lung cancer.
Collapse
Affiliation(s)
- Marybeth Sechler
- Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, CO, USA ; Program in Cancer Biology, University of Colorado, Aurora, CO, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Kim HS, Mitsudomi T, Soo RA, Cho BC. Personalized therapy on the horizon for squamous cell carcinoma of the lung. Lung Cancer 2013; 80:249-55. [PMID: 23489560 DOI: 10.1016/j.lungcan.2013.02.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 02/15/2013] [Accepted: 02/16/2013] [Indexed: 12/28/2022]
Abstract
Squamous cell carcinoma (SQCC) of the lung is the second-largest subtype of non-small cell lung cancer (NSCLC), causing an estimated 400,000 deaths per year worldwide. Recent developments in cancer genome sequencing technology expanded our knowledge of driver mutations, which were identified as novel candidates for targeted therapy in various cancers. Successful targeted treatments for lung adenocarcinoma, NSCLC's primary subtype, with EGFR mutation or ALK fusion are clinically available, and a clinical trial of personalized targeted therapy in patients with lung adenocarcinoma is underway by the Lung Cancer Mutation Consortium. Although there are targeted treatments for lung adenocarcinoma, no personalized therapies currently exist for SQCC. Recently, comprehensive genomic characterization of lung SQCC using massively parallel sequencing has enabled us to identify several potential driver mutations/signaling pathways. These are FGFR1 amplifications, PI3KCA mutations, PTEN mutations/deletions, PDGFRA amplifications/mutations, and DDR2 mutations. The march toward personalized therapy may have taken a step forward with the discovery of these potential biomarkers for the treatment of SQCC of the lung. This article reviewed the current knowledge of genomic landscape of lung SQCC and summarized ongoing clinical trials of targeted agents for lung SQCC. Also, we will suggest several other actionable mutations with matching drugs that should be investigated in future clinical trials for the personalized treatment of lung SQCC.
Collapse
Affiliation(s)
- Han Sang Kim
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
73
|
Wang N, Lu Y, Pinard M, Pilotte A, Gilbert R, Massie B, Brodt P. Sustained production of a soluble IGF-I receptor by gutless adenovirus-transduced host cells protects from tumor growth in the liver. Cancer Gene Ther 2013; 20:229-36. [PMID: 23470563 DOI: 10.1038/cgt.2013.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The IGF-I receptor (IGF-IR) has an important role in malignant disease and is the target of several drugs presently in clinical trials. Gene therapy has been explored as cancer treatment, mainly for delivery of genes that induce cell death or enhance the immunological response to cancer. Previously, we have shown that the implantation of autologous bone-marrow stromal cells producing a soluble form of IGF-IR (sIGFIR) inhibited experimental liver metastasis of several tumor types in mice. Here, we evaluated the utility of adenovirus-based gene delivery for generating therapeutically effective plasma levels of this decoy. We constructed a third generation gutless adenovirus expressing sIGFIR and found that HEK-293 cells transduced by this, but not control adenoviruses, secreted soluble receptor protein that blocked IGF-I-induced tumor cell migration, proliferation and survival in vitro. Following virus injection in vivo, viral DNA was detectable by PCR in several host organs, particularly the liver, and this resulted in the production of measurable sIGFIR plasma levels for up to 21 days post injection. In mice producing virus-encoded sIGFIR, experimental liver metastasis was inhibited, indicating that sIGFIR levels were therapeutically effective. The results show that adenovirus-based delivery of inhibitory soluble proteins can provide an effective anticancer strategy.
Collapse
Affiliation(s)
- N Wang
- Department of Surgery, McGill University Health Center, Royal Victoria Hospital, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
74
|
Macaulay VM, Middleton MR, Protheroe AS, Tolcher A, Dieras V, Sessa C, Bahleda R, Blay JY, LoRusso P, Mery-Mignard D, Soria JC. Phase I study of humanized monoclonal antibody AVE1642 directed against the type 1 insulin-like growth factor receptor (IGF-1R), administered in combination with anticancer therapies to patients with advanced solid tumors. Ann Oncol 2013; 24:784-91. [PMID: 23104723 PMCID: PMC3574548 DOI: 10.1093/annonc/mds511] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 07/23/2012] [Accepted: 08/27/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Type 1 insulin-like growth factor receptor (IGF-1R) mediates resistance to chemotherapy and targeted agents. This study assessed the safety, pharmacokinetics (PK), and tolerability of humanized IGF-1R antibody AVE1642 with other cancer treatments. PATIENTS Patients with advanced solid tumors received three weekly AVE1642 dosed at 6 mg/kg, chosen following previous study, with 75 (cohort A) or 100 mg/m(2) (B) docetaxel, 1250 mg/m(2) gemcitabine/100 mg erlotinib (C1), or 60 mg/m(2) doxorubicin (D1). Blood samples were assayed for PK, IGFs, and IGF-BP3. RESULTS Fifty-eight patients received 317 AVE1642 infusions. The commonest adverse events were diarrhea (37/58 patients), asthenia (34/58), nausea (30/58), and stomatitis (21/58). Dose-limiting toxic effects in cohorts C1 (diarrhea) and D1 (neutropenia) prompted addition of cohorts C2 (1000 mg/m(2) gemcitabine/75 mg erlotinib) and D2 (50 mg/m(2) doxorubicin). Grade 3-4 hyperglycemia (three cases) accompanied steroid premedication for docetaxel administration. No PK interactions were detected. There were three partial responses in cohorts B (melanoma) and C (leiomyosarcoma, two cases) and 22 stabilizations ≥12 weeks, giving a control rate of 25/57 (44%). On treatment IGF-II rose by 68 ± 25 ng/ml in patients discontinuing treatment <12 weeks, and fell by 55.5 ± 21 ng/ml with disease control (P < 0.001). CONCLUSION AVE1642 was tolerable with 75-100 mg/m(2) docetaxel and 1000 mg/m(2) gemcitabine/75 mg erlotinib, achieving durable disease control in 44%, with an association between IGF-II and response.
Collapse
Affiliation(s)
- V M Macaulay
- Department of Oncology, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford OX3 7LE, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Ludovini V, Flacco A, Bianconi F, Ragusa M, Vannucci J, Bellezza G, Chiari R, Minotti V, Pistola L, Tofanetti FR, Siggillino A, Baldelli E, Sidoni A, Daddi N, Puma F, Varella-Garcia M, Crinò L. Concomitant high gene copy number and protein overexpression of IGF1R and EGFR negatively affect disease-free survival of surgically resected non-small-cell-lung cancer patients. Cancer Chemother Pharmacol 2013; 71:671-80. [PMID: 23314677 PMCID: PMC3963139 DOI: 10.1007/s00280-012-2056-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/12/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND Insulin-like growth factor 1 receptor (IGF1R) represents a novel molecular target in non-small-cell-lung cancer (NSCLC). IGF1R and epidermal growth factor receptor (EGFR) activation are essential to mediate tumor cell survival, proliferation, and invasion. This study investigates the prognostic role of IGF1R and EGFR in surgically resected NSCLC. MATERIALS AND METHODS IGF1R and EGFR copy number gain (CNG) were tested by fluorescence in situ hybridization (FISH) and protein expression by immunohistochemistry (IHC) in 125 stage I-II-IIIA NSCLC patients. RESULTS Fourty-six tumors (40.3%) were IGF1R FISH-positive (FISH+), and 76 (67.2%) were EGFR FISH+. Tumors with concomitant IGF1R/EGFR FISH+ were observed in 34 cases (30.1%). IGF1R and EGFR FISH+ were associated with SCC histology (p = 0.01 and p = 0.04, respectively). IGF1R and EGFR protein over-expression (IHC+) were detected in 45 (36.0%) and 69 (55.2%) cases, respectively. Tumors with concomitant IGF1R/EGFR IHC+ were detected in 31 (24.8%) patients. IGF1R/EGFR FISH+ and IGF1R/EGFR IHC+ were significantly associated (χ(2) = 4.02, p = 0.04). Patients with IGF1R/EGFR FISH+ and IGF1R/EGFR IHC+ were associated with shorter disease-free survival (DFS) (p = 0.05 and p = 0.05, respectively). Patients with concomitant IGF1R/EGFR FISH+/IHC+ had a worse DFS and overall survival (p = 0.005 and p = 0.01, respectively). The multivariate model confirmed that IGF1R/EGFR FISH+/IHC+ (hazard ratio (HR), 4.08; p = 0.01) and tumor stage (II-III vs I) (HR, 4.77; p = 0.003) were significantly associated with worse DFS. CONCLUSIONS IGF1R/EGFR FISH+ correlates with IGF1R/EGFR IHC+. IGF1R/EGFR FISH+/IHC+ is an independent negative prognostic factor for DFS in early NSCLC. These features may have important implications for future anti-IGF1R therapeutic approaches.
Collapse
Affiliation(s)
- V Ludovini
- Department of Medical Oncology, S. Maria Della Misericordia Hospital, 1, G. Dottori Street, 06132 Perugia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
A dose finding, safety and pharmacokinetic study of AVE1642, an anti-insulin-like growth factor-1 receptor (IGF-1R/CD221) monoclonal antibody, administered as a single agent and in combination with docetaxel in patients with advanced solid tumours. Eur J Cancer 2013; 49:1799-807. [PMID: 23485230 DOI: 10.1016/j.ejca.2013.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 12/31/2012] [Accepted: 01/06/2013] [Indexed: 01/13/2023]
Abstract
PURPOSE AVE1642, a humanised mAb, binds the human IGF-1R specifically and with high affinity. This study aimed to select the dose of AVE1642 alone and then combined with docetaxel 75mg/m(2) (D). MATERIAL AND METHODS AVE1642 was administered alone at cycle (cy) 1 and then combined with D from cy2, q3w. RESULTS A total of 27 patients received a median number of 5 cy (range, 1-10). The most common tumour types were sarcoma (18.5%), osseous tumours (11.1%) and colon cancer (11.1%). Two DLTs were reported in cy1 at dose level (DL) 18mg/kg and dose escalation was stopped. No major safety issue was observed. No anti-drug antibodies were detected. The Maximal Tolerated Dose of AVE1642 was 12mg/kg. The dose selected for further combinations is 6mg/kg, based on PK/PD data. Three objective responses, (two in sarcoma and one breast cancer) were observed but only one was confirmed. Eleven patients appeared to benefit from treatment with prolonged disease stabilisation ⩾4months. CONCLUSION AVE1642 is well tolerated as a single agent and combined with D. The selected dose of AVE1642 combined with D is 6mg/kg. Promising activity was seen in sarcoma and breast cancer patients.
Collapse
|
77
|
Identification of driver mutations in lung cancer: first step in personalized cancer. Target Oncol 2013; 8:3-14. [PMID: 23371030 DOI: 10.1007/s11523-013-0263-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/16/2013] [Indexed: 12/20/2022]
Abstract
Non-small cell lung cancer (NSCLC) has recently been associated with interesting molecular characteristics that have important implications in carcinogenesis and response to targeted therapies. Targeted therapies, if given to a patient subpopulation enriched by the presence of relevant molecular targets, can often abrogate cell signaling that perpetuates cancer progression. For instance, several molecular alterations have been defined as "driver mutations," such as mutations in EGFR and EML4-ALK fusion gene. Other key signaling pathways have also been identified as novel targets for lung cancer treatment. These first steps towards personalized medicine represent a shift in the management of NSCLC. Indeed, NSCLC should no longer be viewed as one common generic tumor but rather as a collection of more rare diseases with different biological behaviors and different sensitivities to targeted treatments. We are now clearly entering an era of personalized medicine for NSCLC cancers, and the development of molecular profiling technologies to assess DNA provides the potential to tailored medical care.
Collapse
|
78
|
Pillai RN, Ramalingam SS. Inhibition of insulin-like growth factor receptor: end of a targeted therapy? Transl Lung Cancer Res 2013; 2:14-22. [PMID: 25806201 PMCID: PMC4367644 DOI: 10.3978/j.issn.2218-6751.2012.11.05] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 11/08/2012] [Indexed: 11/14/2022]
Abstract
The Insulin-like Growth Factor 1 (IGF-1) signaling pathway activates several downstream signals important to lung cancer development and survival. IGF-1R activation has been linked to cancer risk in epidemiological studies and tumorigenesis in preclinical models. Several inhibitors of the insulin-like growth factor 1 receptor (IGF-1R) have been tested in clinical trials. Despite promising data in early phase studies, most studies of IGF-1R antagonists in combination with chemotherapy or with epidermal growth factor receptor (EGFR) inhibitors in non-small cell lung cancer (NSCLC) yielded disappointing results. Biomarker studies of clinical trials have identified IGF-1 levels as a potential marker of sensitivity to IGF-1R inhibition. Further study will need to focus on selection of NSCLC patients most likely to benefit from the addition of IGF-1R antagonists to standard therapy and the development of rational strategies for combination therapy in NSCLC.
Collapse
|
79
|
Bhutani D, Vaishampayan UN. Monoclonal antibodies in oncology therapeutics: present and future indications. Expert Opin Biol Ther 2013; 13:269-82. [PMID: 23286740 DOI: 10.1517/14712598.2012.758705] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Over the last decade, the field of oncology has undergone revolutionary changes. One of the major reasons contributing to this change is the improvement in our understanding of the biology of cancer. Recognition of novel targets on the cancer cell has enabled development of tools to attack those targets. Monoclonal antibodies represent such a therapy that has rapidly been adapted in almost all major cancer subtypes. AREAS COVERED This review intends to give a comprehensive overview of monoclonal antibodies, including mechanism of action, the currently approved agents and future targets. The authors reviewed published data as well as information from the ongoing clinical trials. EXPERT OPINION Monoclonal antibodies represent a major new advance in oncology therapy but there remains significant room for improvement.
Collapse
Affiliation(s)
- Divaya Bhutani
- Wayne State University, Barbara Ann Karmanos Cancer Institute, Department of Oncology, Department of Medicine, 4 Hudson Webber Cancer Research Center, 4100 John R, Detroit, MI 48201, USA
| | | |
Collapse
|
80
|
Scagliotti GV, Novello S, Rapetti S, Papotti M. Current state-of-the-art therapy for advanced squamous cell lung cancer. Am Soc Clin Oncol Educ Book 2013:354-358. [PMID: 23714545 DOI: 10.14694/edbook_am.2013.33.354] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Squamous cell carcinoma (SCC) represents the second most common histologic subtype of lung cancer (preceded only by adenocarcinoma). SSC of the lung is prevalently diagnosed in smokers and has been described as a preferentially centrally located tumor in which the main airways are commonly involved. Clinically, it presents with predominant locoregional signs and symptoms, but in recent years an increasing frequency of peripheral SCC of the lung has been reported. Pathologic diagnosis can be easily made through light microscopy and immunohistochemistry. The treatment approach for early-stage disease does not differ from that of other histologic subtypes of non-small cell lung cancer; in locally advanced unresectable or metastatic disease, doublet chemotherapy regimens (including cisplatin or carboplatin and a third-generation agent such as gemcitabine, taxanes, or vinorelbine) remain the cornerstone of front-line systemic treatment. Conversely, a single agent, mainly docetaxel, is the preferred treatment in second-line treatment. In unselected patient populations, targeted therapies have been extensively tested in combination with cytotoxic chemotherapy with disappointing results because of increased toxicity or lack of improvement in efficacy outcomes. Genomic alterations in SCC of the lung have not been comprehensively characterized, and no molecularly targeted therapies have been specifically developed for the treatment of this disease, but recently immune checkpoints have emerged as new therapeutic agent.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/classification
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/epidemiology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Clinical Trials as Topic
- Genes, Neoplasm
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/epidemiology
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Mutation
- Neoplasm Proteins/analysis
- Neoplasm Proteins/genetics
- Salvage Therapy
- Treatment Outcome
Collapse
Affiliation(s)
- Giorgio V Scagliotti
- From the Department of Oncology, University of Torino, San Luigi Hospital, Orbassano, Torino, Italy
| | | | | | | |
Collapse
|
81
|
Gombos A, Metzger-Filho O, Dal Lago L, Awada-Hussein A. Clinical development of insulin-like growth factor receptor--1 (IGF-1R) inhibitors: at the crossroad? Invest New Drugs 2012; 30:2433-42. [PMID: 22415797 PMCID: PMC3484277 DOI: 10.1007/s10637-012-9811-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 02/28/2012] [Indexed: 12/20/2022]
Abstract
Insulin like growth factor receptor (IGF-1R) targeting became one of the most investigated areas in anticancer drug development during the last decade. Strategies aiming to block IGF-1R activity include monoclonal antibodies, tyrosine kinase inhibitors and anti-ligands antibodies. Initial enthusiasm quickly encountered challenges. Unfortunately the validation of the efficacy of IGF-1R targeted agents in large clinical trials failed, however anecdotal single agent activity was seen in early studies. Consequently, questions regarding the selection of right target population and the appropriate trial design are arising. Despite the plethora of clinical trials conducted no predictive biomarker has been validated so far and resistance mechanisms to IGF-1R inhibitors remain unclear. The other issue to be addressed is how to best combine IGF-1R inhibitors with other therapeutic approaches. This review highlights the most relevant clinical data emphasizing the main tumor types where IGF-1R inhibition showed potential interest. We also tried to extract based on clinical and translational data some candidate biomarkers that could help better to select patient population who potentially could benefit most from this therapeutic approach.
Collapse
Affiliation(s)
- Andrea Gombos
- Medical Oncology Clinic, Institut Jules Bordet, 121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Otto Metzger-Filho
- Medical Oncology Clinic, Institut Jules Bordet, 121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Lissandra Dal Lago
- Medical Oncology Clinic, Institut Jules Bordet, 121 Boulevard de Waterloo, 1000 Brussels, Belgium
| | - Ahmad Awada-Hussein
- Medical Oncology Clinic, Institut Jules Bordet, 121 Boulevard de Waterloo, 1000 Brussels, Belgium
- Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
82
|
Sereno M, Esteban IR, Zambrana F, Merino M, Gómez-Raposo C, López-Gómez M, Sáenz EC. Squamous-cell carcinoma of the lungs: Is it really so different? Crit Rev Oncol Hematol 2012; 84:327-39. [DOI: 10.1016/j.critrevonc.2012.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 06/22/2012] [Accepted: 06/27/2012] [Indexed: 12/24/2022] Open
|
83
|
Abstract
Identifying specific somatic mutations that drive tumor growth has transformed the treatment of lung cancer. For example, cancers with sensitizing epidermal growth factor receptor mutations and echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase translocations can have remarkable responses to epidermal growth factor receptor and ALK inhibitors respectively, leading to significant clinical benefit. However, effective molecularly targeted therapies have disproportionately impacted adenocarcinomas compared to squamous cell carcinomas, and never or light smokers compared to heavy smokers. Further progress in non-small-cell lung cancer will require the identification and effective targeting of molecular alterations in all subtypes of lung cancer. Here, we review the current knowledge about the molecular alterations found in squamous cell carcinoma of the lung. First, we will discuss the ongoing efforts to comprehensively assess the squamous cell carcinoma genome. We will then discuss the evidence supporting the role of specific genes in driving squamous cell carcinomas. By describing the landscape of somatic targets in squamous cell lung cancer, we hope to crystallize the current understanding of potential targets, spur development of therapies that can have clinical impact, and underscore the importance of new discoveries in this field.
Collapse
|
84
|
Akt kinase-interacting protein1, a novel therapeutic target for lung cancer with EGFR-activating and gatekeeper mutations. Oncogene 2012; 32:4427-35. [DOI: 10.1038/onc.2012.446] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 08/07/2012] [Accepted: 08/09/2012] [Indexed: 01/17/2023]
|
85
|
de Melo Gagliato D, Shimada AK, Jardim DL, Katz A. Primary treatment of advanced non-small-cell lung cancer: role of monoclonal antibodies. Lung Cancer Manag 2012. [DOI: 10.2217/lmt.12.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Metastatic non-small-cell lung cancer (NSCLC) is one of the most incident cancers for which no curative therapies exist. New insights into the genetic background of the disease have led to the development of new effective therapies, including monoclonal antibodies. Bevacizumab demonstrated improved response rate and progression-free survival in two Phase III trials in association with conventional chemotherapy as first-line treatment for metastatic NSCLC. Cetuximab has proved to be effective in patients with advanced NSCLC expressing EGFR by immunohistochemistry. One of the promising targets in NSCLC is the MET pathway, for which monoclonal antibodies are under development. This review aims to describe the role of monoclonal antibodies for advanced NSCLC and future perspectives in this field.
Collapse
Affiliation(s)
| | | | | | - Artur Katz
- Centro de Oncologia, Hospital Sirio Libanes, Sao Paulo, Brazil
| |
Collapse
|
86
|
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that functions as a key regulatory protein in normal cell growth, survival, metabolism, development, and angiogenic pathways. Deregulation of these processes is a required hallmark of cancer, and dysregulation of mTOR signaling frequently occurs in a wide variety of malignancies, including lung cancer. Targeting of mTOR is thus an attractive strategy in the development of therapeutic agents against lung cancer. In this review, the mTOR-signaling pathway is described, highlighting opportunities for therapeutic intervention and biomarker analysis, and clinical trials in lung cancer including both non-small cell lung cancer and small cell lung cancer.
Collapse
|
87
|
邵 岚, 宋 正, 张 沂, 苏 丹. [Advances of molecular subtype and targeted therapy of lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2012; 15:545-52. [PMID: 22989458 PMCID: PMC5999864 DOI: 10.3779/j.issn.1009-3419.2012.09.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 07/30/2012] [Indexed: 11/24/2022]
Abstract
The discovery of multiple molecular mechanisms underlying the development, progression, and prognosis of lung cancer, has created new opportunities for targeted therapy. Each subtype is associated with molecular tests that define the subtype and drugs that may have potential therapeutic effect on lung cancer. In 2004, mutations in the epidermal growth factor receptor (epidermal growth factor receptor, EGFR) gene were discovered in non-small cell lung cancers (NSCLC), especially in adenocarcinomas. And they are strongly associated with sensitivity to EGFR-tyrosine kinase inhibitors (EGFR-TKIs). Moreover, in 2007 the existence of the echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) fusion gene was discovered in NSCLC, and the same as EGFR-TKIs, ALK inhibitors are being found to be highly effective in lung cancers. At present, multiple molecular subtype of lung cancer and relevant targeted drugs are undering study. Here, we review the remarkable progress in molecular subtype of lung cancer and the related targeted therapy.
Collapse
Affiliation(s)
- 岚 邵
- 310022 杭州,浙江省肿瘤医院化疗中心Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
- 310022 杭州,浙江省胸部肿瘤重点实验室Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou 310022, China
| | - 正波 宋
- 310022 杭州,浙江省肿瘤医院化疗中心Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
- 310022 杭州,浙江省胸部肿瘤重点实验室Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou 310022, China
| | - 沂平 张
- 310022 杭州,浙江省肿瘤医院化疗中心Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
- 310022 杭州,浙江省胸部肿瘤重点实验室Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou 310022, China
| | - 丹 苏
- 310022 杭州,浙江省胸部肿瘤重点实验室Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou 310022, China
- 310022 杭州,浙江省肿瘤医院肿瘤研究所Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
88
|
Chang CH, Wang Y, Trisal P, Li R, Rossi DL, Nair A, Gupta P, Losman M, Cardillo TM, Rossi EA, Goldenberg DM. Evaluation of a novel hexavalent humanized anti-IGF-1R antibody and its bivalent parental IgG in diverse cancer cell lines. PLoS One 2012; 7:e44235. [PMID: 22952934 PMCID: PMC3432068 DOI: 10.1371/journal.pone.0044235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/30/2012] [Indexed: 12/11/2022] Open
Abstract
A major mechanism of monoclonal antibodies that selectively target the insulin-like growth factor type 1 receptor (IGF-1R) to inhibit tumor growth is by downregulating the receptor, regardless whether they are capable (antagonistic) or incapable (agonistic) of blocking the binding of cognate ligands. We have developed and characterized a novel agonistic anti-IGF-1R humanized antibody, hR1, and used the Dock-and-Lock (DNL) method to construct Hex-hR1, the first multivalent antibody comprising 6 functional Fabs of hR1, with the aim of enhancing potency of hR1. Based on cross-blocking experiments, hR1 recognizes a region of cysteine-rich domain on the α-subunit, different from the epitopes mapped for existing anti-IGF-1R antibodies, yet hR1 is similar to other anti-IGF-1R antibodies in downregulating IGF-1R and inhibiting proliferation, colony formation, or invasion of selected cancer cell lines in vitro, as well as suppressing growth of the RH-30 rhabdomyosarcoma xenograft in nude mice when combined with the mTOR inhibitor, rapamycin. Hex-hR1 and hR1 are generally comparable in their bioactivities under the in-intro and in-vivo conditions investigated. Nevertheless, in selective experiments involving a direct comparison of potency, Hex-hR1 demonstrated a stronger effect on inhibiting cell proliferation stimulated by IGF-1 and could effectively downregulate IGF-1R at a concentration as low as 20 pM.
Collapse
Affiliation(s)
- Chien-Hsing Chang
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
| | - Yang Wang
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Preeti Trisal
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Rongxiu Li
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Diane L. Rossi
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Anju Nair
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Pankaj Gupta
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Michele Losman
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | | | - Edmund A. Rossi
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
| | - David M. Goldenberg
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
- Center of Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, New Jersey, United States of America
| |
Collapse
|
89
|
Warth A, Muley T, Herpel E, Meister M, Herth FJF, Schirmacher P, Weichert W, Hoffmann H, Schnabel PA. Large-scale comparative analyses of immunomarkers for diagnostic subtyping of non-small-cell lung cancer biopsies. Histopathology 2012; 61:1017-25. [PMID: 22882703 DOI: 10.1111/j.1365-2559.2012.04308.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
AIMS Patient stratification according to histological subtype is important for non-small-cell lung cancer (NSCLC) therapy. For specimens with uncertain histomorphology, rational and material-saving algorithms for specific and sensitive immunotyping need to be established. METHODS AND RESULTS One thousand one hundred and forty-five NSCLCs were immunohistochemically investigated for the expression of cytokeratin 5/6 (CK5/6), CK7, thyroid transcription factor-1 (TTF-1), p63, napsin-A, and desmocollin-3. Overall, napsin-A and desmocollin-3 were the most specific markers (specificity of each, 99%), and CK7 and CK5/6 the most sensitive markers (sensitivity, 96% and 94%) for adenocarcinomatous and squamous differentiation, respectively. However, for NSCLC not otherwise specified (NOS) cases, TTF-1, p63, CK5/6 and CK7 were found to be the most reliable markers. On the basis of morphology alone, approximately two-thirds of all NSCLCs could be reliably diagnosed in biopsy specimens. Immunohistochemistry further reduced the NOS fraction to 10%. CONCLUSIONS When morphology alone is not reliable, the use of selected markers and marker panels is highly sensitive and specific, and allows reliable distinction between squamous cell carcinoma and adenocarcinoma. Considering the impact of typing for the selection of molecular testing and treatment response, one must be aware of immunomarker expression patterns in NSCLC and their diagnostic value, in order to optimize typing and thereby maximize patient benefit from chemotherapy.
Collapse
Affiliation(s)
- Arne Warth
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Goto Y, Sekine I, Tanioka M, Shibata T, Tanai C, Asahina H, Nokihara H, Yamamoto N, Kunitoh H, Ohe Y, Kikkawa H, Ohki E, Tamura T. Figitumumab combined with carboplatin and paclitaxel in treatment-naïve Japanese patients with advanced non-small cell lung cancer. Invest New Drugs 2012; 30:1548-56. [PMID: 21748299 PMCID: PMC3388259 DOI: 10.1007/s10637-011-9715-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 06/29/2011] [Indexed: 01/13/2023]
Abstract
OBJECTIVES The insulin-like growth factor (IGF) signaling pathway has been implicated in the pathogenesis of numerous tumor types, including non-small cell lung cancer (NSCLC). Figitumumab is a fully human IgG2 monoclonal antibody against IGF-1 receptor (IGF-1R). METHODS This phase I, open-label, dose-escalation study (ClinicalTrials.gov: NCT00603538) assessed the safety and tolerability of figitumumab (6, 10 and 20 mg/kg) in combination with carboplatin (area under the curve: 6 mg·min/mL) and paclitaxel (200 mg/m(2)) in Japanese patients (N = 19) with chemotherapy-naïve, advanced NSCLC. Treatments were administered intravenously on day 1 of a 21-day cycle for four to six cycles. Pharmacokinetics, biomarkers, and antitumor activity were also evaluated. RESULTS Figitumumab in combination with carboplatin and paclitaxel was well tolerated at doses up to 20 mg/kg; no dose-limiting toxicities were observed at this dose level. When given in combination, figitumumab plasma exposure increased in an approximately dose-proportional manner. The approximate 2-fold accumulation following repeated administration supported the 21-day regimen as appropriate for figitumumab administration. Serum total IGF-1 and IGF binding protein-3 concentrations increased following figitumumab dosing, but a clear dose-dependent relationship was not demonstrated. Seven of 18 evaluable patients experienced a partial response. CONCLUSIONS Figitumumab 20 mg/kg in combination with carboplatin and paclitaxel was well tolerated in chemotherapy-naïve Japanese patients with NSCLC. Further analysis of biomarker data is necessary for the development of figitumumab therapy.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/blood
- Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Asian People
- Biomarkers, Tumor/blood
- Carboplatin/adverse effects
- Carboplatin/blood
- Carboplatin/pharmacokinetics
- Carboplatin/therapeutic use
- Carcinoma, Non-Small-Cell Lung/blood
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Dose-Response Relationship, Drug
- Female
- Humans
- Immunoglobulins, Intravenous
- Japan
- Lung Neoplasms/blood
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Paclitaxel/adverse effects
- Paclitaxel/blood
- Paclitaxel/pharmacokinetics
- Paclitaxel/therapeutic use
- Time Factors
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- Yasushi Goto
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Ikuo Sekine
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Maki Tanioka
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Takashi Shibata
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Chiharu Tanai
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Hajime Asahina
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Hiroshi Nokihara
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Noboru Yamamoto
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Hideo Kunitoh
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Yuichiro Ohe
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | | | - Emiko Ohki
- Pfizer Oncology, Global Research and Development, Tokyo, Japan
| | - Tomohide Tamura
- Department of Internal Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| |
Collapse
|
91
|
Yee D. Insulin-like growth factor receptor inhibitors: baby or the bathwater? J Natl Cancer Inst 2012; 104:975-81. [PMID: 22761272 DOI: 10.1093/jnci/djs258] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The success of targeted therapies for cancer is undisputed; strong preclinical evidence has resulted in the approval of several new agents for cancer treatment. The type I insulin-like growth factor receptor (IGF1R) appeared to be one of these promising new targets. Substantial population and preclinical data have all pointed toward this pathway as an important regulator of tumor cell biology. Although early results from clinical trials that targeted the IGF1R showed some evidence of response, larger randomized phase III trials have not shown clear clinical benefit of targeting this pathway in combination with conventional strategies. These disappointing results have resulted in the discontinuation of several anti-IGF1R programs. However, the conduct of these trials has brought to the forefront several important factors that need to be considered in the conduct of future clinical trials. The need to develop biomarkers, a clearer understanding of insulin receptor function, and defining rational combination regimens all require further consideration. In this commentary, the current state of IGF1R inhibitors in cancer therapy is reviewed.
Collapse
Affiliation(s)
- Douglas Yee
- Masonic Cancer Center, University of Minnesota420 Delaware Street SE, Minneapolis, MN, USA.
| |
Collapse
|
92
|
Oncogenic Pathways, Molecularly Targeted Therapies, and Highlighted Clinical Trials in Non–Small-Cell Lung Cancer (NSCLC). Clin Lung Cancer 2012; 13:252-66. [DOI: 10.1016/j.cllc.2011.09.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 09/23/2011] [Accepted: 09/24/2011] [Indexed: 02/06/2023]
|
93
|
Asmane I, Watkin E, Alberti L, Duc A, Marec-Berard P, Ray-Coquard I, Cassier P, Decouvelaere AV, Ranchère D, Kurtz JE, Bergerat JP, Blay JY. Insulin-like growth factor type 1 receptor (IGF-1R) exclusive nuclear staining: a predictive biomarker for IGF-1R monoclonal antibody (Ab) therapy in sarcomas. Eur J Cancer 2012; 48:3027-35. [PMID: 22682017 DOI: 10.1016/j.ejca.2012.05.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 05/01/2012] [Accepted: 05/09/2012] [Indexed: 01/16/2023]
Abstract
AIMS A minority of patients with advanced sarcoma achieve prolonged progression free survival (PFS) with insulin growth factor type 1 receptor (IGF-1R) monoclonal antibody (Ab) therapy. A biomarker identifying those patients beforehand would be useful to select patients for the development of these agents. METHODS This single centre series includes patients with unresectable or metastatic soft tissue sarcomas (STS), Ewing sarcoma (ES) and osteosarcoma treated with IGF-1R Ab (R1507, IMC-A12, SCH 717454 and CP-751.871) in the Centre Léon Bérard. Tumour samples were analysed by immunohistochemistry for expression of IGF-1R, insulin-like growth factor binding protein type 3 (IGFBP-3), Ki67, epidermal growth factor receptor (HER1) and human epidermal growth factor receptor 2 (HER2). Predictive factors for PFS and overall survival (OS) were investigated. RESULTS All tumour samples had a positive IGF-1R immunostaining on 60% to 100% of tumour cells. IGFBP-3 immunostaining was observed in 12 (75%) samples with 5% to 100% of positive cells. IGF-1R immunostaining was nuclear (n=9, 56%), cytoplasmic (n=4, 25%), or nuclear +cytoplasmic (n=3, 19%). Neither IGFBP-3 expression, nor Ki67 was correlated to PFS. HER2 and HER1 staining were positive in 0 and 2 samples respectively (both primary resistant to IGF-1R Ab therapy). Exclusive intra-nuclear immunoreactivity for IGF-1R was significantly associated with a better PFS (p=0.01) and OS (p=0.007). CONCLUSION Exclusive nuclear localisation of IGF-1R is an easily testable biomarker associated with a better PFS and OS for patients treated with IGF-1R Ab therapy. Nuclear localisation of IGF-1R in tumour cells might be a hallmark of pathway activation.
Collapse
Affiliation(s)
- Irène Asmane
- CHU Strasbourg, 1, Avenue Molière, 67000 Strasbourg, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Abstract
Insulin-like growth factor (IGF) plays an important role in tissue growth and development. Several studies have demonstrated the association between circulating levels of IGF-1 and -2 and cancer risk, and the IGF system has been implicated in the oncogenesis of essentially all solid and hematologic malignancies. The optimal strategy for targeting IGF signaling in patients with cancer is not clear. The modest benefits reported thus far underscore the need for a better understanding of IGF signaling, which would enable clinicians to identify the subset of patients with the greatest likelihood of attaining benefit from this targeted approach.
Collapse
Affiliation(s)
- S John Weroha
- Department of Oncology, Mayo Clinic College of Medicine, 200 First Street Southwest, Rochester, MN 55905, USA
| | | |
Collapse
|
95
|
Capelletto E, Novello S. Emerging New Agents for the Management of Patients with Non-Small Cell Lung Cancer. Drugs 2012; 72 Suppl 1:37-52. [DOI: 10.2165/1163028-s0-000000000-00000] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
96
|
Gold KA, Wistuba II, Kim ES. New strategies in squamous cell carcinoma of the lung: identification of tumor drivers to personalize therapy. Clin Cancer Res 2012; 18:3002-7. [PMID: 22461458 PMCID: PMC3737591 DOI: 10.1158/1078-0432.ccr-11-2055] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment for non-small cell lung cancer has been improving, with personalized treatment increasingly becoming a reality in the clinic. Unfortunately, these advances have largely been confined to the treatment of adenocarcinomas. Treatment options for squamous cell carcinoma (SCC) of the lung have lagged behind, partly because of a lack of understanding of the oncogenes driving SCC. Cytotoxic chemotherapy continues to be the only treatment option for many of our patients, and no genetic tests are clinically useful for patients with SCC. Recent advances in basic science have identified mutations and alterations in protein expression frequently found in SCCs, and clinical trials are ongoing to target these changes.
Collapse
Affiliation(s)
- Kathryn A. Gold
- Departments of Thoracic/Head and Neck Medical Oncology, Houston, Texas
| | - Ignacio I. Wistuba
- Departments of Thoracic/Head and Neck Medical Oncology, Houston, Texas
- Pathology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Edward S. Kim
- Departments of Thoracic/Head and Neck Medical Oncology, Houston, Texas
| |
Collapse
|
97
|
Abstract
The IGF axis is a tightly controlled endocrine system that regulates cell growth and development, known to have an important function in cancer biology. IGF1 and IGF2 can promote cancer growth in a GH-independent manner both through paracrine and autocrine secretion and can also confer resistance to chemotherapy and radiation. Many alterations of this system have been found in neoplasias, including increased expression of ligands and receptors, loss of heterozygosity of the IGF2 locus and increased IGF1R gene copy number. The IGF1 network is an attractive candidate for targeted therapy, including receptor blockade with monoclonal antibodies and small molecule inhibitors of receptor downstream signaling. This article reviews the role of the IGF axis in the initiation and progression of cancer, and describes the recent advances in IGF inhibition as a therapeutic tool.
Collapse
Affiliation(s)
- Fernanda I Arnaldez
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 CRC Room 1-3816, Bethesda, MD 20892, USA.
| | | |
Collapse
|
98
|
Kalra N, Zhang J, Yu Y, Ho M, Merino M, Cao L, Hassan R. Efficacy of anti-insulin-like growth factor I receptor monoclonal antibody cixutumumab in mesothelioma is highly correlated with insulin growth factor-I receptor sites/cell. Int J Cancer 2012; 131:2143-52. [PMID: 22323052 DOI: 10.1002/ijc.27471] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/24/2012] [Indexed: 12/29/2022]
Abstract
Insulin growth factor-I receptor (IGF-IR) is expressed in mesothelioma and therefore an attractive target for therapy. The antitumor activity of cixutumumab, a humanized monoclonal antibody to IGF-IR, in mesothelioma and relationship to IGF-IR expression was investigated using eight early passage tumor cells obtained from patients, nine established cell lines and an in vivo human mesothelioma tumor xenograft model. Although IGF-IR expression at the mRNA and protein level was present in all mesothelioma cells, using a quantitative ELISA immunoassay, there was considerable variability of IGF-IR expression ranging from 1 to 14 ng/mg of lysate. Using flow cytometry, the number of IGF-IR surface receptors varied from ≈ 2,000 to 50,000 sites/cell. Cells expressing >10,000 sites/cell had greater than 10% growth inhibition when treated with cixutumumab (100 μg/ml). Cixutumumab also induced antibody-dependent cell-mediated toxicity (>10% specific lysis) in cell lines, which had >20,000 IGF-IR sites/cell. Treatment with cixutumumab decreased phosphorylation of IGF-IR, Akt and Erk in cell lines, H226 and H28 having 24,000 and 51,000 IGF-IR sites/cell, respectively, but not in the cell line H2052 with 3,000 IGF-IR sites/cell. In vivo, cixutumumab treatment delayed growth of H226 mesothelioma tumor xenografts in mice and improved the overall survival of these mice compared to mice treated with saline (p < 0.004). Our results demonstrate that the antitumor efficacy of cixutumumab including inhibition of IGF-IR downstream signaling is highly correlated with IGF-IR sites/cell. A phase II clinical trial of cixutumumab is currently ongoing for the treatment of patients with mesothelioma.
Collapse
Affiliation(s)
- Neetu Kalra
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
Fidler MJ, Shersher DD, Borgia JA, Bonomi P. Targeting the insulin-like growth factor receptor pathway in lung cancer: problems and pitfalls. Ther Adv Med Oncol 2012; 4:51-60. [PMID: 22423264 PMCID: PMC3296080 DOI: 10.1177/1758834011427576] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The insulin-like growth factor (IGF) pathway is a complex pathway involving interactions between membrane-bound receptors, ligands, binding proteins, downstream effectors, and other receptor tyrosine kinase signaling cascades. The IGF pathway has been identified as a potential therapeutic target in non-small cell lung cancer (NSCLC) based on the following provocative factors. Preclinical observations in NSCLC have shown that this pathway is involved in tumor cell proliferation, survival, and invasiveness. In addition, IGF-1R protein expression is found in a significant number of non-small cell tumor specimens. Initial therapeutic efforts involved the development of monoclonal antibodies and tyrosine kinase inhibitors that target IGF-1R, a transmembrane receptor tyrosine kinase. Enthusiasm for targeting this pathway increased when a randomized phase II study showed that combining an anti-IGF-1R monoclonal antibody (figitumumab) with a platinum doublet resulted in a higher response rate and trends for superior progression-free survival and overall survival. Subsequently, a phase III study failed to confirm the promising results observed in the phase II trial. Currently, investigators are studying different monoclonal antibodies and tyrosine kinases targeting IGF-1R. In unselected patients, results presented thus far do not suggest efficacy of this agent. However, retrospective subgroup analyses suggest that circulating IGF-1 levels might identify patients who could benefit from treatment with an IGF-1R monoclonal antibody and may warrant further exploratory studies for predictive molecular markers. The purpose of this paper is to briefly discuss the IGF pathway and its relationship with other signaling pathways in lung cancer and to review the ongoing IGF clinical trials and efforts to identify predictive molecular markers.
Collapse
Affiliation(s)
- Mary Jo Fidler
- Section of Medical Oncology, Rush University Medical Center, 1725 West Harrison Street, Suite 821, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
100
|
Gao J, Chang YS, Jallal B, Viner J. Targeting the insulin-like growth factor axis for the development of novel therapeutics in oncology. Cancer Res 2012; 72:3-12. [PMID: 22215692 DOI: 10.1158/0008-5472.can-11-0550] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulin-like growth factors (IGF) are polypeptide hormones with potent anabolic and mitogenic effects that regulate cell growth and differentiation. Dysregulation of the IGF axis has been well documented in the development and progression of multiple types of cancer. As a result, compounds targeting the IGF axis have become an area of intense preclinical and clinical research for cancer therapeutics. The IGF axis is intimately involved with the insulin-signaling pathway because of their close homologies. This homology may explain hurdles encountered in the clinical development of IGF-targeted therapies, such as less-than-expected antitumor efficacy that may arise from compensatory increases in the activity of insulin receptor isoform A (IR-A), in response to IGF-I receptor (IGF-IR) inhibition and perturbations in glucose homeostasis, arising from the inhibition of insulin receptor isoform B (IR-B) activity. In this brief review, we compare differentiating factors that characterize the 3 major classes of IGF-targeting compounds: therapeutic antibodies that target IGF-IR, small molecule tyrosine kinase inhibitors that inhibit kinase activities of IGF-IR and IR, and antibodies that target IGF ligands.
Collapse
Affiliation(s)
- Jin Gao
- MedImmune, LLC, Gaithersburg, Maryland, USA
| | | | | | | |
Collapse
|