1
|
Heumann P, Albert A, Gülow K, Tümen D, Müller M, Kandulski A. Current and Future Therapeutic Targets for Directed Molecular Therapies in Cholangiocarcinoma. Cancers (Basel) 2024; 16:1690. [PMID: 38730642 PMCID: PMC11083102 DOI: 10.3390/cancers16091690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/17/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
We conducted a comprehensive review of the current literature of published data, clinical trials (MEDLINE; ncbi.pubmed.com), congress contributions (asco.org; esmo.org), and active recruiting clinical trains (clinicaltrial.gov) on targeted therapies in cholangiocarcinoma. Palliative treatment regimens were analyzed as well as preoperative and perioperative treatment options. We summarized the current knowledge for each mutation and molecular pathway that is or has been under clinical evaluation and discussed the results on the background of current treatment guidelines. We established and recommended targeted treatment options that already exist for second-line settings, including IDH-, BRAF-, and NTRK-mutated tumors, as well as for FGFR2 fusion, HER2/neu-overexpression, and microsatellite instable tumors. Other options for targeted treatment include EGFR- or VEGF-dependent pathways, which are known to be overexpressed or dysregulated in this cancer type and are currently under clinical investigation. Targeted therapy in CCA is a hallmark of individualized medicine as these therapies aim to specifically block pathways that promote cancer cell growth and survival, leading to tumor shrinkage and improved patient outcomes based on the molecular profile of the tumor.
Collapse
Affiliation(s)
- Philipp Heumann
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases University Hospital Regensburg Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | - Arne Kandulski
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases University Hospital Regensburg Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
2
|
Hua C, Wenyong P, Zhongquan Z, Chang X, Xiayun J. Effects of propofol on IGF-1 activity and cell behaviour in the GES 1 mucosal cell model. Growth Factors 2023; 41:32-42. [PMID: 36441130 DOI: 10.1080/08977194.2022.2150189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Propofol is an important and widely used anaesthetic drug in the clinic. Many works have shown that propofol has important biological functions except as an anaesthetic. In the current study, we mainly explored the effect of propofol on the biological activity of IGF-1, which is an important growth factor involved in regulating the growth and development of the stomach. Here, we explored the effect of propofol on the biological activity of IGF-1 in a GES-1-cell model. We found that propofol affected the biological activity of IGF-1. It not only reduces IGF-1/IGF-1R signalling but also changes IGF-1R cell characteristics. We further explored the mechanism by which propofol affected IGF-1 activity. Through a series of experiments, we found that propofol affected the stability of membrane-localised IGF-1R. It also affects the recycling of the IGF-1R receptor Propofol can affect the degradation of IGF-1R by changing the endocytosis of IGF-1R. In short, the current study found that propofol affected the biological activity of IGF-1, which laid the foundation for related research.
Collapse
Affiliation(s)
- Chai Hua
- Anesthesiology, Jinhua Central Hospital, JinHua, China
| | - Peng Wenyong
- Anesthesiology, Jinhua Central Hospital, JinHua, China
| | - Zhu Zhongquan
- Anesthesiology, Jinhua Central Hospital, JinHua, China
| | - Xiong Chang
- Anesthesiology, Jinhua Central Hospital, JinHua, China
| | - Jin Xiayun
- Oncology, Jinhua Central Hospital, JinHua, China
| |
Collapse
|
3
|
Liu QQ, Wang P, He QJ, Ma R, Lee SC. PPARγ promotes diabetes‐associated centrosome amplification via increasing the expression of SKA1 directly at the transcriptional level. J Cell Physiol 2019; 234:20694-20703. [DOI: 10.1002/jcp.28674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 03/09/2019] [Accepted: 03/19/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Qin Qin Liu
- Department of Biology, School of Life Sciences Shanxi University Taiyuan Shanxi PR China
| | - Pu Wang
- Department of Biology, School of Life Sciences Shanxi University Taiyuan Shanxi PR China
| | - Qin Ju He
- Department of Biology, School of Life Sciences Shanxi University Taiyuan Shanxi PR China
| | - Ronald Ma
- Department of Medicine and Therapeutics Chinese University of Hong Kong Shatin Hong Kong SAR PR China
| | - Shao Chin Lee
- Department of Biology, School of Life Sciences Shanxi University Taiyuan Shanxi PR China
- Department of Biology, School of Life Sciences Jiangsu Normal University Xuzhou Jiangsu PR China
| |
Collapse
|
4
|
Zhou X, Zhao X, Li X, Ping G, Pei S, Chen M, Wang Z, Zhou W, Jin B. PQ401, an IGF-1R inhibitor, induces apoptosis and inhibits growth, proliferation and migration of glioma cells. J Chemother 2017; 28:44-9. [PMID: 25971682 DOI: 10.1179/1973947815y.0000000026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Growth factor signalling pathways transduce extra-cellular physiological cues to guide cells to maintain critical cellular functions, including cell proliferation, survival and metabolism. Dysregulation of certain growth factor signalling pathways has been shown as a major route to promote tumourigenesis. Glioma is a type of aggressive malignant tumour with no effective systematic therapy so far. Overexpression or hyperactivation of IGF-1R has been observed to be tightly associated with glioma progression and poor prognosis. Here, we examined the biological effects of a specific IGF-1R inhibitor, PQ401, on suppressing U87MG glioma cell growth and migration. Specifically, we observed that PQ401 not only induced cellular apoptosis in U87MG cells and subsequently reduced cell viability and proliferation but also attenuated cell mobility in vitro. More importantly, through a mouse xenograft model, we observed that administration of PQ401 on mice led to suppression of glioma tumour growth in vivo. In summary, our study suggests that PQ401 may serve as a promising leading drug for treating glioma patients with elevated IGF-1R signalling.
Collapse
Affiliation(s)
- Xiang Zhou
- a Department of Neurosurgery , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Xinli Zhao
- a Department of Neurosurgery , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Xiangsheng Li
- a Department of Neurosurgery , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Guanfang Ping
- b Department of Pharmacy , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Sujuan Pei
- b Department of Pharmacy , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Ming Chen
- b Department of Pharmacy , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Zhongwei Wang
- a Department of Neurosurgery , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Wenke Zhou
- a Department of Neurosurgery , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| | - Baozhe Jin
- a Department of Neurosurgery , First Affiliated Hospital of Xinxiang Medical College , Weihui , Henan , P. R. China
| |
Collapse
|
5
|
Martelli C, Dico AL, Diceglie C, Lucignani G, Ottobrini L. Optical imaging probes in oncology. Oncotarget 2016; 7:48753-48787. [PMID: 27145373 PMCID: PMC5217050 DOI: 10.18632/oncotarget.9066] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/10/2016] [Indexed: 01/19/2023] Open
Abstract
Cancer is a complex disease, characterized by alteration of different physiological molecular processes and cellular features. Keeping this in mind, the possibility of early identification and detection of specific tumor biomarkers by non-invasive approaches could improve early diagnosis and patient management.Different molecular imaging procedures provide powerful tools for detection and non-invasive characterization of oncological lesions. Clinical studies are mainly based on the use of computed tomography, nuclear-based imaging techniques and magnetic resonance imaging. Preclinical imaging in small animal models entails the use of dedicated instruments, and beyond the already cited imaging techniques, it includes also optical imaging studies. Optical imaging strategies are based on the use of luminescent or fluorescent reporter genes or injectable fluorescent or luminescent probes that provide the possibility to study tumor features even by means of fluorescence and luminescence imaging. Currently, most of these probes are used only in animal models, but the possibility of applying some of them also in the clinics is under evaluation.The importance of tumor imaging, the ease of use of optical imaging instruments, the commercial availability of a wide range of probes as well as the continuous description of newly developed probes, demonstrate the significance of these applications. The aim of this review is providing a complete description of the possible optical imaging procedures available for the non-invasive assessment of tumor features in oncological murine models. In particular, the characteristics of both commercially available and newly developed probes will be outlined and discussed.
Collapse
Affiliation(s)
- Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
| | - Alessia Lo Dico
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Umberto Veronesi Foundation, Milan, Italy
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Tecnomed Foundation, University of Milan-Bicocca, Monza, Italy
| | - Giovanni Lucignani
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Institute for Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| |
Collapse
|
6
|
Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, Folseraas T, Forbes SJ, Fouassier L, Geier A, Calvisi DF, Mertens JC, Trauner M, Benedetti A, Maroni L, Vaquero J, Macias RIR, Raggi C, Perugorria MJ, Gaudio E, Boberg KM, Marin JJG, Alvaro D. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol 2016; 13:261-80. [PMID: 27095655 DOI: 10.1038/nrgastro.2016.51] [Citation(s) in RCA: 951] [Impact Index Per Article: 105.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous group of malignancies with features of biliary tract differentiation. CCA is the second most common primary liver tumour and the incidence is increasing worldwide. CCA has high mortality owing to its aggressiveness, late diagnosis and refractory nature. In May 2015, the "European Network for the Study of Cholangiocarcinoma" (ENS-CCA: www.enscca.org or www.cholangiocarcinoma.eu) was created to promote and boost international research collaboration on the study of CCA at basic, translational and clinical level. In this Consensus Statement, we aim to provide valuable information on classifications, pathological features, risk factors, cells of origin, genetic and epigenetic modifications and current therapies available for this cancer. Moreover, future directions on basic and clinical investigations and plans for the ENS-CCA are highlighted.
Collapse
Affiliation(s)
- Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, Ikerbasque, CIBERehd, Paseo del Dr. Begiristain s/n, E-20014, San Sebastian, Spain
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Guido Carpino
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis 6, 00135, Rome, Italy
| | - Marco Marzioni
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Jesper B Andersen
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Pietro Invernizzi
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
- Program for Autoimmune Liver Diseases, International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Via Cadore 48, 20900, Monza, Italy
| | - Guro E Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310, Oslo, Norway
| | - Trine Folseraas
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Pb. 4950 Nydalen, N-0424, Oslo, Norway
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, 49 Little France Crescent, EH16 4SB, Edinburgh, United Kingdom
| | - Laura Fouassier
- INSERM UMR S938, Centre de Recherche Saint-Antoine, 184 rue du Faubourg Saint-Antoine, 75571, Paris cedex 12, Fondation ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Andreas Geier
- Department of Internal Medicine II, University Hospital Würzburg, Oberdürrbacherstrasse 6, D-97080, Würzburg, Germany
| | - Diego F Calvisi
- Institute of Pathology, Universitätsmedizin Greifswald, Friedrich-Löffler-Strasse 23e, 17489, Greifswald, Germany
| | - Joachim C Mertens
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Antonio Benedetti
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Luca Maroni
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Javier Vaquero
- INSERM UMR S938, Centre de Recherche Saint-Antoine, 184 rue du Faubourg Saint-Antoine, 75571, Paris cedex 12, Fondation ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Rocio I R Macias
- Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Campus Miguel de Unamuno, E.I.D. S-09, University of Salamanca, IBSAL, CIBERehd, 37007, Salamanca, Spain
| | - Chiara Raggi
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, Ikerbasque, CIBERehd, Paseo del Dr. Begiristain s/n, E-20014, San Sebastian, Spain
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Via Alfonso Borelli 50, 00161, Rome, Italy
| | - Kirsten M Boberg
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Pb. 4950 Nydalen, N-0424, Oslo, Norway
| | - Jose J G Marin
- Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Campus Miguel de Unamuno, E.I.D. S-09, University of Salamanca, IBSAL, CIBERehd, 37007, Salamanca, Spain
| | - Domenico Alvaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| |
Collapse
|
7
|
Rudnick EW, Thareja S, Cherpelis B. Oral therapy for nonmelanoma skin cancer in patients with advanced disease and large tumor burden: a review of the literature with focus on a new generation of targeted therapies. Int J Dermatol 2015; 55:249-58; quiz 256, 258. [PMID: 26566923 DOI: 10.1111/ijd.12961] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/26/2014] [Accepted: 12/31/2014] [Indexed: 12/20/2022]
Abstract
Nonmelanoma skin cancer (NMSC) is the most common cancer in patients and includes basal cell carcinoma (BCC) and squamous cell carcinoma (SCC). Treatments useful for SCC and BCC include surgical, topical, and in advanced cases systemic chemo-radiation. This review of the literature aims to describe previous and current treatment options for oral therapy in locally advanced and metastatic NMSC otherwise unamenable to standard treatment. Oral Smoothened (Smo) inhibitors Vismodegib, Sonidegib, and Taladegib have shown to be effective in several trials. Oral tyrosine kinase inhibitors Erlotinib and Gefitinib, which target epidermal growth factor receptor (EGFR), have early supporting data and are currently undergoing large multicenter trials. Other less studied oral therapies which have shown at least partial efficacy include 5-Fluorouracil, capecitabine, and picropodophyllin. In vitro studies have elucidated new targets for dual combination oral therapy targeting both EGFR and insulin-like growth factor 1 receptor (IGF-1R). It is important to stratify treatment options based on patient risk of advanced disease, failure of conservative treatment, and ill-tolerated intravenous chemotherapy adverse events. Oral therapy in NMSC is useful in high risk patients with recurrent and aggressive disease who may not tolerate other systemic therapies.
Collapse
Affiliation(s)
- Eric W Rudnick
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sumeet Thareja
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Basil Cherpelis
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
8
|
Chugh R, Griffith KA, Davis EJ, Thomas DG, Zavala JD, Metko G, Brockstein B, Undevia SD, Stadler WM, Schuetze SM. Doxorubicin plus the IGF-1R antibody cixutumumab in soft tissue sarcoma: a phase I study using the TITE-CRM model. Ann Oncol 2015; 26:1459-64. [PMID: 25858498 DOI: 10.1093/annonc/mdv171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/27/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Insulin-like growth factor receptor (IGF-1R) has been studied as an oncologic target in soft tissue sarcoma (STS), but its role in sarcoma biology is unclear. Anti-IGF-1R antibody cixutumumab demonstrated acceptable toxicity but limited activity as a single agent in STS. We carried out a dose-escalation study of cixutumumab with doxorubicin to evaluate safety and dosing of the combination. PATIENTS AND METHODS Eligible patients with advanced STS were treated with cixutumumab intravenously on days 1/8/15 at one of three dose levels (A: 1 mg/kg, B: 3 mg/kg, C: 6 mg/kg) with doxorubicin at 75 mg/m(2) as a 48 h infusion on day 1 of a 21 day cycle. After six cycles of the combination, patients could receive cixutumumab alone. The Time-to-Event Continual Reassessment Method was used to estimate the probability of dose-limiting toxicity (DLT) and to assign patients to the dose with an estimated probability of DLT≤20%. RESULTS Between September 2008 and January 2012, 30 patients with advanced STS received a median of six cycles of therapy (range <1-22). Two DLTs were observed, grade 3 mucositis (dose level B) and grade 4 hyperglycemia (dose level C). Grade 2 and 3 reduced left ventricular ejection fraction was seen in three and two patients, respectively. Five partial responses were observed, and estimated progression-free survival was 5.3 months (95% confidence interval 3.0-6.3) in 26 response-assessable patients. Immunohistochemical staining of 11 available tumor samples for IGF-1R and phospho-IGF-1R was not significantly different among responders and non-responders, and serum analysis of select single-nucleotide polymorphisms did not predict for cardiotoxicity. CONCLUSION The maximum tolerated dose was doxorubicin 75 mg/m(2) on day 1 and cixitumumab 6 mg/kg on days 1/8/15 of a 21 day cycle. Cardiac toxicity was observed and should be monitored in subsequent studies, which should be considered in STS only if a predictive biomarker of benefit to anti-IGF-1R therapy is identified. TRIAL REGISTRATION ClinicalTrials.gov:NCT00720174.
Collapse
Affiliation(s)
- R Chugh
- Departments of Internal Medicine, University of Michigan, Ann Arbor
| | - K A Griffith
- Biostatistics, University of Michigan, Ann Arbor
| | - E J Davis
- Departments of Internal Medicine, University of Michigan, Ann Arbor
| | - D G Thomas
- Pathology, University of Michigan, Ann Arbor
| | - J D Zavala
- Cancer Clinical Trials Office, University of Chicago, Chicago
| | - G Metko
- Clinical Trials Office, University of Michigan, Ann Arbor
| | | | - S D Undevia
- Department of Medicine, University of Chicago, Chicago, USA
| | - W M Stadler
- Department of Medicine, University of Chicago, Chicago, USA
| | - S M Schuetze
- Departments of Internal Medicine, University of Michigan, Ann Arbor
| |
Collapse
|
9
|
Tian D, Kreeger PK. Analysis of the quantitative balance between insulin-like growth factor (IGF)-1 ligand, receptor, and binding protein levels to predict cell sensitivity and therapeutic efficacy. BMC SYSTEMS BIOLOGY 2014; 8:98. [PMID: 25115504 PMCID: PMC4236724 DOI: 10.1186/s12918-014-0098-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 08/05/2014] [Indexed: 01/06/2023]
Abstract
Background The insulin-like growth factor (IGF) system impacts cell proliferation and is highly activated in ovarian cancer. While an attractive therapeutic target, the IGF system is complex with two receptors (IGF1R, IGF2R), two ligands (IGF1, IGF2), and at least six high affinity IGF-binding proteins (IGFBPs) that regulate the bioavailability of IGF ligands. We hypothesized that a quantitative balance between these different network components regulated cell response. Results OVCAR5, an immortalized ovarian cancer cell line, were found to be sensitive to IGF1, with the dose of IGF1 (i.e., the total mass of IGF1 available) a more reliable predictor of cell response than ligand concentration. The applied dose of IGF1 was depleted by both cell-secreted IGFBPs and endocytic trafficking, with IGFBPs sequestering up to 90% of the available ligand. To explore how different variables (i.e., IGF1, IGFBPs, and IGF1R levels) impacted cell response, a mass-action steady-state model was developed. Examination of the model revealed that the level of IGF1-IGF1R complexes per cell was directly proportional to the extent of proliferation induced by IGF1. Model analysis suggested, and experimental results confirmed, that IGFBPs present during IGF1 treatment significantly decreased IGF1-mediated proliferation. We utilized this model to assess the efficacy of IGF1 and IGF1R antibodies against different network compositions and determined that IGF1R antibodies were more globally effective due to the receptor-limited state of the network. Conclusions Changes that affect IGF1R occupancy have predictable effects on IGF1-induced proliferation and our model captured these effects. Analysis of this model suggests that IGF1R antibodies will be more effective than IGF1 antibodies, although the difference was minimal in conditions with low levels of IGF1 and IGFBPs. Examining how different components of the IGF system influence cell response will be critical to improve our understanding of the IGF signaling network in ovarian cancer.
Collapse
|
10
|
Rota LM, Albanito L, Shin ME, Goyeneche CL, Shushanov S, Gallagher EJ, LeRoith D, Lazzarino DA, Wood TL. IGF1R inhibition in mammary epithelia promotes canonical Wnt signaling and Wnt1-driven tumors. Cancer Res 2014; 74:5668-79. [PMID: 25092896 DOI: 10.1158/0008-5472.can-14-0970] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease subtype that, unlike other subtypes, lacks an effective targeted therapy. Inhibitors of the insulin-like growth factor receptor (IGF1R) have been considered for use in treating TNBC. Here, we provide genetic evidence that IGF1R inhibition promotes development of Wnt1-mediated murine mammary tumors that offer a model of TNBC. We found that in a double transgenic mouse model carrying activated Wnt1 and mutant Igf1r, a reduction in IGF1R signaling reduced tumor latency and promoted more aggressive phenotypes. These tumors displayed a squamous phenotype with increased expression of keratins 5/6 and β-catenin. Notably, cell lineage analyses revealed an increase in basal (CD29(hi)/CD24(+)) and luminal (CD24(+)/CD61+/CD29(lo)) progenitor cell populations, along with increased Nanog expression and decreased Elf5 expression. In these doubly transgenic mice, lung metastases developed with characteristics of the primary tumors, unlike MMTV-Wnt1 mice. Mechanistic investigations showed that pharmacologic inhibition of the IGF1R in vitro was sufficient to increase the tumorsphere-forming efficiency ofMMTV-Wnt1 tumor cells. Tumors from doubly transgenic mice also exhibited an increase in the expression ratio of the IGF-II-sensitive, A isoform of the insulin receptor versus the IR-B isoform, which when stimulated in vitro resulted in enhanced expression of β-catenin. Overall, our results revealed that in Wnt-driven tumors, an attenuation of IGF1R signaling accelerates tumorigenesis and promotes more aggressive phenotypes with potential implications for understanding TNBC pathobiology and treatment.
Collapse
Affiliation(s)
- Lauren M Rota
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Lidia Albanito
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Marcus E Shin
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Corey L Goyeneche
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Sain Shushanov
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, New York
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, New York
| | - Deborah A Lazzarino
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Teresa L Wood
- Department of Neurology and Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical and Health Sciences, Newark, New Jersey.
| |
Collapse
|
11
|
Shin SJ, Gong G, Lee HJ, Kang J, Bae YK, Lee A, Cho EY, Lee JS, Suh KS, Lee DW, Jung WH. Positive expression of insulin-like growth factor-1 receptor is associated with a positive hormone receptor status and a favorable prognosis in breast cancer. J Breast Cancer 2014; 17:113-20. [PMID: 25013431 PMCID: PMC4090312 DOI: 10.4048/jbc.2014.17.2.113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 05/23/2014] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Insulin-like growth factor 1 receptor (IGF-1R) is commonly expressed in primary breast cancers. Understanding the role of IGF-1R signaling in the different subtypes of breast cancer is important because each subtype has a different outcome and requires different treatment modalities. However, the precise biological significance of IGF-1R expression in cancer cells is still unclear. In this study, we examined the expression of IGF-1R in the different molecular subtypes of breast cancer. The effects of IGF-1R expression on the survival rates and outcomes of breast cancer were also examined. METHODS IGF-1R expression was evaluated immunohistochemically in tissue microarray blocks constructed from 1,198 invasive breast cancer samples collected from six medical institutions. IGF-1R expression was interpreted according to the human epidermal growth factor receptor 2 (HER2)/neu immunohistochemistry scoring system. Scores of 2+ and 3+ were considered positive. RESULTS Positive IGF-1R expression was observed in 65.4% of invasive breast cancer samples. IGF-1R expression was detected in all cancer subtypes (luminal A, 84.4%; luminal B, 75.9%; HER2, 21.2%; triple-negative, 46.6%) and was found to be associated with a positive hormone receptor status and the absence of HER2 amplification (p<0.001). Positive IGF-1R expression was significantly associated with high survival rates (p=0.014). However, a multivariate analysis revealed that the expression levels of IGF-1R did not achieve statistical significance. In the triple-negative cancer subtype, IGF-1R expression was found to be associated with a lower disease-free survival rate (p=0.031). CONCLUSION Positive IGF-1R expression is associated with a favorable prognosis in breast cancer. IGF-1R is frequently expressed in the luminal A/B subtypes of breast cancer, and its expression is related to the hormone receptor status.
Collapse
Affiliation(s)
- Su-Jin Shin
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jun Kang
- Department of Pathology, Incheon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Incheon, Korea
| | - Young Kyung Bae
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Ahwon Lee
- Department of Pathology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Eun Yoon Cho
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Shin Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju, Korea
| | - Kwang-Sun Suh
- Department of Pathology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Dong Wha Lee
- Department of Pathology, Soonchunhyang University Hospital, Seoul, Korea
| | - Woo Hee Jung
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Singh P, Alex JM, Bast F. Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: novel treatment strategies for cancer. Med Oncol 2013; 31:805. [PMID: 24338270 DOI: 10.1007/s12032-013-0805-3] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/04/2013] [Indexed: 02/07/2023]
Abstract
Insulin and insulin-like growth factor (IGF) signaling system, commonly known for fine-tuning numerous biological processes, has lately made its mark as a much sought-after therapeutic targets for diabetes and cancer. These receptors make an attractive anticancer target owing to their overexpression in variety of cancer especially in prostate and breast cancer. Inhibitors of IGF signaling were subjected to clinical cancer trials with the main objective to confirm the effectiveness of these receptors as a therapeutic target. However, the results that these trials produced proved to be disappointing as the role played by the cross talk between IGF and insulin receptor (IR) signaling pathways at the receptor level or at downstream signaling level became more lucid. Therapeutic strategy for IGF-1R and IR inhibition mainly encompasses three main approaches namely receptor blockade with monoclonal antibodies, tyrosine kinase inhibition (ATP antagonist and non-ATP antagonist), and ligand neutralization via monoclonal antibodies targeted to ligand or recombinant IGF-binding proteins. Other drug-discovery approaches are employed to target IGF-1R, and IR includes antisense oligonucleotides and recombinant IGF-binding proteins. However, therapies with monoclonal antibodies and tyrosine kinase inhibition targeting the IGF-1R are not evidenced to be satisfactory as expected. Factors that are duly held responsible for the unsuccessfulness of these therapies include (a) the existence of the IR isoform A overexpressed on a variety of cancers, enhancing the mitogenic signals to the nucleus leading to the endorsement of cell growth, (b) IGF-1R and IR that form hybrid receptors sensitive to the stimulation of all three IGF axis ligands, and (c) IGF-1R and IR that also have the potential to form hybrid receptors with other tyrosine kinase to potentiate the cellular transformation, tumorigenesis, and tumor vascularization. This mini review is a concerted effort to explore and fathom the well-recognized roles of the IRA signaling system in human cancer phenotype and the main strategies that have been so far evaluated to target the IR and IGF-1R.
Collapse
Affiliation(s)
- Pushpendra Singh
- Centre for Biosciences, School of Basic and Applied Science, Central University of Punjab, Bathinda, 151001, Punjab, India
| | | | | |
Collapse
|
13
|
Arunkumar R, Sharmila G, Elumalai P, Senthilkumar K, Banudevi S, Gunadharini DN, Benson CS, Daisy P, Arunakaran J. Effect of diallyl disulfide on insulin-like growth factor signaling molecules involved in cell survival and proliferation of human prostate cancer cells in vitro and in silico approach through docking analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2012; 19:912-923. [PMID: 22739413 DOI: 10.1016/j.phymed.2012.04.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 03/21/2012] [Accepted: 04/29/2012] [Indexed: 06/01/2023]
Abstract
PURPOSE Diallyl Disulfide (DADS) is one of the major components of garlic, which inhibits the proliferation of various cancer cells. Our previous studies showed that DADS inhibits cell growth and induces apoptosis on prostate cancer cells. Insulin like growth factor signaling pathway plays a significant role on prostate cancer cell growth and survival and it's over expression also identified in human prostate cancers. The molecular mechanism of IGF mediated PI3K/Akt signaling remains to be elucidated. The present study was designed to evaluate the effects of diallyl disulfide on IGF signaling in androgen independent prostate cancer cells (PC-3). METHODS DADS (10-50 μM) caused dose-dependent inhibition of PC-3 cells, were analyzed by MTT, IC50 value of PC-3 cells was 40 μM for 24h. Interestingly, DADS also altered the mRNA and protein expressions of IGF signaling and apoptotic molecules which were confirmed by semi quantitative PCR and western blot method. Further the docking study of DADS with IGF receptor was carried out by Ligand Fit of Discovery studio. Accord Excel Package was used for the prediction of ADME properties of the compound. RESULTS The results suggests that DADS decreases the survival rate of androgen independent prostate cancer cells by modulating the expression of IGF system, which leads to inhibition of phosphorylation of Akt, thereby inhibits cell cycle progression and survival by lowering the expression of cyclin D1, NFkB and anti-apoptotic Bcl-2 molecule and increasing the level of pro-apoptotic (Bad and Bax) signaling molecules which leads to apoptosis. CONCLUSION The present investigation showed downregulation of Akt and a concomitant increase in apoptosis in DADS treated prostate cancer cells. Since inhibition of this Akt pathway by DADS leads to inhibition in cancer cell progression, it is highly suggested that DADS has the potential use as a therapy for prostate cancer.
Collapse
Affiliation(s)
- R Arunkumar
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai 600113, India
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Hou X, Huang F, Macedo LF, Harrington SC, Reeves KA, Greer A, Finckenstein FG, Brodie A, Gottardis MM, Carboni JM, Haluska P. Dual IGF-1R/InsR inhibitor BMS-754807 synergizes with hormonal agents in treatment of estrogen-dependent breast cancer. Cancer Res 2011; 71:7597-607. [PMID: 22042792 DOI: 10.1158/0008-5472.can-11-1080] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin-like growth factor (IGF) signaling has been implicated in the resistance to hormonal therapy in breast cancer. Using a model of postmenopausal, estrogen-dependent breast cancer, we investigated the antitumor effects of the dual IGF-1R/InsR tyrosine kinase inhibitor BMS-754807 alone and in combination with letrozole or tamoxifen. BMS-754807 exhibited antiproliferative effects in vitro that synergized strongly in combination with letrozole or 4-hydroxytamoxifen and fulvestrant. Similarly, combined treatment of BMS-754807 with either tamoxifen or letrozole in vivo elicited tumor regressions not achieved by single-agent therapy. Notably, hormonal therapy enhanced the inhibition of IGF-1R/InsR without major side effects in animals. Microarray expression analysis revealed downregulation of cell-cycle control and survival pathways and upregulation of erbB in response to BMS-754807 plus hormonal therapy, particularly tamoxifen. Overall, these results offer a preclinical proof-of-concept for BMS-754807 as an antitumor agent in combination with hormonal therapies in hormone-sensitive breast cancer. Cooperative cell-cycle arrest, decreased proliferation, and enhanced promotion of apoptosis may contribute to antitumor effects to be gauged in future clinical investigations justified by our findings.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/administration & dosage
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Synergism
- Estradiol/administration & dosage
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Estrogens/metabolism
- Female
- Fulvestrant
- Gene Expression Profiling
- Humans
- Letrozole
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Nitriles/administration & dosage
- Nitriles/pharmacology
- Oligonucleotide Array Sequence Analysis
- Pyrazoles/pharmacology
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/antagonists & inhibitors
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Tamoxifen/administration & dosage
- Tamoxifen/analogs & derivatives
- Tamoxifen/pharmacology
- Triazines/pharmacology
- Triazoles/administration & dosage
- Triazoles/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Med Chem 2011; 1:1153-71. [PMID: 21425998 DOI: 10.4155/fmc.09.89] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The IGF-1 receptor (IGF-1R) has been implicated in the promotion of tumorigenesis, metastasis and resistance to cancer therapies. Therefore, this receptor has become a major focus for the development of anticancer agents. RESULTS Our lead optimization efforts that blended structure-based design and empirical medicinal chemistry led to the discovery of OSI-906, a novel small-molecule dual IGF-1R/insulin receptor (IR) kinase inhibitor. OSI-906 potently and selectively inhibits autophosphorylation of both human IGF-1R and IR, displays in vitro antiproliferative effects in a variety of tumor cell lines and shows robust in vivo anti-tumor efficacy in an IGF-1R-driven xenograft model when administered orally once daily. CONCLUSION OSI-906 is a novel, potent, selective and orally bioavailable dual IGF-1R/IR kinase inhibitor with favorable preclinical drug-like properties, which has demonstrated in vivo efficacy in tumor models and is currently in clinical testing.
Collapse
|
16
|
Spallone G, Botti E, Costanzo A. Targeted therapy in nonmelanoma skin cancers. Cancers (Basel) 2011; 3:2255-73. [PMID: 24212808 PMCID: PMC3757416 DOI: 10.3390/cancers3022255] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 04/11/2011] [Accepted: 04/26/2011] [Indexed: 12/13/2022] Open
Abstract
Nonmelanoma skin cancer (NMSC) is the most prevalent cancer in light-skinned populations, and includes mainly Basal Cell Carcinomas (BCC), representing around 75% of NMSC and Squamous Cell Carcinomas (SCC). The incidence of these tumors is continuously growing. It was found that the overall number of procedures for NMSC in US rose by 76%, from 1,158,298 in 1992 to 2,048,517 in 2006. Although mortality from NMSC tends to be very low, clearly the morbidity related to these skin cancers is very high. Treatment options for NMSC include both surgical and nonsurgical interventions. Surgery was considered the gold standard therapy, however, advancements in the knowledge of pathogenic mechanisms of NMSCs led to the identification of key targets for drug intervention and to the consequent development of several targeted therapies. These represent the future in treatment of these common forms of cancer ensuring a high cure rate, preservation of the maximal amount of normal surrounding tissue and optimal cosmetic outcome. Here, we will review recent advancements in NMSC targeted therapies focusing on BCC and SCC.
Collapse
Affiliation(s)
- Giulia Spallone
- Department of Dermatology, University of Rome "Tor Vergata", Via Montpellier 1, 00199, Rome, Italy.
| | | | | |
Collapse
|
17
|
Discovery of 2,4-bis-arylamino-1,3-pyrimidines as insulin-like growth factor-1 receptor (IGF-1R) inhibitors. Bioorg Med Chem Lett 2011; 21:2394-9. [PMID: 21414779 DOI: 10.1016/j.bmcl.2011.02.075] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 11/22/2022]
Abstract
The insulin-like growth factor-1 receptor (IGF-1R) plays an important role in the regulation of cell growth and differentiation, and in protection from apoptosis. IGF-1R has been shown to be an appealing target for the treatment of human cancer. Herein, we report the synthesis, structure-activity relationships (SAR), X-ray cocrystal structure and in vivo tumor study results for a series of 2,4-bis-arylamino-1,3-pyrimidines.
Collapse
|
18
|
Galer CE, Corey CL, Wang Z, Younes MN, Gomez-Rivera F, Jasser SA, Ludwig DL, El-Naggar AK, Weber RS, Myers JN. Dual inhibition of epidermal growth factor receptor and insulin-like growth factor receptor I: reduction of angiogenesis and tumor growth in cutaneous squamous cell carcinoma. Head Neck 2011; 33:189-98. [PMID: 20848439 PMCID: PMC3010504 DOI: 10.1002/hed.21419] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (CSCC) is the second most common nonmelanoma skin cancer. Most of the approximately 250,000 cases occurring annually in the United States are small, nonaggressive, and cured by excision alone. However, a subset of these tumors which are defined by poorly differentiated histology, large tumor size, invasion of adjacent structures, and/or regional metastases can prove resistant to treatment despite adjuvant radiotherapy and can have an increased risk of recurrence and nodal metastasis. Novel therapeutic approaches are necessary to improve the outcomes for patients with aggressive CSCC. METHODS We analyzed the effect of targeted therapy on the growth and survival of CSCC cell lines using an anti-insulin-like growth factor-I receptor (IGF-IR) antibody, A12, alone or in combination with an anti-epidermal growth factor receptor (EGFR) antibody, cetuximab, both in vitro and in vivo in an athymic nude mouse model of CSCC. RESULTS Treatment with A12 and cetuximab inhibited the signaling pathways of IGF-IR and EGFR and inhibited proliferation and induced apoptosis of squamous cell carcinoma (SCC) cell lines in vitro. Immunohistochemical staining revealed decreased proliferating cell nuclear antigen (PCNA), microvessel density, and increased apoptosis within the treated tumor xenografts. In addition, the administration of A12, alone or in combination with cetuximab inhibited the growth of tumors by 51% and 92%, respectively, and significantly enhanced survival in the nude mouse model of CSCC (p = .044 and p < .001, respectively). CONCLUSION These data suggest that dual treatment with monoclonal antibodies to the EGFR and IGF-IR may be therapeutically useful in the treatment of CSCC.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Anthracenes/pharmacology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Cetuximab
- ErbB Receptors/antagonists & inhibitors
- Humans
- In Vitro Techniques
- Mice
- Mice, Nude
- Neovascularization, Pathologic/prevention & control
- Receptor, IGF Type 1/antagonists & inhibitors
- Skin Neoplasms/drug therapy
- Skin Neoplasms/pathology
- Treatment Outcome
- Tumor Cells, Cultured/drug effects
Collapse
Affiliation(s)
- Chad E. Galer
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Christina L. Corey
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Zhuoying Wang
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Maher N. Younes
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Fernando Gomez-Rivera
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Samar A. Jasser
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Adel K. El-Naggar
- Department of Head and Neck Surgery, Cancer Hospital of Fudan University, Shanghai, China
| | - Randal S. Weber
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey N. Myers
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Bobby R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Hou X, Huang F, Carboni JM, Flatten K, Asmann YW, Eyck CT, Nakanishi T, Tibodeau JD, Ross DD, Gottardis MM, Erlichman C, Kaufmann SH, Haluska P. Drug efflux by breast cancer resistance protein is a mechanism of resistance to the benzimidazole insulin-like growth factor receptor/insulin receptor inhibitor, BMS-536924. Mol Cancer Ther 2011; 10:117-25. [PMID: 21220496 PMCID: PMC3057506 DOI: 10.1158/1535-7163.mct-10-0438] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Preclinical investigations have identified insulin-like growth factor (IGF) signaling as a key mechanism for cancer growth and resistance to clinically useful therapies in multiple tumor types including breast cancer. Thus, agents targeting and blocking IGF signaling have promise in the treatment of solid tumors. To identify possible mechanisms of resistance to blocking the IGF pathway, we generated a cell line that was resistant to the IGF-1R/InsR benzimidazole inhibitors, BMS-554417 and BMS-536924, and compared expression profiles of the parental and resistant cells lines using Affymetrix GeneChip Human Genome U133 arrays. Compared with MCF-7 cells, breast cancer resistance protein (BCRP) expression was increased 9-fold in MCF-7R4, which was confirmed by immunoblotting and was highly statistically significant (P = 7.13E-09). BCRP was also upregulated in an independently derived resistant cell line, MCF-7 924R. MCF-7R4 cells had significantly lower intracellular accumulation of BMS-536924 compared with MCF-7 cells. Expression of BCRP in MCF-7 cells was sufficient to reduce sensitivity to BMS-536924. Furthermore, knockdown of BCRP in MCF-7R4 cells resensitized cells to BMS-536924. Four cell lines selected for resistance to the pyrrolotriazine IGF-1R/InsR inhibitor, BMS-754807, did not have upregulation of BCRP. These data suggest that benzimidazole IGF-1R/InsR inhibitors may select for upregulation and be effluxed by the ATP-binding cassette transporter, BCRP, contributing to resistance. However, pyrrolotriazine IGF-1R/InsR inhibitors do not appear to be affected by this resistance mechanism.
Collapse
Affiliation(s)
- Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, MN 55905
| | - Fei Huang
- Oncology Drug Discovery, Bristol Myers Squibb Pharmaceutical Research Institute, Princeton, NJ 08534
| | - Joan M. Carboni
- Oncology Drug Discovery, Bristol Myers Squibb Pharmaceutical Research Institute, Princeton, NJ 08534
| | - Karen Flatten
- Department of Oncology, Mayo Clinic, Rochester, MN 55905
| | - Yan W. Asmann
- Division of Biomedical Informatics, Mayo Clinic, Rochester, MN 55905
| | | | - Takeo Nakanishi
- University of Maryland Greenebaum Cancer Center and Baltimore Veterans Medical Center, Baltimore, MD 21201
| | | | - Douglas D. Ross
- University of Maryland Greenebaum Cancer Center and Baltimore Veterans Medical Center, Baltimore, MD 21201
| | - Marco M. Gottardis
- Oncology Drug Discovery, Bristol Myers Squibb Pharmaceutical Research Institute, Princeton, NJ 08534
| | | | | | - Paul Haluska
- Department of Oncology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
20
|
Couderc C, Poncet G, Villaume K, Blanc M, Gadot N, Walter T, Lepinasse F, Hervieu V, Cordier-Bussat M, Scoazec JY, Roche C. Targeting the PI3K/mTOR pathway in murine endocrine cell lines: in vitro and in vivo effects on tumor cell growth. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:336-44. [PMID: 21224070 DOI: 10.1016/j.ajpath.2010.11.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 08/13/2010] [Accepted: 09/09/2010] [Indexed: 11/29/2022]
Abstract
The mammalian target of rapamycin (mTOR) inhibitors, such as rapalogues, are a promising new tool for the treatment of metastatic gastroenteropancreatic endocrine tumors. However, their mechanisms of action remain to be established. We used two murine intestinal endocrine tumoral cell lines, STC-1 and GLUTag, to evaluate the antitumor effects of rapamycin in vitro and in vivo in a preclinical model of liver endocrine metastases. In vitro, rapamycin inhibited the proliferation of cells in the basal state and after stimulation by insulin-like growth factor-1. Simultaneously, p70S6 kinase and 4EBP1 phosphorylation was inhibited. In vivo, rapamycin substantially inhibited the intrahepatic growth of STC-1 cells, irrespectively of the timing of its administration and even when the treatment was administered after cell intrahepatic engraftment. In addition, treated animals had significantly prolonged survival (mean survival time: 47.7 days in treated animals versus 31.8 days in controls) and better clinical status. Rapamycin treatment was associated with a significant decrease in mitotic index and in intratumoral vascular density within STC-1 tumors. Furthermore, the antitumoral effect obtained after treatment with a combination of rapamycin and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 was more significant than with rapamycin alone in both cell lines. Our results suggest that the antitumor efficacy of rapamycin in neuroendocrine tumors results from a combination of antiproliferative and antiangiogenic effects. Interestingly, a more potent antitumor efficiency could be obtained by simultaneously targeting several levels of the PI3K/mTOR pathway.
Collapse
Affiliation(s)
- Christophe Couderc
- INSERM, U865, Faculté Laënnec, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Senthilkumar K, Elumalai P, Arunkumar R, Banudevi S, Gunadharini ND, Sharmila G, Selvakumar K, Arunakaran J. Quercetin regulates insulin like growth factor signaling and induces intrinsic and extrinsic pathway mediated apoptosis in androgen independent prostate cancer cells (PC-3). Mol Cell Biochem 2010; 344:173-84. [PMID: 20658310 DOI: 10.1007/s11010-010-0540-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 07/14/2010] [Indexed: 01/02/2023]
Abstract
Progression of prostate cancer is facilitated by growth factors that activate critical signaling cascades thereby promote prostate cancer cell growth, survival, and migration. To investigate the effect of quercetin on insulin-like growth factor signaling and apoptosis in androgen independent prostate cancer cells (PC-3), IGF-IR, PI-3K, p-Akt, Akt, cyclin D1, Bad, cytochrome c, PARP, caspases-9 and 10 protein levels were assessed by western blot analysis. Mitochondrial membrane potency was detected by rhodamine-123 staining. Quercetin induced caspase-3 activity assay was performed for activation of apoptosis. Further, RT-PCR was also performed for Bad, IGF-I, II, IR, and IGFBP-3 mRNA expression. Quercetin significantly increases the proapoptotic mRNA levels of Bad, IGFBP-3 and protein levels of Bad, cytochrome C, cleaved caspase-9, caspase-10, cleaved PARP and caspase-3 activity in PC-3 cells. IGF-IRβ, PI3K, p-Akt, and cyclin D1 protein expression and mRNA levels of IGF-I, II and IGF-IR were decreased significantly. Further, treatment with PI3K inhibitor (LY294002) and quercetin showed decreased p-Akt levels. Apoptosis is confirmed by loss of mitochondrial membrane potential in quercetin treated PC-3 cells. This study suggests that quercetin decreases the survival of androgen independent prostate cancer cells by modulating the expression of insulin-like growth factors (IGF) system components, signaling molecules and induces apoptosis, which could be very useful for the androgen independent prostate cancer treatment.
Collapse
Affiliation(s)
- Kalimuthu Senthilkumar
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, Tamilnadu, India.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Geoerger B, Brasme JF, Daudigeos-Dubus E, Opolon P, Venot C, Debussche L, Vrignaud P, Vassal G. Anti-insulin-like growth factor 1 receptor antibody EM164 (murine AVE1642) exhibits anti-tumour activity alone and in combination with temozolomide against neuroblastoma. Eur J Cancer 2010; 46:3251-62. [PMID: 20591650 DOI: 10.1016/j.ejca.2010.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 05/28/2010] [Accepted: 06/02/2010] [Indexed: 12/16/2022]
Abstract
Insulin-like growth factor 1 receptor (IGF-1R) is overexpressed in many tumours and contributes to tumourigenicity, cell proliferation, metastasis and resistance, thus representing a promising therapeutic target. The human IGF-1R antagonistic monoclonal antibody EM164 (murine AVE1642) has shown activity in adult cancers and is being evaluated in patients with advanced malignancies. We investigated the EM164 for its therapeutic potential against childhood neuroblastoma. EM164 at 0.07, 0.7 and 7 μg/mL exhibited anti-proliferative activity against all nine cell lines tested in (3)H-thymidine incorporation assay in vitro. Cell proliferation after EM164 exposure ranged between 24% and 80% compared to controls. Sensitivity was independent from culture serum conditions, intensity of IGF-1R expression and IGF-II secretion, although associated with inhibition of AKT activation. In vivo, EM164 administered intravenously at 40 mg/kg twice weekly for 4 weeks yielded significant tumour growth delays (TGD) of 13.4d in advanced stage IGR-N91 and 12.9 d in SK-N-AS tumours compared to controls (p = 0.02 and p = 0.0059, respectively). Simultaneous treatment of EM164 0.7 μg/mL and temozolomide resulted in enhanced activity in vitro. In vivo, treatment with temozolomide at the maximum tolerated dose (100mg/kg/d for 5 consecutive days) and EM164 yielded a significantly greater TGD of 29.1d (p<0.01) and two complete tumour regressions (CR) compared to 18.1d (p = ns) and one CR for EM164 alone and 16.1d (p = ns) for temozolomide alone. Our results demonstrate the potential of the anti-IGF-1R antibody alone and in combination with alkylating agents and support the therapeutic development of the AVE1642 for aggressive neuroblastoma.
Collapse
Affiliation(s)
- Birgit Geoerger
- UPRES EA 3535, Pharmacology and New Treatments of Cancer, Université Paris-Sud XI, Institut Gustave Roussy, 94805 Villejuif, France.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Nemecek C, Metz WA, Wentzler S, Ding FX, Venot C, Souaille C, Dagallier A, Maignan S, Guilloteau JP, Bernard F, Henry A, Grapinet S, Lesuisse D. Design of Potent IGF1-R Inhibitors Related to Bis-azaindoles. Chem Biol Drug Des 2010; 76:100-6. [DOI: 10.1111/j.1747-0285.2010.00991.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
Tonini G, Virzì V, Fratto ME, Vincenzi B, Santini D. Targeted therapy in biliary tract cancer: 2009 update. Future Oncol 2010; 5:1675-84. [PMID: 20001803 DOI: 10.2217/fon.09.130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Biliary tract cancers (BTCs) include cholangiocarcinoma (intrahepatic, perihilar and extrahepatic), carcinoma of the gall bladder and ampullary carcinoma. In patients with advanced disease the prognosis is poor. There is not a consensus regarding treatment strategy. Chemotherapy has only limited efficacy. This review summarizes the new approaches for BTC patients and the rationale for targeted therapies. The prognostic factors and the molecular features of BTC are analyzed. The clinical trials evaluating the targeted agents are accurately described, especially those assessing the role of anti-EGFR and antiangiogenic drugs. The ongoing trials are also analyzed. In fact, only the results of these trials will establish which is the most effective agent or combination for this setting.
Collapse
Affiliation(s)
- Giuseppe Tonini
- Department of Medical Oncology, University Campus Bio-Medico, 00128 Rome, Italy.
| | | | | | | | | |
Collapse
|
25
|
Fluorescent tumour imaging of type I IGF receptor in vivo: comparison of antibody-conjugated quantum dots and small-molecule fluorophore. Br J Cancer 2009; 101:71-9. [PMID: 19491901 PMCID: PMC2713715 DOI: 10.1038/sj.bjc.6605103] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background: The type I insulin-like growth factor receptor (IGF1R) is a transmembrane tyrosine kinase involved in cancer proliferation, survival, and metastasis. Methods: In this study, we used two different fluorescent technologies (small-molecule fluorophores and quantum dot (QD) nanoparticles) to detect receptor expression and its downregulation by antibodies in vivo. Results: After conjugation with AVE-1642, a humanised anti-IGF1R monoclonal antibody, both QDs (705 nm) or Alexa 680 (small-molecule fluorophore) detected expression and downregulation of IGF1R in vitro. To examine their utility in vivo, either AVE-1642 conjugates were intravenously delivered to mice bearing xenograft tumours of mouse embryo fibroblasts expressing human IGF1R or MCF-7 human breast cancer cells. Quantum dot fluorescence was mainly localised to the reticuloendothelial system in several organs and engulfed by macrophages, with only very small amount of QDs detected in the xenograft tumours. Depletion of macrophages by clodronate liposomes did not alter the nonspecific uptake of QDs. In contrast, AVE-1642-conjugated Alexa 680 solely targeted to xenograft tumour and was able to detect IGF1R downregulation, with little nonspecific targeting to other tissues or organs in mice. Conclusion: Taken together, our data suggest that small-molecule fluorophores, not QDs, are suitable to detect the expression and downregulation of IGF1R in vivo.
Collapse
|
26
|
Abstract
Cancer metastasis is the result of complex processes, including alteration of cell adhesion/motility in the microenvironment and neoangiogenesis, that are necessary to support cancer growth in tissues distant from the primary tumor. The molecular chaperone heat-shock protein 90 (Hsp90), also termed the 'cancer chaperone', plays a crucial role in maintaining the stability and activity of numerous signaling proteins involved in these processes. Small-molecule Hsp90 inhibitors display anticancer activity both in vitro and in vivo, and multiple Phase II and Phase III clinical trials of several structurally distinct Hsp90 inhibitors are currently underway. In this review, we will highlight the importance of Hsp90 in cancer metastasis and the therapeutic potential of Hsp90 inhibitors as antimetastasis drugs.
Collapse
Affiliation(s)
- Shinji Tsutsumi
- Urologic Oncology Branch, National Cancer Institute, 9000 Rockville Pike, Bldg. 10/CRC, 1-5940, Bethesda, MD, 20892-1107, USA
| | - Kristin Beebe
- Urologic Oncology Branch, National Cancer Institute, 9000 Rockville Pike, Bldg. 10/CRC, 1-5940, Bethesda, MD, 20892-1107, USA
| | - Len Neckers
- Urologic Oncology Branch, National Cancer Institute, 9000 Rockville Pike, Bldg. 10/CRC, 1-5940, Bethesda, MD, 20892-1107, USA Tel.: +1 301 496 5899 Fax: +1 301 402 0922
| |
Collapse
|
27
|
From biology to clinical experience: evolution in the knowledge of neuroendocrine tumours. Oncol Rev 2009. [DOI: 10.1007/s12156-009-0011-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
28
|
Law JH, Habibi G, Hu K, Masoudi H, Wang MYC, Stratford AL, Park E, Gee JMW, Finlay P, Jones HE, Nicholson RI, Carboni J, Gottardis M, Pollak M, Dunn SE. Phosphorylated insulin-like growth factor-i/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Cancer Res 2009; 68:10238-46. [PMID: 19074892 DOI: 10.1158/0008-5472.can-08-2755] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Drugs that target the insulin-like growth factor-I receptor (IGF-IR) and/or insulin receptor (IR) are currently under investigation for a variety of malignancies including breast cancer. Although we have previously reported that IGF-IR expression in primary breast tumors is common, the activation status of this receptor has not been examined in relation to survival. Phosphorylated IGF-IR/IR (P-IGF-IR/IR) and its downstream signaling partner phospho-S6 (P-S6) were evaluated immunohistochemically in tumor tissue microarrays representing 438 cases of invasive breast cancer. P-IGF-IR/IR (n = 114; P = 0.046) and total levels of IR (n = 122; P = 0.009) were indicative of poor survival, whereas total IGF-IR (n = 112; P = 0.304) was not. P-IGF-IR/IR and P-S6 were coordinately expressed in primary breast tumors (likelihood ratio, 11.57; P = 6.70 x 10(-4)). Importantly, P-IGF-IR/IR was detected in all breast cancer subtypes (luminal, 48.1%; triple negative, 41.9%; and HER2, 64.3%). In vitro, the IGF-IR/IR inhibitor BMS-536924 decreased phospho-RSK and P-S6, and significantly suppressed the growth of breast cancer cell lines MCF-7, SUM149, and AU565 representing the luminal, triple negative, and HER2 subtypes, respectively, in monolayer and soft agar. BMS-536924 also inhibited growth in tamoxifen resistant MCF-7 Tam-R cells while having little effect on immortalized normal breast epithelial cells. Thus, we can determine which patients have the activated receptor and provide evidence that P-IGF-IR/IR is a prognostic factor for breast cancer. Beyond this, P-IGF-IR/IR could be a predictive marker for response to IGF-IR and/or IR-targeted therapies, as these inhibitors may be of benefit in all breast cancer subtypes including those with acquired resistance to tamoxifen.
Collapse
Affiliation(s)
- Jennifer H Law
- Department of Pediatrics, Laboratory for Oncogenomic Research, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Höpfner M, Schuppan D, Scherübl H. Targeted medical therapy of biliary tract cancer: recent advances and future perspectives. World J Gastroenterol 2008; 14:7021-32. [PMID: 19084910 PMCID: PMC2776833 DOI: 10.3748/wjg.14.7021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Revised: 11/12/2008] [Accepted: 11/19/2008] [Indexed: 02/06/2023] Open
Abstract
The limited efficacy of cytotoxic therapy for advanced biliary tract and gallbladder cancers emphasizes the need for novel and more effective medical treatment options. A better understanding of the specific biological features of these neoplasms led to the development of new targeted therapies, which take the abundant expression of several growth factors and cognate tyrosine kinase receptors into account. This review will briefly summarize the status and future perspectives of antiangiogenic and growth factor receptor-based pharmacological approaches for the treatment of biliary tract and gallbladder cancers. In view of multiple novel targeted approaches, the rationale for innovative therapies, such as combinations of growth factor (receptor)-targeting agents with cytotoxic drugs or with other novel anticancer drugs will be highlighted.
Collapse
|
30
|
Girnita A, All-Ericsson C, Economou MA, Aström K, Axelson M, Seregard S, Larsson O, Girnita L. The insulin-like growth factor-I receptor inhibitor picropodophyllin causes tumor regression and attenuates mechanisms involved in invasion of uveal melanoma cells. Acta Ophthalmol 2008; 86 Thesis 4:26-34. [PMID: 19032679 DOI: 10.1111/j.1755-3768.2008.01183.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE Uveal melanoma has a high mortality rate due to a high incidence of metastasis (up to 50%) which preferentially occurs in the liver. Conventional chemotherapy being the only therapeutic option today against metastatic uveal melanoma, has not proved to be effective. Therefore, new molecular targets important for malignant phenotype of uveal melanoma have to be found to design efficient pharmacologic agents. EXPERIMENTAL DESIGN We previously reported data indicating that the insulin-like growth factor-1 receptor (IGF-IR) is a metastasis predictor as well as a therapeutic target for uveal melanoma. In the present study, we made use of the cyclolignan picropodophyllin (PPP), which is an inhibitor of the IGF-IR. RESULT We showed that PPP efficiently block growth and viability of uveal melanoma cells in cultures and causes tumor regression in xenografted mice. In addition, treatment with PPP inhibited several mechanism involved in metastasis, including tumor cells adhesion to extracellular matrix proteins, activity and expression of matrix metalloproteinase 2, and cell migration as well as invasion through basement membranes and endothelial cell layer. Furthermore, PPP significantly delayed established of uveal melanoma tumor and drastically reduced the incidence of liver metastasis in mice. CONCLUSIONS Our data suggest that IGR-IR is crucial for growth and survival as well as invasion and metastasis of uveal melanoma cells. Targeting this receptor may therefore comprise a strategy to treat ongoing disease (today incurable) as well as a strategy to prevent development of metastases in patients with primary disease.
Collapse
Affiliation(s)
- Ada Girnita
- Cellular and Molecular Tumor Pathology, Department of Oncology and Pathology, Cancer Centre Karolinska R8:04, Karolinska Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Haluska P, Carboni JM, TenEyck C, Attar RM, Hou X, Yu C, Sagar M, Wong TW, Gottardis MM, Erlichman C. HER receptor signaling confers resistance to the insulin-like growth factor-I receptor inhibitor, BMS-536924. Mol Cancer Ther 2008; 7:2589-98. [PMID: 18765823 PMCID: PMC2614316 DOI: 10.1158/1535-7163.mct-08-0493] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We have reported previously the activity of the insulin-like growth factor-I (IGF-IR)/insulin receptor (InsR) inhibitor, BMS-554417, in breast and ovarian cancer cell lines. Further studies indicated treatment of OV202 ovarian cancer cells with BMS-554417 increased phosphorylation of HER-2. In addition, treatment with the pan-HER inhibitor, BMS-599626, resulted in increased phosphorylation of IGF-IR, suggesting a reciprocal cross-talk mechanism. In a panel of five ovarian cancer cell lines, simultaneous treatment with the IGF-IR/InsR inhibitor, BMS-536924 and BMS-599626, resulted in a synergistic antiproliferative effect. Furthermore, combination therapy decreased AKT and extracellular signal-regulated kinase activation and increased biochemical and nuclear morphologic changes consistent with apoptosis compared with either agent alone. In response to treatment with BMS-536924, increased expression and activation of various members of the HER family of receptors were seen in all five ovarian cancer cell lines, suggesting that inhibition of IGF-IR/InsR results in adaptive up-regulation of the HER pathway. Using MCF-7 breast cancer cell variants that overexpressed HER-1 or HER-2, we then tested the hypothesis that HER receptor expression is sufficient to confer resistance to IGF-IR-targeted therapy. In the presence of activating ligands epidermal growth factor or heregulin, respectively, MCF-7 cells expressing HER-1 or HER-2 were resistant to BMS-536924 as determined in a proliferation and clonogenic assay. These data suggested that simultaneous treatment with inhibitors of the IGF-I and HER family of receptors may be an effective strategy for clinical investigations of IGF-IR inhibitors in breast and ovarian cancer and that targeting HER-1 and HER-2 may overcome clinical resistance to IGF-IR inhibitors.
Collapse
Affiliation(s)
- Paul Haluska
- Department of Oncology, Mayo Clinic, 200 First Street, South West, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hubbard RD, Wilsbacher JL. Advances towards the development of ATP-competitive small-molecule inhibitors of the insulin-like growth factor receptor (IGF-IR). ChemMedChem 2008; 2:41-6. [PMID: 17089440 DOI: 10.1002/cmdc.200600174] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Robert D Hubbard
- Cancer Research, Global Pharmaceutical Research and Development, Abbott Laboratories, Abbott Park, Illinois 60064, USA.
| | | |
Collapse
|
33
|
Biochemical constitution of extracellular medium is critical for control of human breast cancer MDA-MB-231 cell motility. J Membr Biol 2008; 223:27-36. [PMID: 18575796 DOI: 10.1007/s00232-008-9110-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Accepted: 05/07/2008] [Indexed: 01/04/2023]
Abstract
Although voltage-gated sodium channel (VGSC) activity, upregulated significantly in strongly metastatic human breast cancer cells, has been found to potentiate a variety of in vitro metastatic cell behaviors, the mechanism(s) regulating channel expression/activity is not clear. As a step toward identifying possible serum factors that might be responsible for this, we tested whether medium in which fetal bovine serum (FBS) was substituted with a commercial serum replacement agent (SR-2), comprising insulin and bovine serum albumin, would influence the VGSC-dependent in vitro metastatic cell behaviors. Human breast cancer MDA-MB-231 cells were used as a model. Measurements of lateral motility, transverse migration and adhesion showed consistently that the channel's involvement in metastatic cell behaviors depended on the extracellular biochemical conditions. In normal medium (5% FBS), tetrodotoxin (TTX), a highly specific blocker of VGSCs, suppressed these cellular behaviors, as reported before. In contrast, in SR-2 medium, TTX had opposite effects. However, blocking endogenous insulin/insulin-like growth factor receptor signaling with AG1024 eliminated or reversed the anomalous effects of TTX. Insulin added to serum-free medium increased migration, and TTX increased it further. In conclusion, (1) the biochemical constitution of the extracellular medium had a significant impact upon breast cancer cells' in vitro metastatic behaviors and (2) insulin, in particular, controlled the mode of the functional association between cells' VGSC activity and metastatic machinery.
Collapse
|
34
|
Höpfner M, Schuppan D, Scherübl H. Treatment of gastrointestinal neuroendocrine tumors with inhibitors of growth factor receptors and their signaling pathways: recent advances and future perspectives. World J Gastroenterol 2008; 14:2461-73. [PMID: 18442192 PMCID: PMC2708356 DOI: 10.3748/wjg.14.2461] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 03/15/2008] [Indexed: 02/06/2023] Open
Abstract
The limited efficacy of conventional cytotoxic treatment regimes for advanced gastrointestinal neuroendocrine cancers emphasizes the need for novel and more effective medical treatment options. Recent findings on the specific biological features of this family of neoplasms has led to the development of new targeted therapies, which take into account the high vascularization and abundant expression of specific growth factors and cognate tyrosine kinase receptors. This review will briefly summarize the status and future perspectives of antiangiogenic, mTOR- or growth factor receptor-based pharmacological approaches for the innovative treatment of gastrointestinal neuroendocrine tumors. In view of the multitude of novel targeted approaches, the rationale for innovative combination therapies, i.e. combining growth factor (receptor)-targeting agents with chemo- or biotherapeutics or with other novel anticancer drugs such as HDAC or proteasome inhibitors will be taken into account.
Collapse
|
35
|
Zhang H, Sachdev D, Wang C, Hubel A, Gaillard-Kelly M, Yee D. Detection and downregulation of type I IGF receptor expression by antibody-conjugated quantum dots in breast cancer cells. Breast Cancer Res Treat 2008; 114:277-85. [PMID: 18418709 DOI: 10.1007/s10549-008-0014-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 04/07/2008] [Indexed: 11/29/2022]
Abstract
The type I insulin-like growth factor (IGF) receptor (IGF1R) is a transmembrane tyrosine kinase involved in breast cancer proliferation, survival, and metastasis. Several monoclonal antibodies directed against the receptor are in clinical trials. In order to develop a methodology to detect and measure IGF1R levels in breast cancer cells, we covalently conjugated an IGF1R antibody, AVE-1642, with quantum dots (Qdots), which are nanocrystals that emit fluorescence upon excitation. AVE-1642 Qdots only bound to cells that express IGF1R, and measured IGF1R levels by fluorescence emission at 655 nm. After binding to the cell surface, AVE-1642 Qdots underwent receptor mediated endocytosis, localized to endosome, and later translocated into the nucleus. Treating MCF-7 cells with AVE-1642 Qdots, but not unconjugated Qdots alone, downregulated IGF1R levels and rendered cells refractory to IGF-I stimulation. Furthermore, cell proliferation was slightly inhibited by AVE-1642 Qdots, but not the unconjugated Qdots. Our data suggest that AVE-1642 Qdots can be used to detect IGF1R expression and measure changes in cell surface receptor levels. In addition, the inhibitory effect of AVE-1642 Qdots to cell proliferation implies that it may serve as a traceable therapeutic agent.
Collapse
Affiliation(s)
- Hua Zhang
- University of Minnesota Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
36
|
Wang Z, Chakravarty G, Kim S, Yazici YD, Younes MN, Jasser SA, Santillan AA, Bucana CD, El-Naggar AK, Myers JN. Growth-inhibitory effects of human anti-insulin-like growth factor-I receptor antibody (A12) in an orthotopic nude mouse model of anaplastic thyroid carcinoma. Clin Cancer Res 2007; 12:4755-65. [PMID: 16899627 DOI: 10.1158/1078-0432.ccr-05-2691] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The insulin-like growth factor-I receptor (IGF-IR) and its ligands have been implicated in the pathogenesis and progression of various cancers, including those arising in the thyroid gland. We therefore evaluated whether the IGF-IR could serve as a potential target for therapy of anaplastic thyroid carcinoma (ATC). EXPERIMENTAL DESIGN The expression and activation of the IGF-IR and some of its downstream signaling pathway components were evaluated in both human thyroid cancer specimens and thyroid cancer cell lines. The therapeutic potential of a humanized monoclonal antibody (A12) directed against IGF-IR was assessed in vitro and in vivo in an orthotopic model of ATC. Tumor volume and overall survival time were analyzed to evaluate the efficacy of A12 in vivo. RESULTS IGF-IR was overexpressed in 94% of the thyroid cancers. Blockade of IGF-IR with A12 was effective in attenuating IGF-IR signaling both in vitro and in vivo. However, the inhibitory effects of A12 on cell proliferation were cell line dependent, as those ATC cell lines that had detectable levels of pIGF-IR were more sensitive to A12 treatment. A12 was equally effective in vivo, where it brought approximately 57% (P = 0.041) inhibition in tumor volume. The concomitant use of A12 and irinotecan produced additive effects and resulted in a 93% (P < 0.001) reduction in tumor volume. Blocking IGF-IR blocked Akt phosphorylation and decreased proliferation and microvessel density but increased apoptosis within the tumor xenografts. Our results also highlighted a previously undefined IGF-IR-mediated antiangiogenic effect on tumor-associated endothelium in thyroid cancers. CONCLUSION Blocking the IGF-IR with A12 seems to be a potential avenue for treating patients with ATC by its direct antitumor effects and its effects on the tumor vasculature.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigen-Antibody Reactions
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Carcinoma/drug therapy
- Carcinoma/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Humans
- Insulin-Like Growth Factor I/pharmacology
- Insulin-Like Growth Factor II/pharmacology
- Male
- Methylation
- Mice
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/biosynthesis
- Receptor, IGF Type 1/immunology
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Structure-Activity Relationship
- Thyroid Neoplasms/drug therapy
- Thyroid Neoplasms/pathology
- Transplantation, Heterologous
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Zhuoying Wang
- Department of Head and Neck Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, 77030-4009, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ji QS, Mulvihill MJ, Rosenfeld-Franklin M, Cooke A, Feng L, Mak G, O'Connor M, Yao Y, Pirritt C, Buck E, Eyzaguirre A, Arnold LD, Gibson NW, Pachter JA. A novel, potent, and selective insulin-like growth factor-I receptor kinase inhibitor blocks insulin-like growth factor-I receptor signaling in vitro and inhibits insulin-like growth factor-I receptor dependent tumor growth in vivo. Mol Cancer Ther 2007; 6:2158-67. [PMID: 17671083 DOI: 10.1158/1535-7163.mct-07-0070] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin-like growth factor-I receptor (IGF-IR) and its ligands, IGF-I and IGF-II, are up-regulated in a variety of human cancers. In tumors, such as colorectal, non-small cell lung, ovarian, and pediatric cancers, which may drive their own growth and survival through autocrine IGF-II expression, the role of IGF-IR is especially critical. Here, we present a novel small-molecule IGF-IR kinase inhibitor, cis-3-[3-(4-methyl-piperazin-l-yl)-cyclobutyl]-1-(2-phenyl-quinolin-7-yl)-imidazo[1,5-a]pyrazin-8-ylamine (PQIP), which displayed a cellular IC(50) of 19 nmol/L for inhibition of ligand-dependent autophosphorylation of human IGF-IR with 14-fold cellular selectivity relative to the human insulin receptor. PQIP showed minimal activity against a panel of 32 other protein kinases. It also abolished the ligand-induced activation of downstream phosphorylated AKT and phosphorylated extracellular signal-regulated kinase 1/2 in both IGF-IR transfectant cells and a GEO human colorectal cancer cell line. Analysis of GEO cells revealed a significant level of both phosphorylated IGF-IR and IGF-II expression. Furthermore, inactivation of IGF-II in conditioned GEO culture medium by a neutralizing antibody diminished IGF-IR activation, indicating the presence of a functional IGF-II/IGF-IR autocrine loop in GEO cells. Once daily oral dosing of PQIP induced robust antitumor efficacy in GEO xenografts. The antitumor efficacy correlated with the degree and duration of inhibition of tumor IGF-IR phosphorylation in vivo by this compound. Moreover, when mice were treated for 3 days with a dose of PQIP that maximally inhibited tumor growth, only minor changes in blood glucose were observed. Thus, PQIP represents a potent and selective IGF-IR kinase inhibitor that is especially efficacious in an IGF-II-driven human tumor model.
Collapse
Affiliation(s)
- Qun-sheng Ji
- Oncology, OSI Pharmaceuticals, Inc., New York, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Barnes CJ, Ohshiro K, Rayala SK, El-Naggar AK, Kumar R. Insulin-like Growth Factor Receptor as a Therapeutic Target in Head and Neck Cancer. Clin Cancer Res 2007; 13:4291-9. [PMID: 17634559 DOI: 10.1158/1078-0432.ccr-06-2040] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Insulin-like growth factor type I receptor (IGF-IR) plays critical roles in epithelial cancer cell development, proliferation, motility, and survival, and new therapeutic agents targeting IGF-IR are in development. Another receptor tyrosine kinase, the epidermal growth factor receptor (EGFR), is an established therapeutic target in head and neck cancer and IGF-IR/EGFR heterodimerization has been reported in other epithelial cancers. The present study was undertaken to determine the effects of anti-IGF-IR therapeutic targeting on cell signaling and cancer cell phenotypes in squamous cell carcinomas of the head and neck (SCCHN). EXPERIMENTAL DESIGN The therapeutic efficacy of the human anti-IGF-IR antibody IMC-A12 alone and in combination with the EGFR blocking antibody cetuximab (C225) was tested in SCCHN cell lines and in tumor xenografts. RESULTS IGF-IR was overexpressed in human head and neck cancer cell lines and tumors. Pretreatment of serum-starved 183A or TU159 SCCHN cell lines with A12 (10 microg/mL) blocked IGF-stimulated activation of IGF-IR, insulin receptor substrate (IRS)-1 and IRS-2, mitogen-activated protein kinase, and phosphatidylinositol 3-kinase. A12 induced G(0)-G(1) cell cycle arrest and blocked cell growth, motility, and anchorage-independent growth. Stimulation of head and neck cancer cells with either IGF or EGF resulted in IGF-IR and EGFR heterodimerization, but only IGF caused activating phosphorylation of both receptors. Combined treatment with A12 and the EGFR blocking antibody C225 was more effective at reducing cell proliferation and migration than either agent alone. Finally, TU159 tongue cancer cell xenografts grown in athymic nude mice were treated thrice weekly for 4 weeks with vehicle, A12 (40 mg/kg i.p.), C225 (40 mg/kg i.p.), or both agents (n=8 mice per group; 2 tumors per mouse). Linear regression slope analysis showed significant differences in median tumor volume over time between all three treatment groups and the control group. Complete regression was seen in 31% (A12), 31% (C225), and 44% (A12 + C225) of tumors. CONCLUSION Here we found the overexpression of IGF-IR, the functional heterodimerization of IGF-IR and EGFR, and effective therapeutic targeting of these receptors in human head and neck cancer xenografts.
Collapse
Affiliation(s)
- Christopher J Barnes
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
39
|
de Bono JS, Attard G, Adjei A, Pollak MN, Fong PC, Haluska P, Roberts L, Melvin C, Repollet M, Chianese D, Connely M, Terstappen LWMM, Gualberto A. Potential applications for circulating tumor cells expressing the insulin-like growth factor-I receptor. Clin Cancer Res 2007; 13:3611-6. [PMID: 17575225 DOI: 10.1158/1078-0432.ccr-07-0268] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To detect insulin-like growth factor-IR (IGF-IR) on circulating tumor cells (CTC) as a biomarker in the clinical development of a monoclonal human antibody, CP-751,871, targeting IGF-IR. EXPERIMENTAL DESIGN An automated sample preparation and analysis system for enumerating CTCs (CellTracks) was adapted for detecting IGF-IR-positive CTCs with a diagnostic antibody targeting a different IGF-IR epitope to CP-751,871. This assay was used in three phase I trials of CP-751,871 as a single agent or with chemotherapy and was validated using cell lines and blood samples from healthy volunteers and patients with metastatic carcinoma. RESULTS There was no interference between the analytic and therapeutic antibodies. Eighty patients were enrolled on phase I studies of CP-751,871, with 47 (59%) patients having CTCs detected during the study. Before treatment, 26 patients (33%) had CTCs, with 23 having detectable IGF-IR-positive CTCs. CP-751,871 alone, and CP-751,871 with cytotoxic chemotherapy, decreased CTCs and IGF-IR-positive CTCs; these increased toward the end of the 21-day cycle in some patients, falling again with retreatment. CTCs were commonest in advanced hormone refractory prostate cancer (11 of 20). Detectable IGF-IR expression on CTCs before treatment with CP-751,871 and docetaxel was associated with a higher frequency of prostate-specific antigen decline by >50% (6 of 10 versus 2 of 8 patients). A relationship was observed between sustained decreases in CTC counts and prostate-specific antigen declines by >50%. CONCLUSIONS IGF-IR expression is detectable by immunofluorescence on CTCs. These data support the further evaluation of CTCs in pharmacodynamic studies and patient selection, particularly in advanced prostate cancer.
Collapse
|
40
|
Velaparthi U, Liu P, Balasubramanian B, Carboni J, Attar R, Gottardis M, Li A, Greer A, Zoeckler M, Wittman MD, Vyas D. Imidazole moiety replacements in the 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one inhibitors of insulin-like growth factor receptor-1 (IGF-1R) to improve cytochrome P450 profile. Bioorg Med Chem Lett 2007; 17:3072-6. [PMID: 17398093 DOI: 10.1016/j.bmcl.2007.03.048] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 03/13/2007] [Accepted: 03/14/2007] [Indexed: 11/30/2022]
Abstract
A series of 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one inhibitors of insulin-like growth factor receptor-1 (IGF-1R) were examined in which the pendant imidazole moiety was replaced to improve selectivity for IGF-1R inhibition over cytochrome P450 (CYP). Synthesis and SAR of these compounds is presented.
Collapse
Affiliation(s)
- Upender Velaparthi
- Department of Discovery Chemistry, Bristol-Myers Squibb Pharmaceutical Research Institute, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yin D, Vreeland F, Schaaf LJ, Millham R, Duncan BA, Sharma A. Clinical pharmacodynamic effects of the growth hormone receptor antagonist pegvisomant: implications for cancer therapy. Clin Cancer Res 2007; 13:1000-9. [PMID: 17289896 DOI: 10.1158/1078-0432.ccr-06-1910] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE The present study evaluated and compared the efficacy of pegvisomant and octreotide in blocking the growth hormone (GH) axis in humans based on pharmacodynamic biomarkers associated with the GH axis. The study also evaluated the safety of pegvisomant given at high s.c. doses for 14 days. EXPERIMENTAL DESIGN Eighty healthy subjects were enrolled in five cohorts: cohorts 1 to 3, s.c. pegvisomant at 40, 60, or 80 mg once dailyx14 days (n=18 per cohort); cohort 4, s.c. octreotide at 200 microg thrice dailyx14 days (n=18); and cohort 5, untreated control (n=8). Serial blood samples were collected to measure plasma concentrations of total insulin-like growth factor type I (IGF-I), free IGF-I, IGF-II, IGF-binding protein 3 (IGFBP-3), and GH in all subjects and serum pegvisomant concentrations in subjects of cohorts 1 to 3. All subjects receiving treatment were monitored for adverse events (AE). RESULTS After s.c. dosing of pegvisomant once daily for 14 days, the mean maximum suppression values of total IGF-I were 57%, 60%, and 62%, at 40, 60, and 80 mg dose levels, respectively. The maximum suppression was achieved approximately 7 days after the last dose and was sustained for approximately 21 days. Pegvisomant also led to a sustained reduction in free IGF-I, IGFBP-3, and IGF-II concentrations by up to 33%, 46%, and 35%, respectively, and an increase in GH levels. In comparison, octreotide resulted in a considerably weaker inhibition of total IGF-I and IGFBP-3 for a much shorter duration, and no inhibition of IGF-II. AEs in pegvisomant-treated subjects were generally either grade 1 or 2. The most frequent treatment-related AEs included injection site reactions, headache, and fatigue. CONCLUSIONS Pegvisomant at well-tolerated s.c. doses was considerably more efficacious than octreotide in suppressing the GH axis, resulting in substantial and sustained inhibition of circulating IGF-I, IGF-II, and IGFBP-3 concentrations. These results provide evidence in favor of further testing the hypothesis that pegvisomant, through blocking the GH receptor-mediated signal transduction pathways, could be effective in treating tumors that may be GH, IGF-I, and/or IGF-II dependent, such as breast and colorectal cancer.
Collapse
Affiliation(s)
- Donghua Yin
- Global Research and Development, Groton/New London Laboratories, Pfizer, Inc., New London, Connecticut 06320, USA
| | | | | | | | | | | |
Collapse
|
42
|
Velaparthi U, Wittman M, Liu P, Stoffan K, Zimmermann K, Sang X, Carboni J, Li A, Attar R, Gottardis M, Greer A, Chang CY, Jacobsen BL, Sack JS, Sun Y, Langley DR, Balasubramanian B, Vyas D. Discovery and initial SAR of 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-ones as inhibitors of insulin-like growth factor 1-receptor (IGF-1R). Bioorg Med Chem Lett 2007; 17:2317-21. [PMID: 17317169 DOI: 10.1016/j.bmcl.2007.01.102] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 01/12/2007] [Accepted: 01/18/2007] [Indexed: 11/29/2022]
Abstract
The discovery and synthesis of 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one inhibitors of insulin-like growth factor 1-receptor (IGF-1R) are presented. Installing amine containing side chains at the 4-position of pyridone ring significantly improved the enzyme potency. SAR and biological activity of these compounds is presented.
Collapse
Affiliation(s)
- Upender Velaparthi
- Department of Discovery Chemistry, Bristol-Myers Squibb Pharmaceutical Research Institute, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cosaceanu D, Carapancea M, Alexandru O, Budiu R, Martinsson HS, Starborg M, Vrabete M, Kanter L, Lewensohn R, Dricu A. Comparison of three approaches for inhibiting insulin-like growth factor I receptor and their effects on NSCLC cell lines in vitro. Growth Factors 2007; 25:1-8. [PMID: 17454144 DOI: 10.1080/08977190600702865] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The insulin-like growth factor-1 receptor (IGF-1R) mitogenic signaling mediates malignant cell survival by many complex and redundant pathways. This study compared the effects of IGF-1R inhibition on viability and apoptosis of two NSCLC cell lines, using three different methods for the impairment of IGF-1R function: (IR3, an anti-IGF-1R antibody; tyrphostin AG1024, a tyrosine kinase inhibitor (TKI) and IGF-1R-small interfering RNA (siRNA). IGF-1R inhibition led to a decrease of cell survival and induced apoptosis in a manner depending on the approach used for the receptor inhibition. To find an explanation, we analyzed the effects of these treatments on three major antiapoptotic pathways evoked by IGF-1R signaling: IRS-1, Shc and 14.3.3-dependent mitochondrial translocation of Raf-1 kinase (mitRaf). (IR3 downregulated IRS-1 phosphorylation in A549 cells and Shc phosphorylation in U1810 cells. While in A549 cells AG1024 treatment decreased both IRS-1 and Shc phosphorylation, in U1810 cells the IRS-1 phosphorylation was only slightly affected and the Shc phosphorylation drastically downregulated. Neither (IR3 nor AG1024 had any effect on Raf-1 kinase translocation. Irrespective of the cell line, IGF-1R-siRNA treatment induced downregulation of both IRS-1 and Shc phosphorylation coupled with the abrogation of mitRaf. In addition, the IGF-1R-siRNA proved to be the most potent inducer of apoptosis suggesting that more than one antiapoptotic pathway in IGF-1R signaling should be inhibited to effectively induce apoptosis in lung cancer cells.
Collapse
Affiliation(s)
- Daria Cosaceanu
- Department of Oncology-Pathology, Cancer Center Karolinska and Radiumhemmet Karolinska Institute/Hospital. R8:00, Stockholm, S-171 76. Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Shevah O, Laron Z. Patients with congenital deficiency of IGF-I seem protected from the development of malignancies: a preliminary report. Growth Horm IGF Res 2007; 17:54-57. [PMID: 17166755 DOI: 10.1016/j.ghir.2006.10.007] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2006] [Revised: 09/27/2006] [Accepted: 10/31/2006] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Overexpression of IGF-I occurs in tumors diagnosed in childhood (osteosarcoma, Wilms tumor, neuroblastoma, etc.) and in adults (breast, ovaries, colon and prostate cancer). The aim of our study was to establish the prevalence of malignancies in states of congenital IGF-I deficiency. SUBJECTS We surveyed 222 patients with congenital IGF-I deficiency (Laron syndrome, GH gene deletion, GHRH receptor defects and IGF-I resistance) and 338 first and second-degree relatives. RESULTS None of the IGF-I deficient patients had cancer, whereas 9-24% of the family members had a history of malignancy. CONCLUSIONS Congenital IGF-I deficiency acts as a protecting factor for the development of cancer.
Collapse
Affiliation(s)
- O Shevah
- Endocrinology and Diabetes Research Unit, Schneider Children's Medical Center, 14 Kaplan Street, Petah-Tikva 49202, Israel
| | | |
Collapse
|
45
|
Zhang H, Pelzer AM, Kiang DT, Yee D. Down-regulation of type I insulin-like growth factor receptor increases sensitivity of breast cancer cells to insulin. Cancer Res 2007; 67:391-7. [PMID: 17210722 DOI: 10.1158/0008-5472.can-06-1712] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The type I insulin-like growth factor receptor (IGF1R) and insulin receptor (IR) are structurally and functionally related heterotetrameric receptors. Activation of IGF1R has been shown to regulate breast cancer cell biology, and it has become an attractive therapeutic target. Most strategies have focused on targeting IGF1R alone without affecting IR levels given the known physiologic functions of IR. Human breast cancer cell lines and tissues revealed mRNA expression of both IGF1R and IR. Because alphabeta chains of IGF1R and IR form hybrid receptors, we hypothesized that agents solely targeting IGF1R may affect tumor biology mediated by IGF1R/IR hybrids and IR. We used small interfering RNA (siRNA) technology to specifically down-regulate IGF1R by 60% to 80% in the MDA-435/LCC6 cell line, which was sufficient to diminish activation of IGF1R by IGF-I. IGF1R down-regulation by siRNA did not affect IR levels but, interestingly, sensitized cells to insulin activation of downstream signaling pathways in several breast cancer cell lines. IGF1R siRNA treatment diminished hybrid receptor formation, suggesting that specific down-regulation of IGF1R resulted in enhanced holo-IR formation. In addition, IGF1R down-regulation increased insulin binding consistent with the formation of an increased number of holo-IR on the cell surface. Accordingly, insulin-stimulated glucose uptake was enhanced on IGF1R down-regulation. In conclusion, our data suggest that specific siRNA targeting of IGF1R alone in breast cancer increases insulin sensitivity. Because IR also activates signaling pathways similar to IGF1R in breast cancer cells, agents targeting both receptors may be necessary to disrupt the malignant phenotype regulated by this growth factor system.
Collapse
Affiliation(s)
- Hua Zhang
- University of Minnesota Cancer Center, 420 Delaware Street Southeast, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
46
|
Zhang H, Yee D. Is the type I insulin-like growth factor receptor a therapeutic target in endometrial cancer? Clin Cancer Res 2007; 12:6323-5. [PMID: 17085640 DOI: 10.1158/1078-0432.ccr-06-1707] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Hua Zhang
- University of Minnesota Cancer Center, Minneapolis, Minnesota 55455, USA
| | | |
Collapse
|
47
|
Sarma PKS, Tandon R, Gupta P, Dastidar SG, Ray A, Das B, Cliffe IA. Progress in the development of small molecule inhibitors of insulin-like growth factor-1 receptor kinase. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.1.25] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
Mulvihill MJ, Ji QS, Werner D, Beck P, Cesario C, Cooke A, Cox M, Crew A, Dong H, Feng L, Foreman KW, Mak G, Nigro A, O'Connor M, Saroglou L, Stolz KM, Sujka I, Volk B, Weng Q, Wilkes R. 1,3-Disubstituted-imidazo[1,5-a]pyrazines as insulin-like growth-factor-I receptor (IGF-IR) inhibitors. Bioorg Med Chem Lett 2006; 17:1091-7. [PMID: 17127062 DOI: 10.1016/j.bmcl.2006.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 11/04/2006] [Accepted: 11/07/2006] [Indexed: 01/23/2023]
Abstract
A series of novel 8-amino-1,3-disubstituted-imidazo[1,5-a]pyrazines was designed and synthesized as IGF-IR inhibitors.
Collapse
Affiliation(s)
- Mark J Mulvihill
- (OSI) Oncology, OSI Pharmaceuticals, Inc., 1 Bioscience Park Drive, Farmingdale, NY 11735, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shaw LC, Pan H, Afzal A, Calzi SL, Spoerri PE, Sullivan SM, Grant MB. Proliferating endothelial cell-specific expression of IGF-I receptor ribozyme inhibits retinal neovascularization. Gene Ther 2006; 13:752-60. [PMID: 16437130 DOI: 10.1038/sj.gt.3302718] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Insulin-like growth factor-I (IGF-I) and its receptor (IGF-IR) are essential for normal ocular development and are expressed in numerous ocular cell types including lens epithelial cells, retinal pigment epithelial cells, Müller cells and endothelial cells. Endothelial cell proliferation is a common feature of proliferative retinopathies and involves abnormal growth of blood vessels within and on the surface of the retina. In an effort to inhibit the formation of these aberrant blood vessels, we cloned an IGF-IR ribozyme into an expression vector that limits expression of the ribozyme to proliferating endothelial cells. An endothelin enhancer and Cdc6 promoter chimera drives expression of the IGF-IR ribozyme. This promoter limited retinal expression of the reporter gene to proliferating endothelial cells in two mouse models of proliferative retinopathy. In addition, expression of the IGF-IR ribozyme by this promoter inhibited aberrant retinal angiogenesis in both models while preserving normal vessels. These results demonstrate the feasibility of IGF-IR ribozyme expression in a selective manner for safer treatment of abnormal angiogenesis associated with retinopathy.
Collapse
Affiliation(s)
- L C Shaw
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The insulin-like growth factor (IGF) axis has been implicated in malignant transformation and in tumor cell biology. Human population studies have demonstrated that high levels of circulating IGF-I are associated with an increased risk of certain malignancies. Many model systems show that IGFs stimulate tumor cell proliferation, survival and metastasis. In a new era of anticancer treatments aimed at tumor-specific targets, efforts are in progress for the development of novel anti-IGF therapies. Disrupting type I IGF-receptor (IGF-IR) function in vitro and in vivo results in tumor growth inhibition in several model systems. Antireceptor therapies in particular have provided encouraging results leading to the approval of the first Phase I human clinical trial targeting IGF-IR. Additional methods to decrease levels of circulating IGF-I and II have also been developed. In principle, a benefit of targeted therapies could be their relative lack of toxicity compared with conventional chemotherapy. Anti-IGF-IR therapies, however, raise theoretical concerns for the development of serious side effects, including diabetes. As targeted therapies against the IGF axis continue to be developed, efforts will need to be made to minimize the side effects that result from blocking normal ligand and receptor-induced functions.
Collapse
Affiliation(s)
- Tufia Haddad
- Department of Medicine, Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, MMC 806, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|