1
|
Mehdi SH, Nafees S, Mehdi SJ, Morris CA, Mashouri L, Yoon D. Animal Models of Multiple Myeloma Bone Disease. Front Genet 2021; 12:640954. [PMID: 34163520 PMCID: PMC8215650 DOI: 10.3389/fgene.2021.640954] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a clonal B-cell disorder characterized by the proliferation of malignant plasma cells (PCs) in the bone marrow, the presence of monoclonal serum immunoglobulin, and osteolytic lesions. It is the second most common hematological malignancy and considered an incurable disease despite significant treatment improvements. MM bone disease (MMBD) is defined as the presence of one or more osteolytic bone lesions or diffused osteoporosis with compression fracture attributable to the underlying clonal PC disorder. MMBD causes severe morbidity and increases mortality. Cumulative evidence shows that the interaction of MM cells and bone microenvironment plays a significant role in MM progression, suggesting that these interactions may be good targets for therapy. MM animal models have been developed and studied in various aspects of MM tumorigenesis. In particular, MMBD has been studied in various models, and each model has unique features. As the general features of MM animal models have been reviewed elsewhere, the current review will focus on the features of MMBD animal models.
Collapse
Affiliation(s)
- Syed Hassan Mehdi
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sana Nafees
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Syed Jafar Mehdi
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Carol A Morris
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Ladan Mashouri
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Donghoon Yoon
- Myeloma Center, The University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
2
|
Ochoa MC, Perez-Ruiz E, Minute L, Oñate C, Perez G, Rodriguez I, Zabaleta A, Alignani D, Fernandez-Sendin M, Lopez A, Muntasell A, Sanmamed MF, Paiva B, Lopez-Botet M, Berraondo P, Melero I. Daratumumab in combination with urelumab to potentiate anti-myeloma activity in lymphocyte-deficient mice reconstituted with human NK cells. Oncoimmunology 2019; 8:1599636. [PMID: 31143521 DOI: 10.1080/2162402x.2019.1599636] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/05/2019] [Accepted: 03/12/2019] [Indexed: 10/27/2022] Open
Abstract
Daratumumab is an anti-CD38 fully human IgG1 mAb approved for multiple myeloma treatment. One of the proposed mechanisms of action is the induction of antibody-dependent cellular cytotoxicity (ADCC) mediated by NK cells. NK cells acquire surface CD137 expression in the presence of solid-phase-attached daratumumab and when encountering a daratumumab-coated CD38+ tumor cell line. In this setting, addition of the agonist anti-CD137 mAb urelumab enhances NK-cell activation increasing CD25 expression and IFNɣ production. However, in vitro ADCC is not increased by the addition of urelumab both in 4h or 24h lasting experiments. To study urelumab-increased daratumumab-mediated ADCC activity in vivo, we set up a mouse model based on the intravenous administration of a luciferase-transfected multiple myeloma cell line of human origin, human NK cells and daratumumab to immuno-deficient NSG mice. In this model, intravenous administration of urelumab 24h after daratumumab delayed tumor growth and prolonged mice survival.
Collapse
Affiliation(s)
- Maria C Ochoa
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Luna Minute
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Carmen Oñate
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain
| | - Guiomar Perez
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain
| | - Inmaculada Rodriguez
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain
| | - Aintzane Zabaleta
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Cytometry Unit, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Diego Alignani
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Cytometry Unit, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ascension Lopez
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain.,Cell Therapy Area, University Hospital of Navarra, Pamplona, Spain
| | - Aura Muntasell
- Immunity and Infection Lab, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain
| | - Bruno Paiva
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Cytometry Unit, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Miguel Lopez-Botet
- Immunity and Infection Lab, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut. Immunology Unit, University Pompeu Fabra, Barcelona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Department of Immunology and Immunotherapy, University Hospital of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
3
|
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm which is defined by strong interactions with the bone marrow microenvironment, a compartment with high cellular heterogeneity and unique structural and extracellular components. This necessitates the use of in vivo models for research to fully recapitulate MM growth conditions. The selection of appropriate model system is crucial, as each has advantages and shortcomings. Here, we describe the murine models available for studying MM, and focus on the methods for inoculating mice with MM cells via intravenous, intratibial or subcutaneous delivery, as well as monitoring of disease and organ processing for further analysis. The interaction and destruction of bone is a hallmark symptom of MM, and therefore many other complementary techniques used in calcified tissue research can be used, such as microCT, histomorphometry, and biomechanical testing.
Collapse
|
4
|
Rossi M, Botta C, Arbitrio M, Grembiale RD, Tagliaferri P, Tassone P. Mouse models of multiple myeloma: technologic platforms and perspectives. Oncotarget 2018; 9:20119-20133. [PMID: 29732008 PMCID: PMC5929451 DOI: 10.18632/oncotarget.24614] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/24/2018] [Indexed: 12/19/2022] Open
Abstract
Murine models of human multiple myeloma (MM) are key tools for the study of disease biology as well as for investigation and selection of novel candidate therapeutics for clinical translation. In the last years, a variety of pre-clinical models have been generated to recapitulate a wide spectrum of biological features of MM. These systems range from spontaneous or transgenic models of murine MM, to subcutaneous or orthothopic xenografts of human MM cell lines in immune compromised animals, to platform allowing the engraftment of primary/bone marrow-dependent MM cells within a human bone marrow milieu to fully recapitulate human disease. Selecting the right model for specific pre-clinical research is essential for the successful completion of investigation. We here review recent and most known pre-clinical murine, transgenic and humanized models of MM, focusing on major advantages and/or weaknesses in the light of different research aims.
Collapse
Affiliation(s)
- Marco Rossi
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Cirino Botta
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Mariamena Arbitrio
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | | | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
5
|
Cloos J, Roeten MS, Franke NE, van Meerloo J, Zweegman S, Kaspers GJ, Jansen G. (Immuno)proteasomes as therapeutic target in acute leukemia. Cancer Metastasis Rev 2018; 36:599-615. [PMID: 29071527 PMCID: PMC5721123 DOI: 10.1007/s10555-017-9699-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The clinical efficacy of proteasome inhibitors in the treatment of multiple myeloma has encouraged application of proteasome inhibitor containing therapeutic interventions in (pediatric) acute leukemia. Here, we summarize the positioning of bortezomib, as first-generation proteasome inhibitor, and second-generation proteasome inhibitors in leukemia treatment from a preclinical and clinical perspective. Potential markers for proteasome inhibitor sensitivity and/or resistance emerging from leukemia cell line models and clinical sample studies will be discussed focusing on the role of immunoproteasome and constitutive proteasome (subunit) expression, PSMB5 mutations, and alternative mechanisms of overcoming proteolytic stress.
Collapse
Affiliation(s)
- Jacqueline Cloos
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Margot Sf Roeten
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Niels E Franke
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Johan van Meerloo
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gertjan Jl Kaspers
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Princess Màxima Center, Utrecht, The Netherlands
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Bieghs L, Johnsen HE, Maes K, Menu E, Van Valckenborgh E, Overgaard MT, Nyegaard M, Conover CA, Vanderkerken K, De Bruyne E. The insulin-like growth factor system in multiple myeloma: diagnostic and therapeutic potential. Oncotarget 2018; 7:48732-48752. [PMID: 27129151 PMCID: PMC5217049 DOI: 10.18632/oncotarget.8982] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/16/2016] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a highly heterogeneous plasma cell malignancy. The MM cells reside in the bone marrow (BM), where reciprocal interactions with the BM niche foster MM cell survival, proliferation, and drug resistance. As in most cancers, the insulin-like growth factor (IGF) system has been demonstrated to play a key role in the pathogenesis of MM. The IGF system consists of IGF ligands, IGF receptors, IGF binding proteins (IGFBPs), and IGFBP proteases and contributes not only to the survival, proliferation, and homing of MM cells, but also MM-associated angiogenesis and osteolysis. Furthermore, increased IGF-I receptor (IGF-IR) expression on MM cells correlates with a poor prognosis in MM patients. Despite the prominent role of the IGF system in MM, strategies targeting the IGF-IR using blocking antibodies or small molecule inhibitors have failed to translate into the clinic. However, increasing preclinical evidence indicates that IGF-I is also involved in the development of drug resistance against current standard-of-care agents against MM, including proteasome inhibitors, immunomodulatory agents, and corticoids. IGF-IR targeting has been able to overcome or revert this drug resistance in animal models, enhancing the efficacy of standard-of-care agents. This finding has generated renewed interest in the therapeutic potential of IGF-I targeting in MM. The present review provides an update of the impact of the different IGF system components in MM and discusses the diagnostic and therapeutic potentials.
Collapse
Affiliation(s)
- Liesbeth Bieghs
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Hematology, Aalborg Hospital, Aalborg University, Denmark.,Department of Biomedicin, Aarhus University, Aarhus, Denmark
| | - Hans E Johnsen
- Department of Hematology, Aalborg Hospital, Aalborg University, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Denmark.,Department of Clinical Medicine, Aalborg University, Denmark
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Mette Nyegaard
- Department of Biomedicin, Aarhus University, Aarhus, Denmark
| | - Cheryl A Conover
- Division of Endocrinology, Metabolism and Nutrition, Endocrine Research Unit, Mayo Clinic, Rochester, NY, USA
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
7
|
Perez‐Cornago A, Appleby PN, Tipper S, Key TJ, Allen NE, Nieters A, Vermeulen R, Roulland S, Casabonne D, Kaaks R, Fortner RT, Boeing H, Trichopoulou A, La Vecchia C, Klinaki E, Hansen L, Tjønneland A, Bonnet F, Fagherazzi G, Boutron‐Ruault M, Pala V, Masala G, Sacerdote C, Peeters PH, Bueno‐de‐Mesquita HB, Weiderpass E, Dorronsoro M, Quirós JR, Barricarte A, Gavrila D, Agudo A, Borgquist S, Rosendahl AH, Melin B, Wareham N, Khaw K, Gunter M, Riboli E, Vineis P, Travis RC. Prediagnostic circulating concentrations of plasma insulin-like growth factor-I and risk of lymphoma in the European Prospective Investigation into Cancer and Nutrition. Int J Cancer 2017; 140:1111-1118. [PMID: 27870006 PMCID: PMC5299544 DOI: 10.1002/ijc.30528] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/19/2016] [Accepted: 10/28/2016] [Indexed: 12/20/2022]
Abstract
Insulin-like growth factor (IGF)-I has cancer promoting activities. However, the hypothesis that circulating IGF-I concentration is related to risk of lymphoma overall or its subtypes has not been examined prospectively. IGF-I concentration was measured in pre-diagnostic plasma samples from a nested case-control study of 1,072 cases of lymphoid malignancies and 1,072 individually matched controls from the European Prospective Investigation into Cancer and Nutrition. Odds ratios (ORs) and confidence intervals (CIs) for lymphoma were calculated using conditional logistic regression. IGF-I concentration was not associated with overall lymphoma risk (multivariable-adjusted OR for highest versus lowest third = 0.77 [95% CI = 0.57-1.03], ptrend = 0.06). There was no statistical evidence of heterogeneity in this association with IGF-I by sex, age at blood collection, time between blood collection and diagnosis, age at diagnosis, or body mass index (pheterogeneity for all ≥ 0.05). There were no associations between IGF-I concentration and risk for specific BCL subtypes, T-cell lymphoma or Hodgkin lymphoma, although number of cases were small. In this European population, IGF-I concentration was not associated with risk of overall lymphoma. This study provides the first prospective evidence on circulating IGF-I concentrations and risk of lymphoma. Further prospective data are required to examine associations of IGF-I concentrations with lymphoma subtypes.
Collapse
Affiliation(s)
- Aurora Perez‐Cornago
- Nuffield Department of Population Health, Cancer Epidemiology UnitUniversity of OxfordOxfordUnited Kingdom
| | - Paul N. Appleby
- Nuffield Department of Population Health, Cancer Epidemiology UnitUniversity of OxfordOxfordUnited Kingdom
| | - Sarah Tipper
- Nuffield Department of Population Health, Cancer Epidemiology UnitUniversity of OxfordOxfordUnited Kingdom
| | - Timothy J. Key
- Nuffield Department of Population Health, Cancer Epidemiology UnitUniversity of OxfordOxfordUnited Kingdom
| | - Naomi E. Allen
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of OxfordOxfordUnited Kingdom
| | - Alexandra Nieters
- Center for Chronic Immunodeficiency, Molecular EpidemiologyUniversity Medical Center FreiburgFreiburgGermany
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Division Environmental Epidemiology, Utrecht UniversityUtrechtThe Netherlands
| | - Sandrine Roulland
- Centre d'Immunologie de Marseille‐Luminy, Université d'Aix‐Marseille UM2, Inserm, U1104, CNRSMarseilleFrance
| | - Delphine Casabonne
- Unit of Infections and Cancer (UNIC), IDIBELL, Institut Català d'Oncologia, 08907 L'Hospitalet de LlobregatBarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
| | - Rudolf Kaaks
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Renee T. Fortner
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Heiner Boeing
- Department of EpidemiologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Antonia Trichopoulou
- Hellenic Health FoundationAthensGreece
- Department of Hygiene, Epidemiology and Medical Statistics, WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public HealthUniversity of Athens Medical SchoolGreece
| | - Carlo La Vecchia
- Hellenic Health FoundationAthensGreece
- Department of Clinical Sciences and Community Health Università degli Studi di MilanoItaly
| | | | - Louise Hansen
- Danish Cancer Society Research CenterCopenhagenDenmark
| | | | - Fabrice Bonnet
- Université Paris‐Saclay, Université Paris‐Sud, UVSQ, CESP, INSERMVillejuifFrance
- Gustave RoussyVillejuifFrance
- CHU Rennes, University Rennes 1RennesFrance
| | - Guy Fagherazzi
- Université Paris‐Saclay, Université Paris‐Sud, UVSQ, CESP, INSERMVillejuifFrance
- Gustave RoussyVillejuifFrance
| | | | - Valeria Pala
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei TumoriMilanoItaly
| | - Giovanna Masala
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute—ISPOFlorenceItaly
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, AO Citta' della Salute e della Scienza‐University of Turin and Center for Cancer Prevention (CPO‐Piemonte)TurinItaly
| | - Petra H. Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtThe Netherlands
- Department of Epidemiology and Biostatistics, MRC‐PHE Centre for Environment and Health, School of Public HealthImperial CollegeLondonUnited Kingdom
| | - H. B(as) Bueno‐de‐Mesquita
- Department for Determinants of Chronic Diseases (DCD)National Institute for Public Health and the Environment (RIVM)BilthovenThe Netherlands
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUnited Kingdom
- Department of Social & Preventive Medicine, Faculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health SciencesUniversity of Tromsø, The Arctic University of NorwayTromsøNorway
- Department of Research, Cancer Registry of NorwayInstitute of Population‐Based Cancer ResearchOsloNorway
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Genetic Epidemiology Group, Folkhälsan Research CenterHelsinkiFinland
| | - Miren Dorronsoro
- Basque Regional Health Department San SebastianPublic Health Direction and Biodonostia‐ CiberespSpain
| | | | - Aurelio Barricarte
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- Navarra Public Health InstitutePamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Diana Gavrila
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- Department of EpidemiologyMurcia Regional Health Council, IMIB‐ArrixacaMurciaSpain
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Cancer Epidemiology Research ProgramCatalan Institute of Oncology‐IDIBELL, L'Hospitalet de LlobregatBarcelonaSpain
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Division of Oncology and PathologyLund University, Faculty of MedicineLundSweden
| | - Ann H. Rosendahl
- Department of Clinical Sciences Lund, Division of Oncology and PathologyLund University, Faculty of MedicineLundSweden
| | - Beatrice Melin
- Department of Radiation SciencesOncology Umeå UniversityUmeåSweden
| | - Nick Wareham
- MRC Epidemiology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Kay‐Tee Khaw
- University of Cambridge School of Clinical MedicineCambridgeUnited Kingdom
| | - Marc Gunter
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUnited Kingdom
- Section of Nutrition and Metabolism, International Agency for Research on CancerLyonFrance
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUnited Kingdom
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUnited Kingdom
| | - Ruth C. Travis
- Nuffield Department of Population Health, Cancer Epidemiology UnitUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
8
|
Clinical studies in humans targeting the various components of the IGF system show lack of efficacy in the treatment of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 772:105-122. [PMID: 28528684 DOI: 10.1016/j.mrrev.2016.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 01/28/2023]
Abstract
The insulin-like growth factors (IGFs) system regulates cell growth, differentiation and energy metabolism and plays crucial role in the regulation of key aspects of tumor biology, such as cancer cell growth, survival, transformation and invasion. The current focus for cancer therapeutic approaches have shifted from the conventional treatments towards the targeted therapies and the IGF system has gained a great interest as anti-cancer therapy. The proliferative, anti-apoptotic and transformation effects of IGFs are mainly triggered by the ligation of the type I IGF receptor (IGF-IR). Thus, aiming at developing novel and effective cancer therapies, different strategies have been employed to target IGF system in human malignancies, including but not limited to ligand or receptor neutralizing antibodies and IGF-IR signaling inhibitors. In this review, we have focused on the clinical studies that have been conducted targeting the various components of the IGF system for the treatment of different types of cancer, providing a description and the challenges of each targeting strategy and the degree of success.
Collapse
|
9
|
Ma H, Jiang W, Ding J, Li M, Cheng Y, Sun S, Fu C, Liu Y. Polymer Nanoparticle-Based Chemotherapy for Spinal Malignancies. JOURNAL OF NANOMATERIALS 2016; 2016:1-14. [DOI: 10.1155/2016/4754190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Malignant spinal tumors, categorized into primary and metastatic ones, are one of the most serious diseases due to their high morbidity and mortality rates. Common primary spinal tumors include chordoma, chondrosarcoma, osteosarcoma, Ewing’s sarcoma, and multiple myeloma. Spinal malignancies are not only locally invasive and destructive to adjacent structures, such as bone, neural, and vascular structures, but also disruptive to distant organs (e.g., lung). Current treatments for spinal malignancies, including wide resection, radiotherapy, and chemotherapy, have made significant progress like improving patients’ quality of life. Among them, chemotherapy plays an important role, but its potential for clinical application is limited by severe side effects and drug resistance. To ameliorate the current situation, various polymer nanoparticles have been developed as promising excipients to facilitate the effective treatment of spinal malignancies by utilizing their potent advantages, for example, targeting, stimuli response, and synergetic effect. This review overviews the development of polymer nanoparticles for antineoplastic delivery in the treatment of spinal malignancies and discusses future prospects of polymer nanoparticle-based treatment methods.
Collapse
Affiliation(s)
- Hongyun Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department of Spine Surgery, First Hospital of Jilin University, Changchun 130021, China
| | - Weiqian Jiang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Mingqiang Li
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yilong Cheng
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Shuo Sun
- Department of Spine Surgery, First Hospital of Jilin University, Changchun 130021, China
| | - Changfeng Fu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun 130021, China
| | - Yi Liu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
10
|
Paton-Hough J, Chantry AD, Lawson MA. A review of current murine models of multiple myeloma used to assess the efficacy of therapeutic agents on tumour growth and bone disease. Bone 2015; 77:57-68. [PMID: 25868800 DOI: 10.1016/j.bone.2015.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 03/27/2015] [Accepted: 04/02/2015] [Indexed: 12/21/2022]
Abstract
Pre-clinical in vivo models of multiple myeloma are essential tools for investigating the pathophysiology of multiple myeloma and for testing new therapeutic agents and strategies prior to their potential use in clinical trials. Over the last five decades, several different types of murine models of multiple myeloma have been developed ranging from immunocompetent syngeneic models, e.g. the 5 T series of myeloma cells, to immunocompromised models including the SCID xenograft models, which use human myeloma cell lines or patient-derived cells. Other models include hybrid models featuring the implantation of SCID mice with bone chips (SCID-hu or SCID-rab) or 3-D bone scaffolds (SCID-synth-hu), and mice that have been genetically engineered to develop myeloma. Bearing in mind the differences in these models, it is not surprising that they reflect to varying degrees different aspects of myeloma. Here we review the past and present murine models of myeloma, with particular emphasis on their advantages and limitations, characteristics, and their use in testing therapeutic agents to treat myeloma tumour burden and bone disease.
Collapse
Affiliation(s)
- J Paton-Hough
- Sheffield Myeloma Research Team, Department of Oncology, The University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK.
| | - A D Chantry
- Sheffield Myeloma Research Team, Department of Oncology, The University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK.
| | - M A Lawson
- Sheffield Myeloma Research Team, Department of Oncology, The University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK.
| |
Collapse
|
11
|
Zamykal M, Martens T, Matschke J, Günther HS, Kathagen A, Schulte A, Peters R, Westphal M, Lamszus K. Inhibition of intracerebral glioblastoma growth by targeting the insulin-like growth factor 1 receptor involves different context-dependent mechanisms. Neuro Oncol 2014; 17:1076-85. [PMID: 25543125 DOI: 10.1093/neuonc/nou344] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/24/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Signaling by insulin-like growth factor 1 receptor (IGF-1R) can contribute to the formation and progression of many diverse tumor types, including glioblastoma. We investigated the effect of the IGF-1R blocking antibody IMC-A12 on glioblastoma growth in different in vivo models. METHODS U87 cells were chosen to establish rapidly growing, angiogenesis-dependent tumors in the brains of nude mice, and the GS-12 cell line was used to generate highly invasive tumors. IMC-A12 was administered using convection-enhanced local delivery. Tumor parameters were quantified histologically, and the functional relevance of IGF-1R activation was analyzed in vitro. RESULTS IMC-A12 treatment inhibited the growth of U87 and GS-12 tumors by 75% and 50%, respectively. In GS-12 tumors, the invasive tumor extension and proliferation rate were significantly reduced by IMC-A12 treatment, while apoptosis was increased. In IMC-A12-treated U87 tumors, intratumoral vascularization was markedly decreased, and tumor cell proliferation was moderately reduced. Flow cytometry showed that <2% of U87 cells but >85% of GS-12 cells expressed IGF-1R. Activation of IGF-1R by IGF-1 and IGF-2 in GS-12 cells was blocked by IMC-A12. Both ligands stimulated GS-12 cell proliferation, and IGF-2 also stimulated migration. IMC-A12 inhibited these stimulatory effects and increased apoptosis. In U87 cells, stimulation with either ligand had no functional effect. CONCLUSIONS IGF-1R blockade can inhibit glioblastoma growth by different mechanisms, including direct effects on the tumor cells as well as indirect anti-angiogenic effects. Hence, blocking IGF-1R may be useful to target both the highly proliferative, angiogenesis-dependent glioblastoma core component as well as the infiltrative periphery.
Collapse
Affiliation(s)
- Martin Zamykal
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Tobias Martens
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Jakob Matschke
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Hauke S Günther
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Annegret Kathagen
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Alexander Schulte
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Regina Peters
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Manfred Westphal
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Katrin Lamszus
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| |
Collapse
|
12
|
ZISMANOV VICTORIA, ATTAR-SCHNEIDER OSHRAT, LISHNER MICHAEL, AIZENFELD RACHELHEFFEZ, MATALON SHELLYTARTAKOVER, DRUCKER LIAT. Multiple myeloma proteostasis can be targeted via translation initiation factor eIF4E. Int J Oncol 2014; 46:860-70. [DOI: 10.3892/ijo.2014.2774] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/11/2014] [Indexed: 11/06/2022] Open
|
13
|
Enguita-Germán M, Fortes P. Targeting the insulin-like growth factor pathway in hepatocellular carcinoma. World J Hepatol 2014; 6:716-737. [PMID: 25349643 PMCID: PMC4209417 DOI: 10.4254/wjh.v6.i10.716] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/14/2014] [Accepted: 08/31/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Only 30%-40% of the patients with HCC are eligible for curative treatments, which include surgical resection as the first option, liver transplantation and percutaneous ablation. Unfortunately, there is a high frequency of tumor recurrence after surgical resection and most HCC seem resistant to conventional chemotherapy and radiotherapy. Sorafenib, a multi-tyrosine kinase inhibitor, is the only chemotherapeutic option for patients with advanced hepatocellular carcinoma. Patients treated with Sorafenib have a significant increase in overall survival of about three months. Therefore, there is an urgent need to develop alternative treatments. Due to its role in cell growth and development, the insulin-like growth factor system is commonly deregulated in many cancers. Indeed, the insulin-like growth factor (IGF) axis has recently emerged as a potential target for hepatocellular carcinoma treatment. To this aim, several inhibitors of the pathway have been developed such as monoclonal antibodies, small molecules, antisense oligonucleotides or small interfering RNAs. However recent studies suggest that, unlike most tumors, HCC development requires increased signaling through insulin growth factor II rather than insulin growth factor I. This may have great implications in the future treatment of HCC. This review summarizes the role of the IGF axis in liver carcinogenesis and the current status of the strategies designed to target the IGF-I signaling pathway for hepatocellular carcinoma treatment.
Collapse
|
14
|
Iguchi H, Nishina T, Nogami N, Kozuki T, Yamagiwa Y, Yagawa K. Phase I dose-escalation study evaluating safety, tolerability and pharmacokinetics of MEDI-573, a dual IGF-I/II neutralizing antibody, in Japanese patients with advanced solid tumours. Invest New Drugs 2014; 33:194-200. [PMID: 25342141 DOI: 10.1007/s10637-014-0170-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/02/2014] [Indexed: 11/30/2022]
Abstract
PURPOSE This Phase I, open-label, single-arm, dose-escalation study aimed to evaluate the safety and tolerability of the insulin-like growth factor (IGF-I/II) neutralizing antibody, MEDI-573, in Japanese patients with advanced solid tumours refractory to standard therapy or for which no standard therapy exists. The pharmacokinetics, pharmacodynamics and antitumour activity of MEDI-573 were also evaluated. METHODS Three cohorts of patients received MEDI-573 in escalating order: cohort 1, 5 mg/kg on Day 1, 8 and 15; cohort 2, 15 mg/kg on Day 1, 8 and 15; cohort 3, 45 mg/kg on Day 1, of 21-day cycles. RESULTS Ten patients who received at least one dose of MEDI-573 were evaluated. The median number of treatment cycles was 2.0 (range 1-6) and the median number of MEDI-573 doses received was 4.0 (range 1-17). The most commonly reported drug-related adverse events were fatigue (n = 2 patients), pyrexia (n = 2), diarrhoea (n = 2) and electrocardiogram QT prolongation (n = 2). No patients experienced a dose-limiting toxicity. Pharmacokinetics of MEDI-573 were linear with a dose-dependent increase. There were no complete or partial responses; four patients had an overall best response of stable disease. CONCLUSIONS MEDI-573 is well tolerated at the doses investigated.
Collapse
Affiliation(s)
- Haruo Iguchi
- Clinical Research Center, National Hospital Organization Shikoku Cancer Center, 160 Kou, Minamiumemoto-chou, Matsuyama City, Ehime Prefecture, 791-0280, Japan,
| | | | | | | | | | | |
Collapse
|
15
|
Zi FM, He JS, Li Y, Wu C, Yang L, Yang Y, Wang LJ, He DH, Zhao Y, Wu WJ, Zheng GF, Han XY, Huang H, Yi Q, Cai Z. Metformin displays anti-myeloma activity and synergistic effect with dexamethasone in in vitro and in vivo xenograft models. Cancer Lett 2014; 356:443-53. [PMID: 25305450 DOI: 10.1016/j.canlet.2014.09.050] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 09/17/2014] [Accepted: 09/21/2014] [Indexed: 12/29/2022]
Abstract
Epidemiologic studies and meta-analyses have suggested that patients with type 2 diabetes mellitus (T2DM) have a higher incidence of malignancies, including myeloma. Metformin is a widely prescribed antidiabetic drug. Recently, researchers have shown that metformin has direct anticancer activity against many tumor cell lines, mainly through activating AMP-activated protein kinase (AMPK) or reducing the blood insulin level. In the present study, we investigated whether metformin exerts an anti-myeloma effect in in vitro and in vivo xenograft models and explored the underlying mechanism. We found that metformin can inhibit proliferation of MM cells by inducing apoptosis and cell cycle arrest in the G0/G1 phase. Western blot showed that metformin activated caspase 3, caspase 9, PARP-1, Bak, and p21 and inactivated Mcl-1, HIAP-1, cyclin D1, CDK4, and CDK6. Metformin inhibited the expression of insulin growth factor-I receptor (IGF-IR), and phosphatidyl inositol 3-kinase (PI3K), protein kinase B (PKB/AKT) and the downstream mammalian target of rapamycin (mTOR). IGF-I blocked metformin-induced MM cell apoptosis and reactivation of the PI3K/AKT/mTOR signaling pathway. Metformin also demonstrated synergistic activity with dexamethasone but not bortezomib to eradicate MM cells in vitro and in vivo, especially in MM.1S cells. We conclude that metformin inhibits MM cell proliferation through the IGF-1R/PI3K/AKT/mTOR signaling pathway. Metformin and dexamethasone combination therapy may be an option for MM treatment.
Collapse
Affiliation(s)
- Fu-Ming Zi
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jing-Song He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cai Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Yang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Yang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li-Juan Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dong-Hua He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen-Jun Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gao-Feng Zheng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao-Yan Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qing Yi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
BENABOU NADIA, MIRSHAHI PEZHMAN, BORDU CAMILE, FAUSSAT ANNEMARIE, TANG RUOPING, THERWATH AMU, SORIA JEANETE, MARIE JEANPIERE, MIRSHAHI MASSOUD. A subset of bone marrow stromal cells regulate ATP-binding cassette gene expression via insulin-like growth factor-I in a leukemia cell line. Int J Oncol 2014; 45:1372-1380. [PMID: 25095896 PMCID: PMC4151812 DOI: 10.3892/ijo.2014.2569] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/05/2014] [Indexed: 12/16/2022] Open
Abstract
The importance of the insulin-like growth factor, IGF, as a signaling axis in cancer development, progression and metastasis is highlighted by its effects on cancer cells, notably proliferation and acquired resistance. The role of the microenvironment within which cancer cells evolve and which mediates this effect is far from clear. Here, the involvement of IGF-I in inducing multidrug resistance in a myeloid leukemia cell line, grown in the presence of bone marrow-derived stromal cells called 'Hospicells' (BMH), is demonstrated. We found that i) drug sensitive as well as resistant leukemia cells express IGF-I and its receptor IGF-IR. However, the resistant cells were found to secrete high levels of IGF-I. ii) Presence of exogenous IGF-I promoted cell proliferation, which decreased when an inhibitor of IGF-IR (picropodophyllin, PPP) was added. iii) BMH and IGF-I are both involved in the regulation of genes of the ATP binding cassette (ABC) related to resistance development (MDR1, MRP1, MRP2, MRP3 and BCRP). iv) The levels of ABC gene expression by leukemia cells were found to increase in the presence of increasing numbers of BMH. However, these levels decreased when IGF-IR was inhibited by addition of PPP. v) Co-culture of the drug-sensitive leukemia cells with BMH induced protection against the action of daunorubicin. This chemoresistance was amplified by the presence of IGF-I whereas it decreased when IGF-IR was inhibited. Our results underline the role of microenvironment in concert with the IGF-1 pathway in conferring drug resistance to leukemia cells.
Collapse
Affiliation(s)
- NADIA BENABOU
- UMR, Paris Diderot, Paris 7 University, Lariboisiere Hospital, INSERM U965, Paris, France
| | - PEZHMAN MIRSHAHI
- UMR, Paris Diderot, Paris 7 University, Lariboisiere Hospital, INSERM U965, Paris, France
| | - CAMILE BORDU
- UMR, Paris Diderot, Paris 7 University, Lariboisiere Hospital, INSERM U965, Paris, France
| | | | - RUOPING TANG
- Tumor Bank ‘Leukemia’, Saint-Antoine Hospital, Paris, France
| | - AMU THERWATH
- UMR, Paris Diderot, Paris 7 University, Lariboisiere Hospital, INSERM U965, Paris, France
| | - JEANETE SORIA
- UMR, Paris Diderot, Paris 7 University, Lariboisiere Hospital, INSERM U965, Paris, France
| | | | - MASSOUD MIRSHAHI
- UMR, Paris Diderot, Paris 7 University, Lariboisiere Hospital, INSERM U965, Paris, France
| |
Collapse
|
17
|
Cirstea D, Santo L, Hideshima T, Eda H, Mishima Y, Nemani N, Mahindra A, Yee A, Gorgun G, Hu Y, Ohguchi H, Suzuki R, Cottini F, Guichard SM, Anderson KC, Raje N. Delineating the mTOR Kinase Pathway Using a Dual TORC1/2 Inhibitor, AZD8055, in Multiple Myeloma. Mol Cancer Ther 2014; 13:2489-500. [DOI: 10.1158/1535-7163.mct-14-0147] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Weigel B, Malempati S, Reid JM, Voss SD, Cho SY, Chen HX, Krailo M, Villaluna D, Adamson PC, Blaney SM. Phase 2 trial of cixutumumab in children, adolescents, and young adults with refractory solid tumors: a report from the Children's Oncology Group. Pediatr Blood Cancer 2014; 61:452-6. [PMID: 23956055 PMCID: PMC4511811 DOI: 10.1002/pbc.24605] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 12/12/2022]
Abstract
PURPOSE This phase 2 study was designed to assess the efficacy of single agent cixutumumab (IMC-A12) and gain further information about associated toxicities and pharmacodynamics in children, adolescents, and young adults with recurrent or refractory solid tumors. PATIENTS AND METHODS Patients with relapsed or refractory solid tumors were treated with 9 mg/kg of cixutumumab as a 1-hour IV infusion once weekly. Strata included: osteosarcoma, Ewing sarcoma, rhabdomyosarcoma, neuroblastoma (evaluable disease), neuroblastoma (measurable disease), Wilms tumor, adrenocortical carcinoma, synovial sarcoma, hepatoblastoma, and retinoblastoma. Correlative studies in consenting patients included an assessment of c-peptide, IGFBP-3, IGF-1, IGF-2, hGH, and insulin in consenting patients. RESULTS One hundred sixteen patients with 114 eligible having a median age of 12 years (range, 2-30) were enrolled. Five patients achieved a partial response: 4/20 with neuroblastoma (evaluable only) and 1/20 with rhabdomyosarcoma. Fourteen patients had stable disease for a median of 10 cycles. Hematologic and non-hematologic toxicities were generally mild and infrequent. Serum IGF-1 and IGFBP-3 increased in response to therapy with cixutumumab. CONCLUSION Cixutumumab is well tolerated in children with refractory solid tumors. Limited objective single-agent activity of cixutumumab was observed; however, prolonged stable disease was observed in 15% of patients. Ongoing studies are evaluating the toxicity and benefit of cixutumumab in combination with other agents that inhibit the IGF pathway.
Collapse
Affiliation(s)
| | - Suman Malempati
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | | | - Stephan D. Voss
- Children's Hospital-Boston and Dana Farber Cancer Institute, Boston, MA
| | | | | | - Mark Krailo
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA
| | | | | | - Susan M. Blaney
- Texas Children's Cancer Center/Baylor College of Medicine, Houston, TX
| |
Collapse
|
19
|
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: 144] [Impact Index Per Article: 12.0] [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
|
20
|
Leiphrakpam PD, Agarwal E, Mathiesen M, Haferbier KL, Brattain MG, Chowdhury S. In vivo analysis of insulin-like growth factor type 1 receptor humanized monoclonal antibody MK-0646 and small molecule kinase inhibitor OSI-906 in colorectal cancer. Oncol Rep 2013; 31:87-94. [PMID: 24173770 PMCID: PMC3868504 DOI: 10.3892/or.2013.2819] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/20/2013] [Indexed: 01/15/2023] Open
Abstract
The development and characterization of effective anticancer drugs against colorectal cancer (CRC) is of urgent need since it is the second most common cause of cancer death. The study was designed to evaluate the effects of two IGF-1R antagonists, MK-0646, a recombinant fully humanized monoclonal antibody and OSI-906, a small molecule tyrosine kinase inhibitor on CRC cells. Xenograft study was performed on IGF-1R-dependent CRC cell lines for analyzing the antitumor activity of MK-0646 and OSI-906. Tumor proliferation and apoptosis were assessed using Ki67 and TUNEL assays, respectively. We also performed in vitro characterization of MK-0646 and OSI-906 treatment on CRC cells to identify mechanisms associated with drug-induced cell death. Exposure of the GEO and CBS tumor xenografts to MK-0646 or OSI-906 led to a decrease in tumor growth. TUNEL analysis showed an increase of approximately 45-55% in apoptotic cells in both MK-0646 and OSI-906 treated tumor samples. We report the novel finding that treatment with IGF-1R antagonists led to downregulation of X-linked inhibitor of apoptosis (XIAP) protein involved in cell survival and inhibition of cell death. In conclusion, IGF-1R antagonists (MK-0646 and OSI-906) demonstrated single agent inhibition of subcutaneous CRC xenograft growth. This was coupled to pro-apoptotic effects resulting in downregulation of XIAP and inhibition of cell survival. We report a novel mechanism by which MK-0646 and OSI-906 elicits cell death in vivo and in vitro. Moreover, these results indicate that MK-0646 and OSI-906 may be potential anticancer candidates for the treatment of patients with IGF-1R-dependent CRC.
Collapse
Affiliation(s)
- Premila D Leiphrakpam
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA
| | | | | | | | | | | |
Collapse
|
21
|
Jernberg-Wiklund H, Nilsson K. Targeting the IGF-1R signaling and mechanisms for epigenetic gene silencing in human multiple myeloma. Ups J Med Sci 2012; 117:166-77. [PMID: 22348393 PMCID: PMC3339548 DOI: 10.3109/03009734.2012.659293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Multiple myeloma (MM) is a B cell malignancy characterized by the expansion of clonal plasmablast/plasma cells within the bone-marrow. It is well established that the bone-marrow microenvironment has a pivotal role in providing critical cytokines and cell-cell interactions to support the growth and survival of the MM tumor clone. The pathogenesis of MM is, however, only fragmentarily understood. Detailed genomic analysis reveals a heterogeneous and complex pattern of structural and numerical chromosomal aberrations. In this review we will discuss some of the recent results on the functional role and potential clinical use of the IGF-1R, one of the major mediators of growth and survival for MM. We will also describe some of our results on epigenetic gene silencing in MM, as it may indeed constitute a novel basis for the understanding of tumor initiation and maintenance in MM and thus may change the current view on treatment strategies for MM.
Collapse
Affiliation(s)
- Helena Jernberg-Wiklund
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
22
|
Bevacizumab attenuates major signaling cascades and eIF4E translation initiation factor in multiple myeloma cells. J Transl Med 2012; 92:178-90. [PMID: 22083671 DOI: 10.1038/labinvest.2011.162] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Multiple myeloma (MM), a malignancy of plasma cells, remains fatal despite introduction of novel therapies, partially due to humoral factors, including vascular endothelial growth factor (VEGF), in their microenvironment. The aim of this study was to explore the efficacy of anti-VEGF treatment with bevacizumab directly on MM cells. Particular attention was directed to the affect of VEGF inhibition on protein translation initiation. Experiments were conducted on MM cells (lines, bone marrow (BM) samples) cultured on plastic. Inhibition of VEGF was achieved with the clinically employed anti-VEGF antibody, bevacizumab, as a platform and its consequences on viability, proliferation, and survival was assessed. VEGF downstream signals of established importance to MM cell biology were assayed as well, with particular emphasis on translation initiation factor eIF4E. We showed that blocking VEGF is deleterious to the MM cells and causes cytostasis. This was evidenced in MM cell lines, as well as in primary BM samples (BM MM). A common bevacizumab-induced attenuation of critical signaling effectors was determined: VEGFR1, mTOR, c-Myc, Akt, STAT3, (cell lines) and eIF4E translation initiation factor (lines and BM). ERK1/2 displayed a variegated response to bevacizumab (lines). Utilizing a constitutively Akt-expressing MM model, we showed that the effect of bevacizumab on viability and eIF4E status is Akt-dependent. Of note, the effect of bevacizumab was achieved with high concentrations (2 mg/ml), but was shown to be specific. These findings demonstrate that bevacizumab has a direct influence on major pathways critically activated in MM that is independent from its established effect on angiogenesis. The cytostatic effect of VEGF inhibition on MM cells underscores its potential in combined therapy, and our findings, regarding its influence on translation initiation, suggest that drugs that unbalance cellular proteostasis may be particularly effective.
Collapse
|
23
|
Malempati S, Weigel B, Ingle AM, Ahern CH, Carroll JM, Roberts CT, Reid JM, Schmechel S, Voss SD, Cho SY, Chen HX, Krailo MD, Adamson PC, Blaney SM. Phase I/II trial and pharmacokinetic study of cixutumumab in pediatric patients with refractory solid tumors and Ewing sarcoma: a report from the Children's Oncology Group. J Clin Oncol 2012; 30:256-62. [PMID: 22184397 PMCID: PMC3269952 DOI: 10.1200/jco.2011.37.4355] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 10/12/2011] [Indexed: 12/16/2022] Open
Abstract
PURPOSE A phase I/II study of cixutumumab (IMC-A12) in children with refractory solid tumors was conducted. This study was designed to assess the toxicities, pharmacokinetics, and pharmacodynamics of cixutumumab in children to determine a recommended phase II dose and to assess antitumor activity in Ewing sarcoma (ES). PATIENTS AND METHODS Pediatric patients with relapsed or refractory solid tumors were treated with cixutumumab as a 1-hour intravenous infusion once per week. Two dose levels-6 and 9 mg/kg-were evaluated using a standard three-plus-three cohort design. Patients with refractory ES were treated in an expanded phase II cohort at each dose level. RESULTS Forty-seven eligible patients with a median age of 15 years (range, 4 to 28 years) were enrolled. Twelve patients were treated in the dose-finding phase. Hematologic and nonhematologic toxicities were generally mild and infrequent. Dose-limiting toxicities included grade 4 thrombocytopenia at 6 mg/kg and grade 3 dehydration at 9 mg/kg. Mean trough concentration (± standard deviation) at 9 mg/kg was 106 ± 57 μg/mL, which exceeded the effective trough concentration of 60 μg/mL observed in xenograft models. Three patients with ES had confirmed partial responses: one of 10 at 6 mg/kg and two of 20 at 9 mg/kg. Serum insulin-like growth factor I (IGF-I) levels consistently increased after one dose of cixutumumab. Tumor IGF-I receptor expression by immunohistochemistry did not correlate with response in patients with ES. CONCLUSION Cixutumumab is well tolerated in children with refractory solid tumors. The recommended phase II dose is 9 mg/kg. Limited single-agent activity of cixutumumab was seen in ES.
Collapse
Affiliation(s)
- Suman Malempati
- Department of Pediatrics, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, CDRC-P, Portland, OR 97239-3098, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ozkan EE. Plasma and tissue insulin-like growth factor-I receptor (IGF-IR) as a prognostic marker for prostate cancer and anti-IGF-IR agents as novel therapeutic strategy for refractory cases: a review. Mol Cell Endocrinol 2011; 344:1-24. [PMID: 21782884 DOI: 10.1016/j.mce.2011.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Accepted: 07/01/2011] [Indexed: 12/13/2022]
Abstract
Cancer database analysis indicates that prostate cancer is one of the most seen cancers in men meanwhile composing the leading cause of morbidity and mortality among developed countries. Current available therapies are surgery, radiotherapy and androgene ablation for prostate carcinoma. The response rate is as high nearly 90% however, most of these recur or become refractory and androgene independent (AI). Therefore recent studies intensified on molecular factors playing role on development of prostate carcinoma and novel treatment strategies targetting these factors and their receptors. Insulin-like growth factor-I (IGF-I) and its primary receptor insulin-like growth factor receptor-I (IGF-IR) are among these factors. Biologic functions and role in malign progression are primarily achieved via IGF-IR which is a type 2 tyrosine kinase receptor. IGF-IR plays an important role in mitogenesis, angiogenesis, transformation, apoptosis and cell motility. It also generates intensive proliferative signals leading to carcinogenesis in prostate tissue. So IGF-IR and its associated signalling system have provoked considerable interest over recent years as a novel therapeutic target in cancer. In this paper it is aimed to sum up the lately published literature searching the relation of IGF-IR and prostate cancer in terms of incidence, pathologic features, and prognosis. This is followed by a discussion of the different possible targets within the IGF-1R system, and drugs developed to interact at each target. A systems-based approach is then used to review the in vitro and in vivo data in the published literature of the following compounds targeting IGF-1R components using specific examples: growth hormone releasing hormone antagonists (e.g. JV-1-38), growth hormone receptor antagonists (e.g. pegvisomant), IGF-1R antibodies (e.g. CP-751,871, AVE1642/EM164, IMC-A12, SCH-717454, BIIB022, AMG 479, MK-0646/h7C10), and IGF-1R tyrosine kinase inhibitors (e.g. BMS-536942, BMS-554417, NVP-AEW541, NVP-ADW742, AG1024, potent quinolinyl-derived imidazo (1,5-a)pyrazine PQIP, picropodophyllin PPP, nordihydroguaiaretic acid Insm-18/NDGA). And the other end point is to yield an overview on the recent progress about usage of this receptor as a novel anticancer agent of targeted therapies in treatment of prostate carcinoma.
Collapse
Affiliation(s)
- Emine Elif Ozkan
- OSM Middle East Health Center, Department of Radiation Oncology, Sanliurfa 63000, Turkey.
| |
Collapse
|
25
|
Spiliotaki M, Markomanolaki H, Mela M, Mavroudis D, Georgoulias V, Agelaki S. Targeting the insulin-like growth factor I receptor inhibits proliferation and VEGF production of non-small cell lung cancer cells and enhances paclitaxel-mediated anti-tumor effect. Lung Cancer 2010; 73:158-65. [PMID: 21190751 DOI: 10.1016/j.lungcan.2010.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 11/09/2010] [Accepted: 11/20/2010] [Indexed: 12/16/2022]
Abstract
The effects of AVE1642, a human monoclonal antibody against IGF-IR, were examined in NSCLC cell lines in order to characterize its anti-proliferative and anti-angiogenic activity as a single agent and in combination with chemotherapy. AVE1642 inhibited IGF-IR signaling and suppressed IGF-I-induced, serum-stimulated or autocrine-mediated proliferation of NSCLC cells in vitro. Furthermore, the combination of paclitaxel and AVE1642 resulted in a sequence-dependent increase in the inhibition of cell proliferation, compared to each agent alone, which was associated with a dose-dependent increase in phosphorylated IGF-IR and Akt. Moreover, inhibition of IGF-IR signaling by AVE1642 reduced IGF-I-induced VEGF production by NSCLC cells as well as the migratory capacity of HUVEC cells challenged with conditioned media from lung cancer cells previously exposed to IGF-I. The above results suggest that inhibition of IGF-IR signaling by AVE1642 enhances the efficacy of chemotherapy and modulates VEGF and angiogenesis in NSCLC. These effects may have important clinical implications in the treatment of NSCLC.
Collapse
Affiliation(s)
- Maria Spiliotaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | | | |
Collapse
|
26
|
Riesterer O, Yang Q, Raju U, Torres M, Molkentine D, Patel N, Valdecanas D, Milas L, Ang KK. Combination of anti-IGF-1R antibody A12 and ionizing radiation in upper respiratory tract cancers. Int J Radiat Oncol Biol Phys 2010; 79:1179-87. [PMID: 21129859 DOI: 10.1016/j.ijrobp.2010.10.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 08/27/2010] [Accepted: 10/09/2010] [Indexed: 12/30/2022]
Abstract
PURPOSE The IGF1/IGF-1R signaling pathway has emerged as a potential determinant of radiation resistance in human cancer cell lines. Therefore we investigated the potency of monoclonal anti-IGF-1R antibody, A12, to enhance radiation response in upper respiratory tract cancers. METHODS AND MATERIALS Cell lines were assessed for IGF-1R expression and IGF1-dependent response to A12 or radiation using viability and clonogenic cancer cell survival assays. In vivo response of tumor xenografts to 10 or 20 Gy and A12 (0.25-2 mg × 3) was assessed using growth delay assays. Combined treatment effects were also analyzed by immunohistochemical assays for tumor cell proliferation, apoptosis, necrosis, and vascular endothelial growth factor expression at Days 1 and 6 after start of treatment. RESULTS A12 enhanced the radiosensitivity of HN5 and FaDu head-and-neck carcinomas in vitro (p < 0.05) and amplified the radioresponse of FaDu xenografts in a dose-dependent manner, with enhancement factors ranging from 1.2 to 1.8 (p < 0.01). Immunohistochemical analysis of FaDu xenografts demonstrated that A12 inhibited tumor cell proliferation (p < 0.05) and vascular endothelial growth factor expression. When A12 was combined with radiation, this resulted in apoptosis induction that persisted until 6 days from the start of treatment and in increased necrosis at Day 1 (p < 0.01, respectively). Combined treatment with A12 and radiation resulted in additive or subadditive growth delay in H460 or A549 xenografts, respectively. CONCLUSIONS The results of this study strengthen the evidence for investigating how anti-IGF-1R strategies can be integrated into radiation and radiation-cetuximab regimen in the treatment of cancer of the upper aerodigestive tract cancers.
Collapse
Affiliation(s)
- Oliver Riesterer
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson, Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Tovar V, Alsinet C, Villanueva A, Hoshida Y, Chiang DY, Solé M, Thung S, Moyano S, Toffanin S, Mínguez B, Cabellos L, Peix J, Schwartz M, Mazzaferro V, Bruix J, Llovet JM. IGF activation in a molecular subclass of hepatocellular carcinoma and pre-clinical efficacy of IGF-1R blockage. J Hepatol 2010; 52:550-9. [PMID: 20206398 PMCID: PMC3662876 DOI: 10.1016/j.jhep.2010.01.015] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 10/01/2009] [Accepted: 10/14/2009] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS IGF signaling has a relevant role in a variety of human malignancies. We analyzed the underlying molecular mechanisms of IGF signaling activation in early hepatocellular carcinoma (HCC; BCLC class 0 or A) and assessed novel targeted therapies blocking this pathway. METHODS An integrative molecular dissection of the axis was conducted in a cohort of 104 HCCs analyzing gene and miRNA expression, structural aberrations, and protein activation. The therapeutic potential of a selective IGF-1R inhibitor, the monoclonal antibody A12, was assessed in vitro and in a xenograft model of HCC. RESULTS Activation of the IGF axis was observed in 21% of early HCCs. Several molecular aberrations were identified, such as overexpression of IGF2 -resulting from reactivation of fetal promoters P3 and P4-, IGFBP3 downregulation and allelic losses of IGF2R (25% of cases). A gene signature defining IGF-1R activation was developed. Overall, activation of IGF signaling in HCC was significantly associated with mTOR signaling (p=0.035) and was clearly enriched in the Proliferation subclass of the molecular classification of HCC (p=0.001). We also found an inverse correlation between IGF activation and miR-100/miR-216 levels (FDR<0.05). In vitro studies showed that A12-induced abrogation of IGF-1R activation and downstream signaling significantly decreased cell viability and proliferation. In vivo, A12 delayed tumor growth and prolonged survival, reducing proliferation rates and inducing apoptosis. CONCLUSIONS Integrative genomic analysis showed enrichment of activation of IGF signaling in the Proliferation subclass of HCC. Effective blockage of IGF signaling with A12 provides the rationale for testing this therapy in clinical trials.
Collapse
Affiliation(s)
- Victoria Tovar
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Clara Alsinet
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Augusto Villanueva
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | | | | | - Manel Solé
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Swan Thung
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Susana Moyano
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Sara Toffanin
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA.,Liver Cancer Group. Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, Milan, Italy
| | - Beatriz Mínguez
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Laia Cabellos
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Judit Peix
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Myron Schwartz
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Vincenzo Mazzaferro
- Liver Cancer Group. Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, Milan, Italy
| | - Jordi Bruix
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Josep M. Llovet
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain.,Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA.,Institució Catalana de Recerca i Estudis Avançats (ICREA). Generalitat de Catalunya. Barcelona, Catalonia, Spain
| |
Collapse
|
28
|
Abstract
The IGF pathway plays a major role in cancer cell proliferation, survival and resistance to antineoplastic therapies in many human malignancies. As such, interference with this pathway is the target of many investigational pharmacologic agents. Cixutumumab, a monoclonal antibody to IGF-1R, utilizes this concept. In this review, we summarize preclinical, pharmacologic and early clinical data regarding this agent and discuss the impact this drug might have on the future treatment of human cancers.
Collapse
Affiliation(s)
- Kevin P McKian
- Mayo Clinic College of Medicine, Division of Medical Oncology, 200 First St. SW Rochester, MN 55905, USA
| | | |
Collapse
|
29
|
Bao XH, Naomoto Y, Hao HF, Watanabe N, Sakurama K, Noma K, Motoki T, Tomono Y, Fukazawa T, Shirakawa Y, Yamatsuji T, Matsuoka J, Takaoka M. IGF-IR and its inhibitors in gastrointestinal carcinomas (Review). Oncol Lett 2010; 1:195-201. [PMID: 22966282 DOI: 10.3892/ol_00000036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 09/07/2009] [Indexed: 12/12/2022] Open
Abstract
The type I insulin-like growth factor receptor (IGF-IR) and its associated signaling system play a significant role in tumorigenesis, tumor survival and progression, and cancer therapeutic resistance, and thus has provoked great interest as a promising target for cancer treatment. In this report we present the role of IGF-IR in gastrointestinal carcinomas whose pathology has been identified as tightly correlated with an abnormal expression and activation of IGF-IR. Reported data from experimental studies suggest the feasibility of targeted IGF-IR therapy in gastrointestinal carcinomas. Many types of inhibitors against IGF-IR have been developed. Inhibitors with anti-IGF-IR monoclonal antibodies and tyrosine kinase inhibitors currently undergoing preclinical and clinical evolution are also reviewed.
Collapse
Affiliation(s)
- Xiao Hong Bao
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chapuis N, Tamburini J, Cornillet-Lefebvre P, Gillot L, Bardet V, Willems L, Park S, Green AS, Ifrah N, Dreyfus F, Mayeux P, Lacombe C, Bouscary D. Autocrine IGF-1/IGF-1R signaling is responsible for constitutive PI3K/Akt activation in acute myeloid leukemia: therapeutic value of neutralizing anti-IGF-1R antibody. Haematologica 2009; 95:415-23. [PMID: 20007139 DOI: 10.3324/haematol.2009.010785] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Alterations in the PI3K/Akt pathway are found in a wide range of cancers and the development of PI3K inhibitors represents a promising approach to cancer therapy. Constitutive PI3K activation, reflecting an intrinsic oncogenic deregulation of primary blast cells, is detected in 50% of patients with acute myeloid leukemia. However, the mechanisms leading to this activation are currently unknown. As we previously reported IGF-1 autocriny in acute myeloid leukemia cells, we investigated whether IGF-1 signaling was involved in the constitutive activation of PI3K. DESIGN AND METHODS We analyzed the IGF-1/IGF-1R signaling pathway and PI3K activity in 40 acute myeloid leukemia bone marrow samples. Specific inhibition of IGF-1/IGF-1R signaling was investigated using neutralizing anti-IGF-1R, anti-IGF-1 antibodies or IGF-1 short interfering RNA. The anti-leukemic activity of the neutralizing anti-IGF-1R was tested by analyzing its effects on leukemic progenitor clonogenicity, blast cell proliferation and survival. RESULTS In all samples tested, we found that functional IGF-1R was constantly expressed in leukemic cells. In the acute myeloid leukemia samples with PI3K activation, we found that the IGF-1R was constitutively phosphorylated, although no IGF-1R activating mutation was detected. Specific inhibition of IGF-1R signaling with neutralizing anti-IGF-1R strongly inhibited the constitutive phosphorylation of both IGF-1R and Akt in 70% of the PI3K activated samples. Moreover, both incubation with anti-IGF-1 antibody and IGF-1 short interfering RNA inhibited Akt phosphorylation in leukemic cells. Finally, neutralizing anti-IGF-1R treatment decreased the clonogenicity of leukemic progenitors and the proliferation of PI3K activated acute myeloid leukemia cells. CONCLUSIONS Our current data indicate a critical role for IGF-1 autocriny in constitutive PI3K/Akt activation in primary acute myeloid leukemia cells and provide a strong rationale for targeting IGF-1R as a potential new therapy for this disease.
Collapse
Affiliation(s)
- Nicolas Chapuis
- Département d'Hématologie, Institut Cochin, CNRS, UMR8104, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The insulin-like growth factor-I receptor (IGF-IR) mediates the biological actions of both IGF-I and IGF-II. The IGF-IR is expressed in most transformed cells, where it displays potent antiapoptotic, cell-survival, and transforming activities. IGF-IR expression is a fundamental prerequisite for the acquisition of a malignant phenotype, as suggested by the finding that IGF-IR-null cells (derived from IGF-IR knock-out embryos) are unable to undergo transformation when exposed to cellular or viral oncogenes. This review article will focus on the underlying molecular mechanisms that are responsible for the normal, physiological control of IGF-IR gene expression, as well as the cellular pathways that underlie its aberrant expression in cancer. Examples from the clinics will be presented, including a description of how the IGF system is involved in breast, prostate, pediatric, and gynecological cancers. Finally, current attempts to target the IGF-IR as a therapeutic approach will be described.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | |
Collapse
|
32
|
A humanised anti-IGF-1R monoclonal antibody (AVE1642) enhances Bortezomib-induced apoptosis in myeloma cells lacking CD45. Br J Cancer 2009; 100:366-9. [PMID: 19165200 PMCID: PMC2634719 DOI: 10.1038/sj.bjc.6604839] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The humanised form of an antagonistic anti-IGF-1R mAb (AVE1642) selectively inhibits the growth of CD45neg myeloma cells. AVE1642 strongly increased bortezomib-induced apoptosis, correlated with an increase of Noxa expression. These results support the therapeutic use of anti-IGF-1R/bortezomib in CD45neg Myeloma patients, particularly those with the most aggressive form, t(4,14).
Collapse
|
33
|
Lindsay CR, Evans TRJ. The insulin-like growth factor system and its receptors: A potential novel anticancer target. Biologics 2008; 2:855-64. [PMID: 19707463 PMCID: PMC2727903 DOI: 10.2147/btt.s3841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The current generation of novel anticancer therapies that are in preclinical and clinical development are based on exploiting our increasing understanding of the molecular and cellular basis of cancer development and progression. Accelerated rates of cell division and proliferation have been postulated to predispose to the development of malignant disease. The insulin-like growth factor (IGF) signaling system has an important physiological role in regulating cellular proliferation and apoptosis. This function has led to considerable interest in its relevance to neoplasia over the last decade. In this review, we give an overview of the IGF system physiology, discuss the epidemiological significance of IGF signaling and neoplasia, and review the preclinical and clinical studies in targeting IGF receptors as cancer therapies.
Collapse
Affiliation(s)
- Colin R Lindsay
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - TR Jeffry Evans
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
- University of Glasgow, CR-UK Beatson Laboratories, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
34
|
Rodon J, DeSantos V, Ferry RJ, Kurzrock R. Early drug development of inhibitors of the insulin-like growth factor-I receptor pathway: lessons from the first clinical trials. Mol Cancer Ther 2008; 7:2575-88. [PMID: 18790742 DOI: 10.1158/1535-7163.mct-08-0265] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The insulin-like growth factor-I receptor (IGF-IR) was first cloned in 1986. Since then, intense work has defined classic phosphorelays activated via the IGF-IR, which regulate cell proliferation, apoptosis, motility, and fate. The understanding of the roles of hormones in cancer and the growth hormone-IGF-IGF-binding protein axis specifically has yield to a second wave of development: the design of specific inhibitors that interrupt the signaling associated with this axis. The ability to manipulate these pathways holds not only significant therapeutic implications but also increase the chance of deeper insight about the role of the axis in carcinogenesis and metastasis. Nowadays, >25 molecules with the same goal are at different stages of development. Here, we review the clinical and preclinical experience with the two most-investigated strategies, tyrosine kinase inhibitors and monoclonal antibodies, and the advantages and disadvantages of each strategy, as well as other alternatives and possible drug combinations. We also review the biomarkers explored in the first clinical trials, the strategies that have been explored thus far, and the clinical trials that are going to explore their role in cancer treatment.
Collapse
Affiliation(s)
- Jordi Rodon
- Investigational Cancer Therapeutics, University of Texas M. D. Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
35
|
Shang Y, Mao Y, Batson J, Scales SJ, Phillips G, Lackner MR, Totpal K, Williams S, Yang J, Tang Z, Modrusan Z, Tan C, Liang WC, Tsai SP, Vanderbilt A, Kozuka K, Hoeflich K, Tien J, Ross S, Li C, Lee SH, Song A, Wu Y, Stephan JP, Ashkenazi A, Zha J. Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake. Mol Cancer Ther 2008; 7:2599-608. [DOI: 10.1158/1535-7163.mct-07-2401] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Lacy MQ, Alsina M, Fonseca R, Paccagnella ML, Melvin CL, Yin D, Sharma A, Enriquez Sarano M, Pollak M, Jagannath S, Richardson P, Gualberto A. Phase I, Pharmacokinetic and Pharmacodynamic Study of the Anti–Insulinlike Growth Factor Type 1 Receptor Monoclonal Antibody CP-751,871 in Patients With Multiple Myeloma. J Clin Oncol 2008; 26:3196-203. [DOI: 10.1200/jco.2007.15.9319] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose A phase I first-in-human study was conducted to characterize the safety, tolerability, pharmacokinetic, and pharmacodynamic properties of the anti–insulinlike growth factor 1 receptor (IGF-IR) monoclonal antibody CP-751,871. Patients and Methods After informed consent and screening, 47 patients with multiple myeloma in relapse or refractory phase were enrolled into 11 dose-escalation cohorts of CP-751,871 at doses from 0.025 to 20 mg/kg for 4 weeks. Patients with less than a partial response to CP-751,871 treatment were eligible to receive CP-751,871 in combination with oral dexamethasone at the discretion of the investigator. Treatment with CP-751,871 and rapamycin with or without dexamethasone was also offered to patients enrolled in the 10 and 20 mg/kg cohorts with less than a partial response to initial therapy with single-agent CP-751,871. Results No CP-751,871-related dose-limiting toxicities were identified. Plasma CP-751,871 concentrations increased with dose and concentration-time profiles were consistent with those of antibodies with target-mediated disposition. Importantly, CP-751,871 administration led to a decrease in granulocyte IGF-IR expression and serum insulinlike growth factor 1 accumulation at high doses, suggesting systemic IGF-IR inhibition. Tumor response was assessed according to the European Group for Blood and Marrow Transplantation criteria. Nine responses were reported in 27 patients treated with CP-751,871 in combination with dexamethasone. Of interest, two of the patients with a partial response were progressing from dexamethasone treatment at study entry. Conclusion These data indicate that CP-751,871 is well tolerated and may constitute a novel agent in the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Martha Q. Lacy
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Melissa Alsina
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Rafael Fonseca
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - M. Luisa Paccagnella
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Carrie L. Melvin
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Donghua Yin
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Amarnath Sharma
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - M. Enriquez Sarano
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Michael Pollak
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Sundar Jagannath
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Paul Richardson
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Antonio Gualberto
- From the Mayo Clinic, Divisions of Hematology and Cardiovascular Diseases, Rochester, MN; H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL; Mayo Clinic, Division of Hematology, Scottsdale, AZ; Pfizer Global Research & Development, New London, CT; St Vincent's Comprehensive Cancer Center, New York, NY; Dana Farber Cancer Institute Boston, MA; and the McGill University and Lady Davis Research Institute, Montreal, Quebec, Canada
| |
Collapse
|
37
|
Yuen JSP, Macaulay VM. Targeting the type 1 insulin-like growth factor receptor as a treatment for cancer. Expert Opin Ther Targets 2008; 12:589-603. [PMID: 18410242 DOI: 10.1517/14728222.12.5.589] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The type 1 insulin-like growth factor receptor (IGF1R) plays a critical role in transformation, invasion and apoptosis protection, and is an attractive cancer treatment target. OBJECTIVE To review IGF1R antibodies and kinase inhibitors that are in preclinical and clinical development, and to discuss questions that will influence the success of this approach in clinical practice. METHODS This review is drawn from published literature, meeting abstracts and online resources. RESULTS/CONCLUSION IGF1R blockade is generally well tolerated although it can induce hyperglycaemia. Single-agent activity has been documented in Ewing's sarcoma but not thus far in common solid tumours. Key issues include identification of factors that influence sensitivity to IGF1R blockade, and how most effectively to combine IGF1R inhibitors with other treatments.
Collapse
Affiliation(s)
- John S P Yuen
- Weatherall Institute of Molecular Medicine, University of Oxford, IGF Group, Molecular Oncology Laboratories, Headley Way, Headington, Oxford OX3 9DS, UK
| | | |
Collapse
|
38
|
HoxA9 induces insulin-like growth factor-1 receptor expression in B-lineage acute lymphoblastic leukemia. Leukemia 2008; 22:1161-9. [DOI: 10.1038/leu.2008.57] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Abstract
Multiple myeloma (MM) remains incurable despite high-dose chemotherapy with stem cell support. There is need, therefore, for continuous efforts directed toward the development of novel rational-based therapeutics for MM, which requires a detailed knowledge of the mutations driving this malignancy. In improving the success rate of effective drug development, it is equally imperative that biologic systems be developed to better validate these target genes. Here we review the recent developments in the generation of mouse models of MM and their impact as preclinical models for designing and assessing target-based therapeutic approaches.
Collapse
|
40
|
Rowinsky EK, Youssoufian H, Tonra JR, Solomon P, Burtrum D, Ludwig DL. IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor. Clin Cancer Res 2007; 13:5549s-5555s. [PMID: 17875788 DOI: 10.1158/1078-0432.ccr-07-1109] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Targeted monoclonal antibody therapy is an important strategy in cancer therapeutics. Among the most promising characteristics of therapeutic targets are those that modulate the growth and survival of malignant neoplasms and their sensitivity to anticancer therapies. The insulin-like growth factor-I receptor (IGF-IR) is overexpressed in many types of solid and hematopoietic malignancies, and has been implicated as a principal cause of heightened proliferative and survival signaling. IGF-IR has also been shown to confer resistance to cytotoxic, hormonal, and targeted therapies, suggesting that therapeutics targeting IGF-IR may be effective against a broad range of malignancies. IMC-A12 (ImClone Systems Incorporated), a fully human monoclonal IgG1 antibody that binds with high affinity to the IGF-IR, inhibits ligand-dependent receptor activation and downstream signaling. IMC-A12 also mediates robust internalization and degradation of the IGF-IR. In human tumor xenograft models, IGF-IR blockade by IMC-A12 results in rapid and profound growth inhibition of cancers of the breast, lung, colon, and pancreas, and many other neoplasms. Although promising single-agent activity has been observed, the most impressive effects of targeting the IGF-IR with IMC-A12 have been noted when this agent was combined with cytotoxic agents or other targeted therapeutics. The results with IMC-A12 to date suggest that it may be an effective therapeutic in a diverse array of oncologic indications.
Collapse
Affiliation(s)
- Eric K Rowinsky
- Department of Clinical Research and Regulatory Affairs, ImClone Systems Incorporated, Branchburg, New Jersey 08876, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Menu E, Jernberg-Wiklund H, De Raeve H, De Leenheer E, Coulton L, Gallagher O, Van Valckenborgh E, Larsson O, Axelson M, Nilsson K, Van Camp B, Croucher P, Vanderkerken K. Targeting the IGF-1R using picropodophyllin in the therapeutical 5T2MM mouse model of multiple myeloma: beneficial effects on tumor growth, angiogenesis, bone disease and survival. Int J Cancer 2007; 121:1857-61. [PMID: 17546599 DOI: 10.1002/ijc.22845] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
During the last decade, a central role for insulin-like growth factor 1 (IGF-1) in the pathophysiology of multiple myeloma (MM) has been well established. IGF-I provided by the tumor-microenvironment interaction may directly and indirectly facilitate the migration, survival and expansion of the MM cells in the bone marrow (BM). The inhibition of the IGF-1R-mediated signaling pathway has recently been suggested to be a possible new therapeutic principle in MM. Using the mouse 5T2MM model, we now demonstrate that targeting the IGF-1R using picropodophyllin (PPP) in a therapeutical setting not only has strong antitumor activity on the established MM tumor but also influences the BM microenvironment by inhibiting angiogenesis and bone disease, having a profound effect on the survival of the mice. At therapeutically achievable concentrations of PPP, the average survival was 180 days for the PPP-treated mice as compared to 100 days for vehicle-treated mice. PPP used as single drug treatment in the 5T2MM model resulted in a decrease of tumor burden by 65% while the paraprotein concentrations were reduced by 75%. This decrease was associated with a significant inhibition of tumor-associated angiogenesis and osteolysis. The present studies on the biological effects of PPP in the 5T2MM model constitute an important experimental platform for future therapeutic implementation.
Collapse
Affiliation(s)
- Eline Menu
- Department of Hematology and Immunology, Vrije Universiteit Brussel-VUB, Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hartog H, Wesseling J, Boezen HM, van der Graaf WTA. The insulin-like growth factor 1 receptor in cancer: old focus, new future. Eur J Cancer 2007; 43:1895-904. [PMID: 17624760 DOI: 10.1016/j.ejca.2007.05.021] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 05/25/2007] [Indexed: 12/16/2022]
Abstract
The importance of insulin-like growth factor 1 receptor (IGF-1R) signalling in malignant behaviour of tumour cells is well established. Currently, development of drugs targeting the IGF-1R as anticancer treatment is emerging. Several IGF-1R targeting strategies are being investigated in phases I and II clinical trials. Interactions of IGF-1R with insulin receptor, however, might complicate efficiency and tolerability of such drugs. This review describes mechanisms, recent developments and potential limitations of IGF-1R antibodies and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Hermien Hartog
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
43
|
Abstract
Therapeutic advances in the treatment of multiple myeloma have significantly improved remission duration and overall survival (OS). These strategies have included the use of immunotherapy (interferon), novel agents (bortezomib, thalidomide, and lenalidomide), corticosteroids, and chemotherapy. While novel agents have had a major impact on response rates with initial therapy, most patients with multiple myeloma will eventually relapse. In the setting of minimal residual disease following standard dose or high-dose therapy, a number of different 'maintenance' strategies have emerged to prolong the duration of initial or subsequent remissions. The impact of these strategies on OS and event-free survival (EFS) is critically important, as the use of ineffective maintenance therapy adds the burden of additional cost, morbidity, and may reduce quality of life. Truly successful maintenance therapy will be effective in the setting of minimal residual disease, and will improve not only EFS, but also OS. This review summarizes the currently available data in the maintenance setting for multiple myeloma, and will discuss potential future trials to further address this important issue.
Collapse
Affiliation(s)
- R Mihelic
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|