1
|
Jia K, Cao L, Yu Y, Jing D, Wu W, Van Tine BA, Shao Z. Signaling pathways and targeted therapies in Ewing sarcoma. Pharmacol Ther 2025; 266:108765. [PMID: 39622389 DOI: 10.1016/j.pharmthera.2024.108765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024]
Abstract
Ewing sarcoma, the second most prevalent malignant bone tumor with potential occurrence in soft tissues, exhibits a high level of aggressiveness, primarily afflicting children and adolescents. It is characterized by fusion proteins arising from chromosomal translocations. The fusion proteins induce aberrations in multiple signaling pathways and molecules, constituting a key event in oncogenic transformation. While diagnostic and therapeutic modalities have advanced in recent decades and multimodal treatments, including surgery, radiotherapy, and chemotherapy, have significantly improved survival of patients with localized tumors, patients with metastatic tumors continue to face poor prognoses. There persists a pressing need for novel alternative treatments, yet the translation of our understanding of Ewing sarcoma pathogenesis into improved clinical outcomes remains a critical challenge. Here, we provide a comprehensive review of Ewing sarcoma, including fusion proteins, various signaling pathways, pivotal pathogenetic molecules implicated in its development, and associated targeted therapies and immunotherapies. We summarize past endeavors, current advancements, and deliberate on limitations and future research directions. It is envisaged that this review will furnish novel insights into prospective treatment avenues for Ewing sarcoma.
Collapse
Affiliation(s)
- Ke Jia
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Li Cao
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Washington University School of Medicine, St Louis, MO, USA.
| | - Yihan Yu
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Doudou Jing
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China.
| | - Wei Wu
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | | | - Zengwu Shao
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
2
|
Kumar S, Chaudhri S. Recent update on IGF-1/IGF-1R signaling axis as a promising therapeutic target for triple-negative breast cancer. Pathol Res Pract 2024; 263:155620. [PMID: 39357179 DOI: 10.1016/j.prp.2024.155620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/10/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Insulin-like growth factor 1/Insulin-like growth factor 1-receptor (IGF-1/IGF-1R) pathway is highly breast cancer subtype context-dependent. Triple-negative breast cancer (TNBC) is an aggressive, highly metastatic cancer showing early recurrence and poor prognosis. High expression of IGF-1 and its receptor IGF-1R, their interaction, autophosphorylation, and activation of intracellular signaling cascades have been significantly associated with TNBC pathophysiology. In the last five to seven years, marvelous work has been done to explore the role of IGF-1/IGF-1R axis in TNBC. In the present review, starting from the general introduction to IGF-1/IGF-1R pathway an up-to-date discussion was focused on its role in TNBC pathophysiology. Further we discussed the up/down stream molecular events of IGF-1/IGF-1R axis, clinical relevance of IGF-1 and IGF-1R levels in TNBC patients, anti-TNBC therapy and possible way-out for IGF-1/IGF-1R axis mediate therapy resistance in TNBC. Combination therapy strategy has been researched to overcome direct IGF-1/IGF-1R pathway inhibition mediated therapy resistance and produced promising results in the management of TNBC. The understanding of up/downstream of the IGF-1/IGF-1R axis provide immense focus on the pathway as a therapeutic target. It is expected within the next decade to determine its potentiality, or lack thereof, for TNBC treatment.
Collapse
Affiliation(s)
- Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Guddha, Bathinda, Punjab 151401, India.
| | - Smriti Chaudhri
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Guddha, Bathinda, Punjab 151401, India
| |
Collapse
|
3
|
Kotsifaki A, Maroulaki S, Karalexis E, Stathaki M, Armakolas A. Decoding the Role of Insulin-like Growth Factor 1 and Its Isoforms in Breast Cancer. Int J Mol Sci 2024; 25:9302. [PMID: 39273251 PMCID: PMC11394947 DOI: 10.3390/ijms25179302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/25/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Insulin-like Growth Factor-1 (IGF-1) is a crucial mitogenic factor with important functions in the mammary gland, mainly through its interaction with the IGF-1 receptor (IGF-1R). This interaction activates a complex signaling network that promotes cell proliferation, epithelial to mesenchymal transition (EMT) and inhibits apoptosis. Despite extensive research, the precise molecular pathways and intracellular mechanisms activated by IGF-1, in cancer, remain poorly understood. Recent evidence highlights the essential roles of IGF-1 and its isoforms in breast cancer (BC) development, progression, and metastasis. The peptides that define the IGF-1 isoforms-IGF-1Ea, IGF-1Eb, and IGF-1Ec-act as key points of convergence for various signaling pathways that influence the growth, metastasis and survival of BC cells. The aim of this review is to provide a detailed exami-nation of the role of the mature IGF-1 and its isoforms in BC biology and their potential use as possible therapeutical targets.
Collapse
Affiliation(s)
- Amalia Kotsifaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sousanna Maroulaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efthymios Karalexis
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Martha Stathaki
- Surgical Clinic, "Elena Venizelou" General Hospital, 11521 Athens, Greece
| | - Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Shahid A, Santos SG, Lin C, Huang Y. Role of Insulin-like Growth Factor-1 Receptor in Tobacco Smoking-Associated Lung Cancer Development. Biomedicines 2024; 12:563. [PMID: 38540176 PMCID: PMC10967781 DOI: 10.3390/biomedicines12030563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer remains a significant global health concern, with lung cancer consistently leading as one of the most common malignancies. Genetic aberrations involving receptor tyrosine kinases (RTKs) are known to be associated with cancer initiation and development, but RTK involvement in smoking-associated lung cancer cases is not well understood. The Insulin-like Growth Factor 1 Receptor (IGF-1R) is a receptor that plays a critical role in lung cancer development. Its signaling pathway affects the growth and survival of cancer cells, and high expression is linked to poor prognosis and resistance to treatment. Several reports have shown that by activating IGF-1R, tobacco smoke-related carcinogens promote lung cancer and chemotherapy resistance. However, the relationship between IGF-1R and cancer is complex and can vary depending on the type of cancer. Ongoing investigations are focused on developing therapeutic strategies to target IGF-1R and overcome chemotherapy resistance. Overall, this review explores the intricate connections between tobacco smoke-specific carcinogens and the IGF-1R pathway in lung carcinogenesis. This review further highlights the challenges in using IGF-1R inhibitors as targeted therapy for lung cancer due to structural similarities with insulin receptors. Overcoming these obstacles may require a comprehensive approach combining IGF-1R inhibition with other selective agents for successful cancer treatment.
Collapse
Affiliation(s)
- Ayaz Shahid
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Shaira Gail Santos
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Carol Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Ying Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
5
|
Andò S, Simões BM. Editorial: Adipokines and hormone-dependent cancers. Front Endocrinol (Lausanne) 2023; 14:1340171. [PMID: 38107522 PMCID: PMC10722399 DOI: 10.3389/fendo.2023.1340171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Affiliation(s)
- Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- Centro Sanitario, University of Calabria, Rende, Italy
| | - Bruno M. Simões
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
6
|
Macaulay VM, Lord S, Hussain S, Maroto JP, Jones RH, Climent MÁ, Cook N, Lin CC, Wang SS, Bianchini D, Bailey M, Schlieker L, Bogenrieder T, de Bono J. A Phase Ib/II study of IGF-neutralising antibody xentuzumab with enzalutamide in metastatic castration-resistant prostate cancer. Br J Cancer 2023; 129:965-973. [PMID: 37537253 PMCID: PMC10491782 DOI: 10.1038/s41416-023-02380-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/10/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND This multicentre, open-label, Phase Ib/II trial evaluated the insulin-like growth factor (IGF) 1/2 neutralising antibody xentuzumab plus enzalutamide in metastatic castrate-resistant prostate cancer (mCRPC). METHODS The trial included Phase Ib escalation and expansion parts and a randomised Phase II part versus enzalutamide alone. Primary endpoints in the Phase Ib escalation, Phase Ib expansion and Phase II parts were maximum tolerated dose (MTD), prostate-specific antigen response and investigator-assessed progression-free survival (PFS), respectively. Patients in the Phase Ib escalation and Phase II parts had progressed on/after docetaxel/abiraterone. RESULTS In the Phase Ib escalation (n = 10), no dose-limiting toxicities were reported, and xentuzumab 1000 mg weekly plus enzalutamide 160 mg daily (Xe1000 + En160) was defined as the MTD and recommended Phase 2 dose. In the Phase Ib expansion (n = 24), median PFS was 8.2 months, and one patient had a confirmed, long-term response. In Phase II (n = 86), median PFS for the Xe1000 + En160 and En160 arms was 7.4 and 6.2 months, respectively. Subgroup analysis suggested trends towards benefit with Xe1000 + En160 in patients whose tumours had high levels of IGF1 mRNA or PTEN protein. Overall, the combination was well tolerated. CONCLUSIONS Xentuzumab plus enzalutamide was tolerable but lacked antitumour activity in unselected patients with mCRPC. CLINICAL TRIAL REGISTRATION EudraCT number 2013-004011-41.
Collapse
Affiliation(s)
| | - Simon Lord
- Department of Oncology, University of Oxford, Oxford, UK
| | - Syed Hussain
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | | | | | | | - Natalie Cook
- The Christie NHS Foundation Trust and the University of Manchester, Manchester, UK
| | - Chia-Chi Lin
- National Taiwan University Hospital, Taipei, Taiwan
| | | | - Diletta Bianchini
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, London, UK
| | - Mark Bailey
- Boehringer Ingelheim Ltd, Bracknell, Berkshire, UK
| | - Laura Schlieker
- External Statistician on behalf of Boehringer Ingelheim GmbH & Co. KG, Staburo GmbH & Co. KG, Munich, Germany
| | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany
| | - Johann de Bono
- The Institute of Cancer Research, London, UK.
- The Royal Marsden NHS Foundation Trust, Sutton, London, UK.
| |
Collapse
|
7
|
Schmid P, Cortes J, Joaquim A, Jañez NM, Morales S, Díaz-Redondo T, Blau S, Neven P, Lemieux J, García-Sáenz JÁ, Hart L, Biyukov T, Baktash N, Massey D, Burris HA, Rugo HS. XENERA-1: a randomised double-blind Phase II trial of xentuzumab in combination with everolimus and exemestane versus everolimus and exemestane in patients with hormone receptor-positive/HER2-negative metastatic breast cancer and non-visceral disease. Breast Cancer Res 2023; 25:67. [PMID: 37308971 DOI: 10.1186/s13058-023-01649-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/20/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Xentuzumab is a humanised monoclonal antibody that binds to IGF-1 and IGF-2, neutralising their proliferative activity and restoring inhibition of AKT by everolimus. This study evaluated the addition of xentuzumab to everolimus and exemestane in patients with advanced breast cancer with non-visceral disease. METHODS This double-blind, randomised, Phase II study was undertaken in female patients with hormone-receptor (HR)-positive/human epidermal growth factor 2 (HER2)-negative advanced breast cancer with non-visceral disease who had received prior endocrine therapy with or without CDK4/6 inhibitors. Patients received a weekly intravenous infusion of xentuzumab (1000 mg) or placebo in combination with everolimus (10 mg/day orally) and exemestane (25 mg/day orally). The primary endpoint was progression-free survival (PFS) per independent review. RESULTS A total of 103 patients were randomised and 101 were treated (n = 50 in the xentuzumab arm and n = 51 in the placebo arm). The trial was unblinded early due to high rates of discordance between independent and investigator assessment of PFS. Per independent assessment, median PFS was 12.7 (95% CI 6.8-29.3) months with xentuzumab and 11.0 (7.7-19.5) months with placebo (hazard ratio 1.19; 95% CI 0.55-2.59; p = 0.6534). Per investigator assessment, median PFS was 7.4 (6.8-9.7) months with xentuzumab and 9.2 (5.6-14.4) months with placebo (hazard ratio 1.23; 95% CI 0.69-2.20; p = 0.4800). Tolerability was similar between the arms, with diarrhoea (33.3-56.0%), fatigue (33.3-44.0%) and headache (21.6-40.0%) being the most common treatment-emergent adverse events. The incidence of grade ≥ 3 hyperglycaemia was similar between the xentuzumab (2.0%) and placebo (5.9%) arms. CONCLUSIONS While this study demonstrated that xentuzumab could be safely combined with everolimus and exemestane in patients with HR-positive/HER2-negative advanced breast cancer with non-visceral disease, there was no PFS benefit with the addition of xentuzumab. Trial registration ClinicalTrials.gov, NCT03659136. Prospectively registered, September 6, 2018.
Collapse
Affiliation(s)
- Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - Ana Joaquim
- Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | | | | | - Tamara Díaz-Redondo
- Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Unidad de Gestión Clínica Intercentros de Oncología, Málaga, Spain
| | - Sibel Blau
- Northwest Medical Specialties, Tacoma, WA, USA
| | | | - Julie Lemieux
- Centre Hospitalier Universitaire de Québec-Université Laval Research Centre, Quebec, Canada
| | | | - Lowell Hart
- Florida Cancer Specialists, Fort Myers, FL, USA
| | | | - Navid Baktash
- Boehringer Ingelheim (Canada) Ltd, Burlington, ON, Canada
| | - Dan Massey
- Elderbrook Solutions GmbH on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Hope S Rugo
- University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Lin W, Lin Z, Lin X, Peng Z, Liang X, Wei S. Integrated analysis and clinical correlation analysis of hub genes, immune infiltration, and potential therapeutic agents related to lupus nephritis. Lupus 2023; 32:633-643. [PMID: 36912500 DOI: 10.1177/09612033231161587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
BACKGROUND Lupus nephritis (LN) is the most common complication of systemic lupus erythematosus (SLE). This study aimed to explore biomarkers, mechanisms, and potential novel agents regarding LN through bioinformatic analysis. METHOD Four expression profiles were downloaded from the Gene Expression Omnibus (GEO) database and differentially expressed genes (DEGs) were acquired. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGGs) pathway enrichment analyses of DEGs were performed using the R software. The protein-protein interaction (PPI) network was developed using the STRING database. Additionally, five algorithms were used to screen out the hub genes. Expression of the hub genes were validated using Nephroseq v5. CIBERSORT was used to evaluate the infiltration of immune cells. Finally, The Drug-Gene Interaction Database was used to predict potential targeted drugs. RESULT FOS and IGF1 were identified as hub genes, with excellent specificity and sensitivity diagnosis of LN. FOS was also related to renal injury. LN patients had lower activated and resting dendritic cells (DCs) and higher M1 macrophages and activated NK cells than healthy control (HC). FOS had a positive correlation with activated mast cells and a negative correlation with resting mast cells. IGF1 had a positive correlation with activated DCs and a negative correlation with monocytes. The targeted drugs were dusigitumab and xentuzumab target for IGF1. CONCLUSION We analyzed the transcriptomic signature of LN along with the landscape of the immune cell. FOS and IGF1 are promising biomarkers for diagnosing and evaluating the progression of LN. The drug-gene interaction analyses provide a list of candidate drugs for the precise treatment of LN.
Collapse
Affiliation(s)
- Weiyi Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Zien Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Xiaobing Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Zhishen Peng
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Xiaofeng Liang
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Shanshan Wei
- Department of Dermatology, Zhujiang Hospital, 70570Southern Medical University, Guangzhou, The People's Republic of China
| |
Collapse
|
9
|
Zhong W, Wang X, Wang Y, Sun G, Zhang J, Li Z. Obesity and endocrine-related cancer: The important role of IGF-1. Front Endocrinol (Lausanne) 2023; 14:1093257. [PMID: 36755926 PMCID: PMC9899991 DOI: 10.3389/fendo.2023.1093257] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Obesity is increasingly becoming a global epidemic of concern and is considered a risk factor for several endocrine-related cancers. Moreover, obesity is associated with cancer development and poor prognosis. As a metabolic abnormality, obesity leads to a series of changes in insulin, IGF-1, sex hormones, IGFBPs, and adipokines. Among these factors, IGF-1 plays an important role in obesity-related endocrine cancers. This review describes the role of obesity in endocrine-related cancers, such as prostate cancer, breast cancer and pancreatic cancer, focusing on the mechanism of IGF-1 and the crosstalk with estrogen and adipokines. In addition, this review briefly introduces the current status of IGF-1R inhibitors in clinical practice and shows the prospect of IGF-1R inhibitors in combination with other anticancer drugs.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhuo Li
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Lee JS, Tocheny CE, Shaw LM. The Insulin-like Growth Factor Signaling Pathway in Breast Cancer: An Elusive Therapeutic Target. LIFE (BASEL, SWITZERLAND) 2022; 12:life12121992. [PMID: 36556357 PMCID: PMC9782138 DOI: 10.3390/life12121992] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
In this review, we provide an overview of the role of the insulin-like growth factor (IGF) signaling pathway in breast cancer and discuss its potential as a therapeutic target. The IGF pathway ligands, IGF-1 and IGF-2, and their receptors, primarily IGF-1R, are important for normal mammary gland biology, and dysregulation of their expression and function drives breast cancer risk and progression through activation of downstream signaling effectors, often in a subtype-dependent manner. The IGF signaling pathway has also been implicated in resistance to current therapeutic strategies, including ER and HER2 targeting drugs. Unfortunately, efforts to target IGF signaling for the treatment of breast cancer have been unsuccessful, due to a number of factors, most significantly the adverse effects of disrupting IGF signaling on normal glucose metabolism. We highlight here the recent discoveries that provide enthusiasm for continuing efforts to target IGF signaling for the treatment of breast cancer patients.
Collapse
Affiliation(s)
| | | | - Leslie M. Shaw
- Correspondence: ; Tel.: +1-508-856-8675; Fax: +1-508-856-1310
| |
Collapse
|
11
|
Kang J, Guo Z, Zhang H, Guo R, Zhu X, Guo X. Dual Inhibition of EGFR and IGF-1R Signaling Leads to Enhanced Antitumor Efficacy against Esophageal Squamous Cancer. Int J Mol Sci 2022; 23:ijms231810382. [PMID: 36142299 PMCID: PMC9499412 DOI: 10.3390/ijms231810382] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/02/2022] Open
Abstract
Both the epidermal growth factor receptor (EGFR) and insulin-like growth factor 1 receptor (IGF-1R) have been implicated in the development of cancers, and the increased expression of both receptors has been observed in esophageal cancer. However, the tyrosine kinase inhibitors of both receptors have thus far failed to provide clinical benefits for esophageal cancer patients. Studies have confirmed the complicated crosstalks that exist between the EGFR and IGF-1R pathways. The EGFR and IGF-1R signals act as mutual compensation pathways, thereby conveying resistance to EGFR or IGF-1R inhibitors when used alone. This study evaluated the antitumor efficacy of the EGFR/HER2 inhibitors, gefitinib and lapatinib, in combination with the IGF-1R inhibitor, linsitinib, on the esophageal squamous cell carcinoma (ESCC). Gefitinib or lapatinib, in combination with linsitinib, synergistically inhibited the proliferation, migration, and invasion of ESCC cells, caused significant cell cycle arrest, and induced marked cell apoptosis. Their combination demonstrated stronger inhibition on the activation of EGFR, HER2, and IGF-1R as well as the downstream signaling molecules. In vivo, the addition of linsitinib to gefitinib or lapatinib also potentiated the inhibition effects on the growth of xenografts. Our results suggest the next clinical exploration of the combination of gefitinib or lapatinib with linsitinib in the treatment of ESCC patients.
Collapse
Affiliation(s)
- Jia Kang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Pathogenic Biology, Xinxiang 453003, China
| | - Zanzan Guo
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang 453003, China
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang 453003, China
- Xinxiang Molecular and Immunodiagnostics Research Center for Engineering Technology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang 453003, China
| | - Haoqi Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Pathogenic Biology, Xinxiang 453003, China
| | - Rongqi Guo
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang 453003, China
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang 453003, China
- Xinxiang Molecular and Immunodiagnostics Research Center for Engineering Technology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang 453003, China
| | - Xiaofei Zhu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang 453003, China
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang 453003, China
- Xinxiang Molecular and Immunodiagnostics Research Center for Engineering Technology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang 453003, China
- Correspondence: (X.Z.); (X.G.)
| | - Xiaofang Guo
- Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Pathogenic Biology, Xinxiang 453003, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang 453003, China
- Correspondence: (X.Z.); (X.G.)
| |
Collapse
|
12
|
Diverse Mechanisms of Resistance against Osimertinib, a Third-Generation EGFR-TKI, in Lung Adenocarcinoma Cells with an EGFR-Activating Mutation. Cells 2022; 11:cells11142201. [PMID: 35883645 PMCID: PMC9319811 DOI: 10.3390/cells11142201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Osimertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), is used as a first-line treatment for patients with EGFR-mutant non-small cell lung cancer (NSCLC). However, the mechanisms underlying its anticancer activity, particularly the subsequent development of acquired resistance, are unclear. Herein, we investigated the mechanisms underlying the development of osimertinib resistance by treating NSCLC PC-9 cells (harboring an EGFR-activating mutation) with osimertinib, thereby developing five resistant cell lines, i.e., AZDR3, AZDR6, AZDR9, AZDR11, and AZDR14. The amplification of wild-type EGFR in AZDR3 cells and wild-type EGFR and KRAS in AZDR6 cells was also studied. AZDR3 cells showed dependence on EGFR signaling, in addition to afatinib sensitivity. AZDR9 cells harboring KRASG13D showed sensitivity to MEK inhibitors. Furthermore, combination treatment with EGFR and IGF1R inhibitors resulted in attenuated cell proliferation and enhanced apoptosis. In AZDR11 cells, increased Bim expression could not induce apoptosis, but Bid cleavage was found to be essential for the same. A SHP2/T507K mutation was also identified in AZDR14 cells, and, when associated with GAB1, SHP2 could activate ERK1/2, whereas a SHP2 inhibitor, TNO155, disrupted this association, thereby inhibiting GAB1 activation. Thus, diverse osimertinib resistance mechanisms were identified, providing insights for developing novel therapeutic strategies for NSCLC.
Collapse
|
13
|
Wang P, Mak VCY, Cheung LWT. Drugging IGF-1R in cancer: New insights and emerging opportunities. Genes Dis 2022; 10:199-211. [PMID: 37013053 PMCID: PMC10066341 DOI: 10.1016/j.gendis.2022.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/02/2022] [Indexed: 11/19/2022] Open
Abstract
The insulin-like growth factor (IGF) axis plays important roles in cancer development and metastasis. The type 1 IGF receptor (IGF-1R) is a key member in the IGF axis and has long been recognized for its oncogenic role in multiple cancer lineages. Here we review the occurrence of IGF-1R aberrations and activation mechanisms in cancers, which justify the development of anti-IGF-1R therapies. We describe the therapeutic agents available for IGF-1R inhibition, with focuses on the recent or ongoing pre-clinical and clinical studies. These include antisense oligonucleotide, tyrosine kinase inhibitors and monoclonal antibodies which may be conjugated with cytotoxic drug. Remarkably, simultaneous targeting of IGF-1R and several other oncogenic vulnerabilities has shown early promise, highlighting the potential benefits of combination therapy. Further, we discuss the challenges in targeting IGF-1R so far and new concepts to improve therapeutic efficacy such as blockage of the nuclear translocation of IGF-1R.
Collapse
|
14
|
Wu X, Seraia E, Hatch SB, Wan X, Ebner DV, Aroldi F, Jiang Y, Ryan AJ, Bogenrieder T, Weyer-Czernilofsky U, Rieunier G, Macaulay VM. CHK1 inhibition exacerbates replication stress induced by IGF blockade. Oncogene 2022; 41:476-488. [PMID: 34773074 PMCID: PMC8782724 DOI: 10.1038/s41388-021-02080-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 11/18/2022]
Abstract
We recently reported that genetic or pharmacological inhibition of insulin-like growth factor receptor (IGF-1R) slows DNA replication and induces replication stress by downregulating the regulatory subunit RRM2 of ribonucleotide reductase, perturbing deoxynucleotide triphosphate (dNTP) supply. Aiming to exploit this effect in therapy we performed a compound screen in five breast cancer cell lines with IGF neutralising antibody xentuzumab. Inhibitor of checkpoint kinase CHK1 was identified as a top screen hit. Co-inhibition of IGF and CHK1 caused synergistic suppression of cell viability, cell survival and tumour growth in 2D cell culture, 3D spheroid cultures and in vivo. Investigating the mechanism of synthetic lethality, we reveal that CHK1 inhibition in IGF-1R depleted or inhibited cells further downregulated RRM2, reduced dNTP supply and profoundly delayed replication fork progression. These effects resulted in significant accumulation of unreplicated single-stranded DNA and increased cell death, indicative of replication catastrophe. Similar phenotypes were induced by IGF:WEE1 co-inhibition, also via exacerbation of RRM2 downregulation. Exogenous RRM2 expression rescued hallmarks of replication stress induced by co-inhibiting IGF with CHK1 or WEE1, identifying RRM2 as a critical target of the functional IGF:CHK1 and IGF:WEE1 interactions. These data identify novel therapeutic vulnerabilities and may inform future trials of IGF inhibitory drugs.
Collapse
Affiliation(s)
- Xiaoning Wu
- Department of Oncology, University of Oxford, Oxford, UK
| | - Elena Seraia
- Target Discovery Institute, University of Oxford, Oxford, UK
| | | | - Xiao Wan
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - Daniel V Ebner
- Target Discovery Institute, University of Oxford, Oxford, UK
| | | | - Yanyan Jiang
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Anderson J Ryan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- AMAL Therapeutics, c/o Fondation pour Recherches Médicales, 1205 Geneva, Switzerland
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | | | - Guillaume Rieunier
- Department of Oncology, University of Oxford, Oxford, UK.
- Immunocore Ltd, Abingdon, UK.
| | | |
Collapse
|
15
|
Doi T, Kuboki Y, Naito Y, Ishida M, Tanaka T, Takeuchi Y. A phase 1 trial of xentuzumab, an IGF-neutralizing antibody, in Japanese patients with advanced solid tumors. Cancer Sci 2021; 113:1010-1017. [PMID: 34870878 PMCID: PMC8898728 DOI: 10.1111/cas.15231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/10/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
Xentuzumab is an insulin‐like growth factor (IGF) ligand‐neutralizing antibody. This phase 1 trial assessed xentuzumab in Japanese patients with solid tumors. Patients aged ≥20 y old with solid tumors that were refractory or not amenable to standard therapy were enrolled. Patients received xentuzumab intravenously at a starting dose of 750 mg/wk. Dose escalation used a 3 + 3 design with dose de‐escalation. The primary endpoint was to determine the maximum tolerated dose (MTD) of xentuzumab. Safety, pharmacokinetics, pharmacodynamics, and anti‐tumor activity were also assessed. Fifteen patients received xentuzumab in the dose escalation part (750 mg/wk [n = 6]; 1000 mg/wk [n = 3]; 1400 mg/wk [n = 6]). There were no dose‐limiting toxicities at any dose; the MTD of xentuzumab was not reached. Xentuzumab 1000 mg/wk was recommended as the relevant biological dose. Six further patients received xentuzumab 1000 mg/wk in an expansion cohort. Of 21 patients, 13 (61.9%) experienced a drug‐related adverse event, most commonly fatigue (23.8%), neutropenia (19.0%), diarrhea, nausea, white blood cell count decrease, and muscle spasms (14.3% each). No relevant deviations from dose linearity of xentuzumab exposure were observed during dose escalation. Total IGF‐1 and IGF‐2 levels increased and bioactive IGF levels decreased from baseline to 24 h after the first infusion in cycle 1. Partial response was observed in 2 (9.5%) patients with desmoid‐type fibromatosis. Disease control was achieved in 6 (28.6%) patients (median duration 42.4 mo). Xentuzumab monotherapy was well tolerated in Japanese patients and showed evidence of anti‐tumor activity. This study was registered with www.clinicaltrials.gov (NCT02145741).
Collapse
Affiliation(s)
- Toshihiko Doi
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yasutoshi Kuboki
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoichi Naito
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | | |
Collapse
|
16
|
Randomized Phase II Trial of Capecitabine and Lapatinib with or without IMC-A12 (Cituxumumab) in Patients with HER2-Positive Advanced Breast Cancer Previously Treated with Trastuzumab and Chemotherapy: NCCTG N0733 (Alliance). Breast Cancer Res Treat 2021; 188:477-487. [PMID: 33852121 PMCID: PMC8262517 DOI: 10.1007/s10549-021-06221-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/30/2021] [Indexed: 02/01/2023]
Abstract
PURPOSE To compare efficacy and safety of capecitabine and lapatinib with or without IMC-A12 (cituxumumab) in patients with HER2-positive metastatic breast cancer (MBC) previously treated with trastuzumab. PATIENTS AND METHODS Following an initial safety run-in cohort, patients were randomized 1:2 to Arm A (capecitabine and lapatinib) or to Arm B (capecitabine, lapatinib, and cituxumumab). Given the frequency of non-hematologic grade ≥ 3 adverse events in those receiving the three-drug combination in the safety cohort, lapatinib and capecitabine doses were reduced in Arm B only. The primary objective was to determine if the addition of cituxumumab to capecitabine and lapatinib improved progression-free survival (PFS) compared with capecitabine and lapatinib. Secondary objectives included a comparison between arms of other clinical endpoints, safety, change in overall quality of life (QOL) and self-assessed fatigue, rash, diarrhea, and hand-foot syndrome. RESULTS From July 2008 to March 2012, 68 patients (out of 142 planned) were enrolled and 63 were evaluable, including 8 for the safety run-in and 55 for the randomized cohort. Study enrollment was stopped early due to slow accrual. The addition of cituxumumab to capecitabine and lapatinib did not improve PFS (HR 0.93, 95% CI: 0.52-1.64). Furthermore, no difference in objective response rate or overall survival (OS) was observed. No difference between arms was observed in grade ≥ 3 adverse events, overall QOL change from baseline after 4 cycles of treatment. CONCLUSION The addition of cituxumumab to lapatinib and capecitabine did not improve PFS or OS compared with lapatinib and capecitabine in patients with HER2-positive MBC. CLINICAL TRIAL REGISTRY ClinicalTrials.gov Identifier: NCT00684983.
Collapse
|
17
|
Disruption of FOXO3a-miRNA feedback inhibition of IGF2/IGF-1R/IRS1 signaling confers Herceptin resistance in HER2-positive breast cancer. Nat Commun 2021; 12:2699. [PMID: 33976188 PMCID: PMC8113606 DOI: 10.1038/s41467-021-23052-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 04/13/2021] [Indexed: 02/03/2023] Open
Abstract
Resistance to Herceptin represents a significant challenge for successful treatment of HER2-positive breast cancer. Here, we show that in Herceptin-sensitive cells, FOXO3a regulates specific miRNAs to control IGF2 and IRS1 expression, retaining basic IGF2/IGF-1R/IRS1 signaling. The basic activity maintains expression of PPP3CB, a subunit of the serine/threonine-protein phosphatase 2B, to restrict FOXO3a phosphorylation (p-FOXO3a), inducing IGF2- and IRS1-targeting miRNAs. However, in Herceptin-resistant cells, p-FOXO3a levels are elevated due to transcriptional suppression of PPP3CB, disrupting the negative feedback inhibition loop formed by FOXO3a and the miRNAs, thereby upregulating IGF2 and IRS1. Moreover, we detect significantly increased IGF2 in blood and IRS1 in the tumors of breast cancer patients with poor response to Herceptin-containing regimens. Collectively, we demonstrate that the IGF2/IGF-1R/IRS1 signaling is aberrantly activated in Herceptin-resistant breast cancer via disruption of the FOXO3a-miRNA negative feedback inhibition. Such insights provide avenues to identify predictive biomarkers and effective strategies overcoming Herceptin resistance.
Collapse
|
18
|
Badarni M, Prasad M, Golden A, Bhattacharya B, Levin L, Yegodayev KM, Dimitstein O, Joshua BZ, Cohen L, Khrameeva E, Kong D, Porgador A, Braiman A, Grandis JR, Rotblat B, Elkabets M. IGF2 Mediates Resistance to Isoform-Selective-Inhibitors of the PI3K in HPV Positive Head and Neck Cancer. Cancers (Basel) 2021; 13:cancers13092250. [PMID: 34067117 PMCID: PMC8125641 DOI: 10.3390/cancers13092250] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary In the current study, we delineate the molecular mechanisms of acquisition of resistance to two isoform-selective inhibitors of PI3K (isiPI3K), alpelisib and taselisib, in human papillomavirus positive head and neck cell lines. By comparing RNA sequencing of isiPI3K-sensitive tumor cells and their corresponding isiPI3K-acquired-resistant tumor cells, we found that overexpression of insulin growth factor 2 (IGF2) is associated with the resistance phenotype. We further demonstrated by gain and loss of function studies that IGF2 plays a causative role in limiting the sensitivity of human papillomavirus-positive head and neck cell lines. Moreover, we show that blocking IGF2 stimulation activity, using an inhibitor of the IGF1 receptor (IGF1R), enhances isiPI3K efficacy and displays a synergistic anti-tumor effect in vitro and superior anti-tumor activity ex vivo and in vivo. Abstract Over 50% of human papilloma positive head-and-neck cancer (HNCHPV+) patients harbor genomic-alterations in PIK3CA, leading to hyperactivation of the phosphatidylinositol-4, 5-bisphosphate 3-kinase (PI3K) pathway. Nevertheless, despite PI3K pathway activation in HNCHPV+ tumors, the anti-tumor activities of PI3K pathway inhibitors are moderate, mostly due to the emergence of resistance. Thus, for potent and long-term tumor management, drugs blocking resistance mechanisms should be combined with PI3K inhibitors. Here, we delineate the molecular mechanisms of the acquisition of resistance to two isoform-selective inhibitors of PI3K (isiPI3K), alpelisib (BYL719) and taselisib (GDC0032), in HNCHPV+ cell lines. By comparing the transcriptional landscape of isiPI3K-sensitive tumor cells with that of their corresponding isiPI3K-acquired-resistant tumor cells, we found upregulation of insulin growth factor 2 (IGF2) in the resistant cells. Mechanistically, we show that upon isiPI3K treatment, isiPI3K-sensitive tumor cells upregulate the expression of IGF2 to induce cell proliferation via the activation of the IGF1 receptor (IGF1R). Stimulating tumor cells with recombinant IGF2 limited isiPI3K efficacy and released treated cells from S phase arrest. Knocking-down IGF2 with siRNA, or blocking IGF1R with AEW541, resulted in superior anti-tumor activity of isiPI3K in vitro and ex vivo. In vivo, the combination of isiPI3K and IGF1R inhibitor induced stable disease in mice bearing either tumors generated by the HNCHPV+ UM-SCC47 cell line or HPV+ patient-derived xenografts. These findings indicate that IGF2 and the IGF2/IGF1R pathway may constitute new targets for combination therapies to enhance the efficacy of PI3K inhibitors for the treatment of HNCHPV+.
Collapse
Affiliation(s)
- Mai Badarni
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Manu Prasad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Artemiy Golden
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.G.); (E.K.)
| | - Baisali Bhattacharya
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Liron Levin
- Bioinformatics Core Facility, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ksenia M. Yegodayev
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Orr Dimitstein
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
- Department of Otolaryngology—Head and Neck Surgery, Soroka University Medical Center, Beer-Sheva 84105, Israel
| | - Ben-Zion Joshua
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
- Department of Otorhinolaryngology and Head & Neck Surgery, Barzilay Medical Center, Ashkelon 7830604, Israel
| | - Limor Cohen
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Ekaterina Khrameeva
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.G.); (E.K.)
| | - Dexin Kong
- School of Pharmaceutical Sciences, Tianjin Medical University, Tianjin 300070, China;
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
| | - Jennifer R. Grandis
- Department of Otolaryngology—Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Barak Rotblat
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Life Sciences, Faculty of Life Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Correspondence: (B.R.); (M.E.); Tel.: +972-(0)8-6428806 (B.R.); +972-86428846 (M.E.)
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (M.B.); (M.P.); (B.B.); (K.M.Y.); (L.C.); (A.P.); (A.B.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (O.D.); (B.-Z.J.)
- Correspondence: (B.R.); (M.E.); Tel.: +972-(0)8-6428806 (B.R.); +972-86428846 (M.E.)
| |
Collapse
|
19
|
Zhang M, Yu GY, Liu G, Liu WD. Circular RNA circ_0002137 regulated the progression of osteosarcoma through regulating miR-433-3p/ IGF1R axis. J Cell Mol Med 2021; 26:1806-1816. [PMID: 33621401 PMCID: PMC8918411 DOI: 10.1111/jcmm.16166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
Current clinical treatment targeting osteosarcoma (OS) are limited for OS patients with pulmonary metastasis or relapse, which led to high mortality (70%‐85%) for advanced osteosarcoma patients. Although ongoing efforts have been made to illustrate the mechanisms of tumorigenesis and progression in OS; however, it was far for us to learn a comprehensive molecular mechanism implies in OS development. In our study, we implicated a circRNA hsa_circ_0002137, which was higher expressed in osteosarcoma tumours compared with paracancerous tissue. The dysregulated expression pattern was also found in osteosarcoma cell lines. The role of circ_0002137 was explored via down‐ or up‐regulated experiments. It was proved that down‐regulation of circ_0002137 suppressed the progress of OS, including cell invasion, cell cycle and cell apoptosis. Furthermore, the correlation between circ_0002137 and miR‐433‐3p was predicted using bioinformatic tools and verified utilizing RNA pull‐down assay and luciferase reporter assay. Interestingly, we found that the inhibitory effect of circ_0002137 on OS was dependent of insulin‐like growth factor‐1 receptor (IGF1R). In conclusion, it was demonstrated that circ_0002137 could restrain the progression of OS through regulating miR‐433‐3p/IGF1R axis, providing a comprehensive landscape of circ_0002137 in the generation and development of OS.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Orthopedic, The Affiliated Huaian NO. 1 people's Hospital of Nanjing Medical University, Huaian, China
| | - Guang-Yang Yu
- Department of Orthopedic, The Affiliated Huaian NO. 1 people's Hospital of Nanjing Medical University, Huaian, China
| | - Gang Liu
- Department of Orthopedic, The Affiliated Huaian NO. 1 people's Hospital of Nanjing Medical University, Huaian, China
| | - Wei-Dong Liu
- Department of Orthopedic, The Affiliated Huaian NO. 1 people's Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
20
|
Rieunier G, Wu X, Harris LE, Mills JV, Nandakumar A, Colling L, Seraia E, Hatch SB, Ebner DV, Folkes LK, Weyer-Czernilofsky U, Bogenrieder T, Ryan AJ, Macaulay VM. Targeting IGF Perturbs Global Replication through Ribonucleotide Reductase Dysfunction. Cancer Res 2021; 81:2128-2141. [PMID: 33509941 DOI: 10.1158/0008-5472.can-20-2860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/17/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
Inhibition of IGF receptor (IGF1R) delays repair of radiation-induced DNA double-strand breaks (DSB), prompting us to investigate whether IGF1R influences endogenous DNA damage. Here we demonstrate that IGF1R inhibition generates endogenous DNA lesions protected by 53BP1 bodies, indicating under-replicated DNA. In cancer cells, inhibition or depletion of IGF1R delayed replication fork progression accompanied by activation of ATR-CHK1 signaling and the intra-S-phase checkpoint. This phenotype reflected unanticipated regulation of global replication by IGF1 mediated via AKT, MEK/ERK, and JUN to influence expression of ribonucleotide reductase (RNR) subunit RRM2. Consequently, inhibition or depletion of IGF1R downregulated RRM2, compromising RNR function and perturbing dNTP supply. The resulting delay in fork progression and hallmarks of replication stress were rescued by RRM2 overexpression, confirming RRM2 as the critical factor through which IGF1 regulates replication. Suspecting existence of a backup pathway protecting from toxic sequelae of replication stress, targeted compound screens in breast cancer cells identified synergy between IGF inhibition and ATM loss. Reciprocal screens of ATM-proficient/deficient fibroblasts identified an IGF1R inhibitor as the top hit. IGF inhibition selectively compromised growth of ATM-null cells and spheroids and caused regression of ATM-null xenografts. This synthetic-lethal effect reflected conversion of single-stranded lesions in IGF-inhibited cells into toxic DSBs upon ATM inhibition. Overall, these data implicate IGF1R in alleviating replication stress, and the reciprocal IGF:ATM codependence we identify provides an approach to exploit this effect in ATM-deficient cancers. SIGNIFICANCE: This study identifies regulation of ribonucleotide reductase function and dNTP supply by IGFs and demonstrates that IGF axis blockade induces replication stress and reciprocal codependence on ATM. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2128/F1.large.jpg.
Collapse
Affiliation(s)
| | - Xiaoning Wu
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Letitia E Harris
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Jack V Mills
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ashwin Nandakumar
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Laura Colling
- Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
| | - Elena Seraia
- Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Stephanie B Hatch
- Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Daniel V Ebner
- Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Lisa K Folkes
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Thomas Bogenrieder
- AMAL Therapeutics, Geneva, Switzerland
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Anderson J Ryan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Valentine M Macaulay
- Department of Oncology, University of Oxford, Oxford, United Kingdom.
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
21
|
Schmid P, Sablin MP, Bergh J, Im SA, Lu YS, Martínez N, Neven P, Lee KS, Morales S, Pérez-Fidalgo JA, Adamson D, Gonçalves A, Prat A, Jerusalem G, Schlieker L, Espadero RM, Bogenrieder T, Huang DCL, Crown J, Cortés J. A phase Ib/II study of xentuzumab, an IGF-neutralising antibody, combined with exemestane and everolimus in hormone receptor-positive, HER2-negative locally advanced/metastatic breast cancer. Breast Cancer Res 2021; 23:8. [PMID: 33451345 PMCID: PMC7811234 DOI: 10.1186/s13058-020-01382-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
Background Xentuzumab—a humanised IgG1 monoclonal antibody—binds IGF-1 and IGF-2, inhibiting their growth-promoting signalling and suppressing AKT activation by everolimus. This phase Ib/II exploratory trial evaluated xentuzumab plus everolimus and exemestane in hormone receptor-positive, locally advanced and/or metastatic breast cancer (LA/MBC). Methods Patients with hormone receptor-positive/HER2-negative LA/MBC resistant to non-steroidal aromatase inhibitors were enrolled. Maximum tolerated dose (MTD) and recommended phase II dose (RP2D) of xentuzumab/everolimus/exemestane were determined in phase I (single-arm, dose-escalation). In phase II (open-label), patients were randomised 1:1 to the RP2D of xentuzumab/everolimus/exemestane or everolimus/exemestane alone. Randomisation was stratified by the presence of visceral metastases. Primary endpoint was progression-free survival (PFS). Results MTD was determined as xentuzumab 1000 mg weekly plus everolimus 10 mg/day and exemestane 25 mg/day. A total of 140 patients were enrolled in phase II (70 to each arm). Further recruitment was stopped following an unfavourable benefit-risk assessment by the internal Data Monitoring Committee appointed by the sponsor. Xentuzumab was discontinued; patients could receive everolimus/exemestane if clinically indicated. Median PFS was 7.3 months (95% CI 3.3–not calculable) in the xentuzumab/everolimus/exemestane group and 5.6 months (3.7–9.1) in the everolimus/exemestane group (hazard ratio 0.97, 95% CI 0.57–1.65; P = 0.9057). In a pre-specified subgroup of patients without visceral metastases at screening, xentuzumab/everolimus/exemestane showed evidence of PFS benefit versus everolimus/exemestane (hazard ratio 0.21 [0.05–0.98]; P = 0.0293). Most common any-cause adverse events in phase II were diarrhoea (29 [41.4%] in the xentuzumab/everolimus/exemestane group versus 20 [29.0%] in the everolimus/exemestane group), mucosal inflammation (27 [38.6%] versus 21 [30.4%]), stomatitis (24 [34.3%] versus 24 [34.8%]), and asthenia (21 [30.0%] versus 24 [34.8%]). Conclusions Addition of xentuzumab to everolimus/exemestane did not improve PFS in the overall population, leading to early discontinuation of the trial. Evidence of PFS benefit was observed in patients without visceral metastases when treated with xentuzumab/everolimus/exemestane, leading to initiation of the phase II XENERA™-1 trial (NCT03659136). Trial registration ClinicalTrials.gov, NCT02123823. Prospectively registered, 8 March 2013. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-020-01382-8.
Collapse
Affiliation(s)
- Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Marie-Paule Sablin
- Department of Drug Development and Innovation, Institut Curie, Paris, France
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet and Breast Cancer Centre, Cancer Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yen-Shen Lu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Noelia Martínez
- Department of Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Patrick Neven
- Department of Oncology, UZ Leuven, Campus Gasthuisberg, Leuven, Belgium
| | - Keun Seok Lee
- Department of Internal Medicine, National Cancer Center, Goyang, South Korea
| | - Serafín Morales
- Department of Medical Oncology, Hospital Universitario Arnau de Vilanova de Lleida, Lleida, Spain
| | - J Alejandro Pérez-Fidalgo
- Medical Oncology Unit, Hospital Clinico Universitario Valencia, Biomedical Research Institute INCLIVA, CIBERONC, Valencia, Spain
| | - Douglas Adamson
- Department of Medical Oncology, Ninewells Hospital, Tayside Cancer Centre, Dundee, UK
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli Calmettes, Aix-Marseille University, CRCM, CNRS, INSERM, Marseille, France
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Guy Jerusalem
- Department of Medical Oncology, Centre Hospitalier Universitaire de Liège, and Liège University, Liège, Belgium
| | - Laura Schlieker
- External statistician on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG., Staburo GmbH & Co. KG., Munich, Germany
| | - Rosa-Maria Espadero
- Medical Department (Clinical Operations), Boehringer Ingelheim España S.A, Barcelona, Spain
| | - Thomas Bogenrieder
- Medical Department, Boehringer Ingelheim, RCV, Vienna, Austria.,Present Address: Amal Therapeutics SA, Geneva, Switzerland
| | - Dennis Chin-Lun Huang
- Medical Department, Boehringer Ingelheim Taiwan Limited, Taipei, Taiwan.,Present Address: MSD Taiwan, Taipei, Taiwan
| | - John Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Javier Cortés
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Department of Oncology, IOB Institute of Oncology, Quironsalud Group, Madrid and Barcelona, Spain
| |
Collapse
|
22
|
Disrupting Insulin and IGF Receptor Function in Cancer. Int J Mol Sci 2021; 22:ijms22020555. [PMID: 33429867 PMCID: PMC7827299 DOI: 10.3390/ijms22020555] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
The insulin and insulin-like growth factor (IGF) system plays an important role in regulating normal cell proliferation and survival. However, the IGF system is also implicated in many malignancies, including breast cancer. Preclinical studies indicate several IGF blocking approaches, such as monoclonal antibodies and tyrosine kinase inhibitors, have promising therapeutic potential for treating diseases. Uniformly, phase III clinical trials have not shown the benefit of blocking IGF signaling compared to standard of care arms. Clinical and laboratory data argue that targeting Type I IGF receptor (IGF1R) alone may be insufficient to disrupt this pathway as the insulin receptor (IR) may also be a relevant cancer target. Here, we review the well-studied role of the IGF system in regulating malignancies, the limitations on the current strategies of blocking the IGF system in cancer, and the potential future directions for targeting the IGF system.
Collapse
|
23
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020; 11:1986. [PMID: 32983137 PMCID: PMC7485114 DOI: 10.3389/fimmu.2020.01986] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage–derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
24
|
Abstract
The insulin and insulin-like growth factor (IGF) family of proteins are part of a complex network that regulates cell proliferation and survival. While this system is undoubtedly important in prenatal development and postnatal cell growth, members of this family have been implicated in several different cancer types. Increased circulating insulin and IGF ligands have been linked to increased risk of cancer incidence. This observation has led to targeting the IGF system as a therapeutic strategy in a number of cancers. This chapter aims to describe the well-characterized biology of the IGF1R system, outline the rationale for targeting this system in cancer, summarize the clinical data as it stands, and discuss where we can go from here.
Collapse
|
25
|
de Bono J, Lin CC, Chen LT, Corral J, Michalarea V, Rihawi K, Ong M, Lee JH, Hsu CH, Yang JCH, Shiah HS, Yen CJ, Anthoney A, Jove M, Buschke S, Fuertig R, Schmid U, Goeldner RG, Strelkowa N, Huang DCL, Bogenrieder T, Twelves C, Cheng AL. Two first-in-human studies of xentuzumab, a humanised insulin-like growth factor (IGF)-neutralising antibody, in patients with advanced solid tumours. Br J Cancer 2020; 122:1324-1332. [PMID: 32161368 PMCID: PMC7188670 DOI: 10.1038/s41416-020-0774-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/05/2020] [Accepted: 02/17/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Xentuzumab, an insulin-like growth factor (IGF)-1/IGF-2-neutralising antibody, binds IGF-1 and IGF-2, inhibiting their growth-promoting signalling. Two first-in-human trials assessed the maximum-tolerated/relevant biological dose (MTD/RBD), safety, pharmacokinetics, pharmacodynamics, and activity of xentuzumab in advanced/metastatic solid cancers. METHODS These phase 1, open-label trials comprised dose-finding (part I; 3 + 3 design) and expansion cohorts (part II; selected tumours; RBD [weekly dosing]). Primary endpoints were MTD/RBD. RESULTS Study 1280.1 involved 61 patients (part I: xentuzumab 10-1800 mg weekly, n = 48; part II: 1000 mg weekly, n = 13); study 1280.2, 64 patients (part I: 10-3600 mg three-weekly, n = 33; part II: 1000 mg weekly, n = 31). One dose-limiting toxicity occurred; the MTD was not reached for either schedule. Adverse events were generally grade 1/2, mostly gastrointestinal. Xentuzumab showed dose-proportional pharmacokinetics. Total plasma IGF-1 increased dose dependently, plateauing at ~1000 mg/week; at ≥450 mg/week, IGF bioactivity was almost undetectable. Two partial responses occurred (poorly differentiated nasopharyngeal carcinoma and peripheral primitive neuroectodermal tumour). Integration of biomarker and response data by Bayesian Logistic Regression Modeling (BLRM) confirmed the RBD. CONCLUSIONS Xentuzumab was well tolerated; MTD was not reached. RBD was 1000 mg weekly, confirmed by BLRM. Xentuzumab showed preliminary anti-tumour activity. CLINICAL TRIAL REGISTRATION NCT01403974; NCT01317420.
Collapse
Affiliation(s)
- Johann de Bono
- Drug Development Unit, Royal Marsden Hospital & Institute of Cancer Research, Downs Road, Sutton, UK.
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - Li-Tzong Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138 Sheng Li Road, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, 100 Tzyou 1st Road, Kaohsiung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng Li Road, Tainan, Taiwan
| | - Jesus Corral
- Medical Oncology Department, Clinica Universidad de Navarra, Calle Marquesado de Sta. Marta 1, Madrid, Spain
| | - Vasiliki Michalarea
- Drug Development Unit, Royal Marsden Hospital & Institute of Cancer Research, Downs Road, Sutton, UK
| | - Karim Rihawi
- Drug Development Unit, Royal Marsden Hospital & Institute of Cancer Research, Downs Road, Sutton, UK
- Azienda Sanitaria Universitaria Integrata di Udine, Via Pozzuolo, 330, 33100, Udine, Italy
| | - Michael Ong
- The Ottawa Hospital Cancer Centre, 501 Smyth Road, Ottawa, ON, Canada
| | - Jih-Hsiang Lee
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - Chih-Hung Hsu
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - Her-Shyong Shiah
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, 250 Wuxing Street, Taipei, Taiwan
| | - Chia-Jui Yen
- Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138 Sheng Li Road, Tainan, Taiwan
| | - Alan Anthoney
- University of Leeds and Leeds Teaching Hospitals Trust, Beckett Street, Leeds, UK
| | - Maria Jove
- University of Leeds and Leeds Teaching Hospitals Trust, Beckett Street, Leeds, UK
| | - Susanne Buschke
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - René Fuertig
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Ulrike Schmid
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Rainer-Georg Goeldner
- Biostatistics and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Natalja Strelkowa
- Biostatistics and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Dennis Chin-Lun Huang
- Medical Department, Boehringer Ingelheim Taiwan Ltd, 12F, No. 2, Sec 3, Minsheng East Road, Taipei, Taiwan
| | - Thomas Bogenrieder
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, 1 Geschwister-Scholl-Platz, Munich, Germany
- Medicine and Translational Research, Boehringer Ingelheim RCV, 5-11 Doktor-Boehringer-Gasse, Vienna, Austria
| | - Chris Twelves
- University of Leeds and Leeds Teaching Hospitals Trust, Beckett Street, Leeds, UK
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
26
|
Weyer-Czernilofsky U, Hofmann MH, Friedbichler K, Baumgartinger R, Adam PJ, Solca F, Kraut N, Nguyen HM, Corey E, Liu G, Sprenger CC, Plymate SR, Bogenrieder T. Antitumor Activity of the IGF-1/IGF-2-Neutralizing Antibody Xentuzumab (BI 836845) in Combination with Enzalutamide in Prostate Cancer Models. Mol Cancer Ther 2020; 19:1059-1069. [PMID: 32054790 PMCID: PMC10823795 DOI: 10.1158/1535-7163.mct-19-0378] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 11/15/2019] [Accepted: 02/04/2020] [Indexed: 12/24/2022]
Abstract
Androgen deprivation therapy and second-generation androgen receptor signaling inhibitors such as enzalutamide are standard treatments for advanced/metastatic prostate cancer. Unfortunately, most men develop resistance and relapse; signaling via insulin-like growth factor (IGF) has been implicated in castration-resistant prostate cancer. We evaluated the antitumor activity of xentuzumab (IGF ligand-neutralizing antibody), alone and in combination with enzalutamide, in prostate cancer cell lines (VCaP, DuCaP, MDA PCa 2b, LNCaP, and PC-3) using established in vitro assays, and in vivo, using LuCaP 96CR, a prostate cancer patient-derived xenograft (PDX) model. Xentuzumab + enzalutamide reduced the viability of phosphatase and tensin homolog (PTEN)-expressing VCaP, DuCaP, and MDA PCa 2b cells more than either single agent, and increased antiproliferative activity and apoptosis induction in VCaP. Xentuzumab or xentuzumab + enzalutamide inhibited IGF type 1 receptor and AKT serine/threonine kinase (AKT) phosphorylation in VCaP, DuCaP, and MDA PCa 2b cells; xentuzumab had no effect on AKT phosphorylation and proliferation in PTEN-null LNCaP or PC-3 cells. Knockdown of PTEN led to loss of antiproliferative activity of xentuzumab and reduced activity of xentuzumab + enzalutamide in VCaP cells. Xentuzumab + enzalutamide inhibited the growth of castration-resistant LuCaP 96CR PDX with acquired resistance to enzalutamide, and improved survival in vivo The data suggest that xentuzumab + enzalutamide combination therapy may overcome castration resistance and could be effective in patients who are resistant to enzalutamide alone. PTEN status as a biomarker of responsiveness to combination therapy needs further investigation.
Collapse
Affiliation(s)
| | | | | | | | - Paul J Adam
- Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Gang Liu
- Department of Medicine and GRECC VAPSHCS, University of Washington, Seattle, Washington
| | - Cynthia C Sprenger
- Department of Medicine and GRECC VAPSHCS, University of Washington, Seattle, Washington
| | - Stephen R Plymate
- Department of Medicine and GRECC VAPSHCS, University of Washington, Seattle, Washington
| | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
27
|
Manabe T, Yasuda H, Terai H, Kagiwada H, Hamamoto J, Ebisudani T, Kobayashi K, Masuzawa K, Ikemura S, Kawada I, Hayashi Y, Fukui K, Horimoto K, Fukunaga K, Soejima K. IGF2 Autocrine-Mediated IGF1R Activation Is a Clinically Relevant Mechanism of Osimertinib Resistance in Lung Cancer. Mol Cancer Res 2020; 18:549-559. [PMID: 31941753 DOI: 10.1158/1541-7786.mcr-19-0956] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 01/06/2020] [Indexed: 11/16/2022]
Abstract
EGFR-mutated lung cancer accounts for a significant proportion of lung cancer cases worldwide. For these cases, osimertinib, a third-generation EGFR tyrosine kinase inhibitor, is extensively used as a first-line or second-line treatment. However, lung cancer cells acquire resistance to osimertinib in 1 to 2 years. Thus, a thorough clarification of resistance mechanisms to osimertinib is highly anticipated. Recent next-generation sequencing (NGS) of lung cancer samples identified several genetically defined resistance mechanisms to osimertinib, such as EGFR C797S or MET amplification. However, nongenetically defined mechanisms are not well evaluated. For a thorough clarification of osimertinib resistance, both genetic and nongenetic mechanisms are essential. By using our comprehensive protein phosphorylation array, we detected IGF1R bypass pathway activation after EGFR abolishment. Both of our established lung cancer cells and patient-derived lung cancer cells demonstrated IGF2 autocrine-mediated IGF1R pathway activation as a mechanism of osimertinib resistance. Notably, this resistance mechanism was not detected by a previously performed NGS, highlighting the essential roles of living cancer cells for a thorough clarification of resistance mechanisms. Interestingly, the immunohistochemical analysis confirmed the increased IGF2 expression in lung cancer patients who were treated with osimertinib and met the established clinical definition of acquired resistance. The findings highlight the crucial roles of cell-autonomous ligand expression in osimertinib resistance. Here, we report for the first time the IGF2 autocrine-mediated IGF1R activation as a nongenetic mechanism of osimertinib resistance in lung cancer at a clinically relevant level. IMPLICATIONS: Using comprehensive protein phosphorylation array and patient-derived lung cancer cells, we found that IGF2 autocrine-mediated IGF1R pathway activation is a clinically relevant and common mechanism of acquired resistance to osimertinib.
Collapse
Affiliation(s)
- Tadashi Manabe
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan.
| | - Hideki Terai
- Division of Pulmonary Medicine, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Harumi Kagiwada
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Junko Hamamoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Toshiki Ebisudani
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Keigo Kobayashi
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Keita Masuzawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | | | - Ichiro Kawada
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Yuichiro Hayashi
- Department of Diagnostic Pathology, Keio University, School of Medicine, Tokyo, Japan
| | - Kazuhiko Fukui
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Katsuhisa Horimoto
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kenzo Soejima
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020. [PMID: 32983137 DOI: 10.3389/fimmu.2020.01986/bibtex] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage-derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Werner H, Sarfstein R, Bruchim I. Investigational IGF1R inhibitors in early stage clinical trials for cancer therapy. Expert Opin Investig Drugs 2019; 28:1101-1112. [PMID: 31731883 DOI: 10.1080/13543784.2019.1694660] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: The insulin-like growth factors (IGFs) are a family of secreted peptide hormones with important roles in different cellular and organism functions. The biological activities of the IGFs are mediated by the IGF1 receptor (IGF1R), a cell surface, tyrosine kinase-containing heterotetramer that is linked to numerous cytoplasmic signaling cascades. The IGF1R displays potent antiapoptotic, pro-survival capacities and plays a key role in malignant transformation. Research has identified the IGF1R as a candidate therapeutic target in cancer.Areas covered: We offer a synopsis of ongoing efforts to target the IGF axis for therapeutic purposes. Our review includes a digest of early experimental work that led to the identification of IGF1R as a candidate therapeutic target in oncology.Expert opinion: Targeting of the IGF axis has yielded disappointing results in phase III trials, but it is important to learn from this to improve future trials in a rational manner. The potential of anti-IGF1R antibodies and small molecular weight inhibitors, alone or in combination with chemotherapy or other biological agents, should be investigated further in randomized studies. Moreover, the implementation of predictive biomarkers for patient selection will improve the outcome of future trials. Emerging personalized medicine could have a major impact on IGF1R targeting.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv, Israel
| | - Rive Sarfstein
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Bruchim
- Gynecologic Oncology Division, Hillel Yaffe Medical Center, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
30
|
Parra-Guillen ZP, Schmid U, Janda A, Freiwald M, Troconiz IF. Model-Informed Dose Selection for Xentuzumab, a Dual Insulin-Like Growth Factor-I/II-Neutralizing Antibody. Clin Pharmacol Ther 2019; 107:597-606. [PMID: 31562819 DOI: 10.1002/cpt.1648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/22/2019] [Indexed: 01/13/2023]
Abstract
Over the past decade, the insulin-like growth factor (IGF)-signaling pathway has gained substantial interest as potential therapeutic target in oncology. Xentuzumab, a humanized IgG1 monoclonal antibody, binds to IGF-I and IGF-II thereby inhibiting the downstream signaling essential for survival and tumor growth. This pathway is further regulated by circulating IGF binding proteins (IGFBPs). In this work, a mechanistic model characterizing the dynamics and interactions of IGFs, IGFBPs, and Xentuzumab has been developed to guide dose selection. Therefore, in vitro and in vivo literature information was combined with temporal IGF-I, IGF-II, and IGFBP-3 total plasma concentrations from two phase I studies. Based on the established quantitative framework, the time-course of free IGFs as ultimate drug targets not measured in clinics was predicted. Finally, a dose of 1000 mg/week-predicted to reduce free IGF-I and free IGF-II at steady-state by at least 90% and 64%, respectively-was suggested for phase II.
Collapse
Affiliation(s)
- Zinnia P Parra-Guillen
- Pharmacometrics & Systems Pharmacology Research Unit, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
| | - Ulrike Schmid
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Alvaro Janda
- Pharmacometrics & Systems Pharmacology Research Unit, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
| | - Matthias Freiwald
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Iñaki F Troconiz
- Pharmacometrics & Systems Pharmacology Research Unit, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
| |
Collapse
|
31
|
Qureishi A, Rieunier G, Shah KA, Aleksic T, Winter SC, Møller H, Macaulay VM. Radioresistant laryngeal cancers upregulate type 1 IGF receptor and exhibit increased cellular dependence on IGF and EGF signalling. Clin Otolaryngol 2019; 44:1026-1036. [PMID: 31536667 DOI: 10.1111/coa.13434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/01/2019] [Accepted: 09/13/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Patients failing radiotherapy for laryngeal squamous cell carcinoma (LSCC) often require salvage total laryngectomy which has major functional consequences, highlighting a need for biomarkers of radiotherapy resistance. In other tumour types, radioresistance has been linked to epidermal growth factor receptor (EGFR) and type 1 insulin-like growth factor receptor (IGF-1R). Here, we evaluated IGF-1R and EGFR as predictors and mediators of LSCC radioresistance. DESIGN We compared IGF-1R and EGFR immunohistochemical scores in patients with LSCC achieving long-term remission post-radiotherapy (n = 23), patients treated with primary laryngectomy (n = 22) or salvage laryngectomy following radiotherapy recurrence (n = 18). To model radioresistance in vitro, two LSCC cell lines underwent clinically relevant irradiation to 55 Gy in 2.75 Gy fractions. RESULTS Type 1 insulin-like growth factor receptor expression was higher in pre-treatment biopsies of radiotherapy failures compared with those in long-term remission and was upregulated post-radiotherapy. Patients undergoing primary laryngectomy had more advanced T/N stage and greater tumour IGF-1R content than those achieving long-term remission. Pre-treatment EGFR did not associate with radiotherapy outcomes but showed a trend to upregulation post-irradiation. In vitro, radiosensitivity was enhanced by inhibition of EGFR but not IGF. Repeated irradiation upregulated IGF-1R in BICR18 and SQ20B cells and EGFR in SQ20B, and enhanced SQ20B radioresistance. Repeatedly irradiated SQ20B_55 cells were not radiosensitised by inhibition of IGF and/or EGFR, but IGF-1R:EGFR co-inhibition suppressed baseline cell survival more effectively than blockade of either pathway alone, and more effectively than in parental cells. CONCLUSIONS Radiation upregulates IGF-1R and may enhance IGF/EGFR dependence, suggesting that IGF/EGFR blockade may have activity in LSCCs that recur post-radiotherapy.
Collapse
Affiliation(s)
- Ali Qureishi
- Department of Oncology, University of Oxford, Oxford, UK.,Nuffield Department of Surgery, University of Oxford and Department of Head and Neck Surgery, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | | | - Ketan A Shah
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Tamara Aleksic
- Department of Oncology, University of Oxford, Oxford, UK
| | - Stuart C Winter
- Nuffield Department of Surgery, University of Oxford and Department of Head and Neck Surgery, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Henrik Møller
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Valentine M Macaulay
- Department of Oncology, University of Oxford, Oxford, UK.,Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| |
Collapse
|
32
|
Osher E, Macaulay VM. Therapeutic Targeting of the IGF Axis. Cells 2019; 8:E895. [PMID: 31416218 PMCID: PMC6721736 DOI: 10.3390/cells8080895] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/04/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
The insulin like growth factor (IGF) axis plays a fundamental role in normal growth and development, and when deregulated makes an important contribution to disease. Here, we review the functions mediated by ligand-induced IGF axis activation, and discuss the evidence for the involvement of IGF signaling in the pathogenesis of cancer, endocrine disorders including acromegaly, diabetes and thyroid eye disease, skin diseases such as acne and psoriasis, and the frailty that accompanies aging. We discuss the use of IGF axis inhibitors, focusing on the different approaches that have been taken to develop effective and tolerable ways to block this important signaling pathway. We outline the advantages and disadvantages of each approach, and discuss progress in evaluating these agents, including factors that contributed to the failure of many of these novel therapeutics in early phase cancer trials. Finally, we summarize grounds for cautious optimism for ongoing and future studies of IGF blockade in cancer and non-malignant disorders including thyroid eye disease and aging.
Collapse
Affiliation(s)
- Eliot Osher
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | | |
Collapse
|
33
|
An engineered analog of insulin-like growth factor 1 with reduced immunogenicity and retained mitogenicity. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
34
|
Rieunier G, Wu X, Macaulay VM, Lee AV, Weyer-Czernilofsky U, Bogenrieder T. Bad to the Bone: The Role of the Insulin-Like Growth Factor Axis in Osseous Metastasis. Clin Cancer Res 2019; 25:3479-3485. [PMID: 30745299 DOI: 10.1158/1078-0432.ccr-18-2697] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/10/2019] [Accepted: 02/06/2019] [Indexed: 11/16/2022]
Abstract
Bone metastases are a frequent complication of cancer that are associated with considerable morbidity. Current treatments may temporarily palliate the symptoms of bone metastases but often fail to delay their progression. Bones provide a permissive environment because they are characterized by dynamic turnover, secreting factors required for bone maintenance but also stimulating the establishment and growth of metastases. Insulin-like growth factors (IGF) are the most abundant growth factors in bone and are required for normal skeletal development and function. Via activation of the IGF-1 receptors (IGF-1R) and variant insulin receptors, IGFs promote cancer progression, aggressiveness, and treatment resistance. Of specific relevance to bone biology, IGFs contribute to the homing, dormancy, colonization, and expansion of bone metastases. Furthermore, preclinical evidence suggests that tumor cells can be primed to metastasize to bone by a high IGF-1 environment in the primary tumor, suggesting that bone metastases may reflect IGF dependency. Therapeutic targeting of the IGF axis may therefore provide an effective method for treating bone metastases. Indeed, anti-IGF-1R antibodies, IGF-1R tyrosine kinase inhibitors, and anti-IGF-1/2 antibodies have demonstrated antitumor activity in preclinical models of prostate and breast cancer metastases, either alone or in combination with other agents. Several studies suggest that such treatments can inhibit bone metastases without affecting growth of the primary tumor. Although previous trials of anti-IGF-1R drugs have generated negative results in unselected patients, these considerations suggest that future clinical trials of IGF-targeted agents may be warranted in patients with bone metastases.
Collapse
Affiliation(s)
| | - Xiaoning Wu
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Thomas Bogenrieder
- RCV Medicine, Boehringer Ingelheim RCV, Vienna, Austria.,Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
35
|
Jeong I, Kang SK, Kwon WS, Kim HJ, Kim KH, Kim HM, Lee A, Lee SK, Bogenrieder T, Chung HC, Rha SY. Regulation of proliferation and invasion by the IGF signalling pathway in Epstein-Barr virus-positive gastric cancer. J Cell Mol Med 2018; 22:5899-5908. [PMID: 30247804 PMCID: PMC6237558 DOI: 10.1111/jcmm.13859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/07/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Several carcinomas including gastric cancer have been reported to contain Epstein-Barr virus (EBV) infection. EBV-associated gastric cancer (EBVaGC) is classified as one of four molecular subtypes of gastric cancer by The Cancer Genome Atlas (TCGA) group with increased immune-related signatures. Identification of EBV-dependent pathways with significant biological roles is needed for EBVaGC. To compare the biological changes between AGS gastric epithelial cells and EBV-infected AGS (AGS-EBV) cells, proliferation assay, CCK-8 assay, invasion assay, cell cycle analysis, RT-PCR, Western blot and ELISA were performed. BI836845, a humanized insulin-like growth factor (IGF) ligand-neutralizing antibody, was used for IGF-related signalling pathway inhibition. AGS-EBV cells showed slower proliferating rate and higher sensitivity to BI836845 compared to AGS cells. Moreover, invasiveness of AGS-EBV was increased than that of AGS, and BI836845 treatment significantly decreased the invasiveness of AGS-EBV. Although no apoptosis was detected, entry into the S phase of the cell cycle was delayed in BI836845-treated AGS-EBV cells. In conclusion, AGS-EBV cells seem to modulate their proliferation and invasion through the IGF signalling pathway. Inhibition of the IGF signalling pathway therefore could be a potential therapeutic strategy for EBVaGC.
Collapse
Affiliation(s)
- Inhye Jeong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Sun Kyoung Kang
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Jeong Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Kyoo Hyun Kim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Myong Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Andre Lee
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Department of Biological Sciences, Columbia University, New York, New York
| | - Suk Kyeong Lee
- Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria.,Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Hyun Cheol Chung
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Sun Young Rha
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Fajka-Boja R, Marton A, Tóth A, Blazsó P, Tubak V, Bálint B, Nagy I, Hegedűs Z, Vizler C, Katona RL. Increased insulin-like growth factor 1 production by polyploid adipose stem cells promotes growth of breast cancer cells. BMC Cancer 2018; 18:872. [PMID: 30185144 PMCID: PMC6126028 DOI: 10.1186/s12885-018-4781-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/29/2018] [Indexed: 02/08/2023] Open
Abstract
Background Adipose-tissue stem cells (ASCs) are subject of intensive research since their successful use in regenerative therapy. The drawback of ASCs is that they may serve as stroma for cancer cells and assist tumor progression. It is disquieting that ASCs frequently undergo genetic and epigenetic changes during their in vitro propagation. In this study, we describe the polyploidization of murine ASCs and the accompanying phenotypical, gene expressional and functional changes under long term culturing. Methods ASCs were isolated from visceral fat of C57BL/6 J mice, and cultured in vitro for prolonged time. The phenotypical changes were followed by microscopy and flow cytometry. Gene expressional changes were determined by differential transcriptome analysis and changes in protein expression were shown by Western blotting. The tumor growth promoting effect of ASCs was examined by co-culturing them with 4 T1 murine breast cancer cells. Results After five passages, the proliferation of ASCs decreases and cells enter a senescence-like state, from which a proportion of cells escape by polyploidization. The resulting ASC line is susceptible to adipogenic, osteogenic and chondrogenic differentiation, and expresses the stem cell markers CD29 and Sca-1 on an upregulated level. Differential transcriptome analysis of ASCs with normal and polyploid karyotype shows altered expression of genes that are involved in regulation of cancer, cellular growth and proliferation. We verified the increased expression of Klf4 and loss of Nestin on protein level. We found that elevated production of insulin-like growth factor 1 by polyploid ASCs rendered them more potent in tumor growth promotion in vitro. Conclusions Our model indicates how ASCs with altered genetic background may support tumor progression. Electronic supplementary material The online version of this article (10.1186/s12885-018-4781-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Roberta Fajka-Boja
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Genetics, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Annamária Marton
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Biochemistry, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Anna Tóth
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Genetics, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Péter Blazsó
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Genetics, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Vilmos Tubak
- Creative Laboratory Ltd, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Balázs Bálint
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Biochemistry, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - István Nagy
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Biochemistry, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Zoltán Hegedűs
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Biophysics, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Csaba Vizler
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Biochemistry, H-6726 Temesvári krt. 62, Szeged, Hungary
| | - Robert L Katona
- Biological Research Centre of the Hungarian Academy of Sciences, Institute of Genetics, H-6726 Temesvári krt. 62, Szeged, Hungary.
| |
Collapse
|
37
|
Abstract
Early preclinical and population data suggested a role for the type I insulin-like growth factor receptor (IGF1R) in the regulation of breast cancer growth and survival. To target this pathway, multiple monoclonal antibodies and tyrosine kinase inhibitors were developed and tested in clinical trials. While some of the early clinical trials suggested a benefit for these drugs, none of the attempts showed improved outcomes when compared to conventional therapy. This failure of the IGF1R inhibitors was pronounced in breast cancer; multiple trials testing IGF1R inhibition in estrogen receptor-positive breast cancer were conducted, none showed benefit. This review will evaluate the rationale for IGF1R inhibition, discuss results of the clinical trials and suggest a path forward.
Collapse
Affiliation(s)
- Douglas Yee
- Masonic Cancer CenterUniversity of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
38
|
Mancarella C, Scotlandi K. IGF system in sarcomas: a crucial pathway with many unknowns to exploit for therapy. J Mol Endocrinol 2018; 61:T45-T60. [PMID: 29273680 DOI: 10.1530/jme-17-0250] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022]
Abstract
The insulin-like growth factor (IGF) system has gained substantial interest due to its involvement in regulating cell proliferation, differentiation and survival during anoikis and after conventional and targeted therapies. However, results from clinical trials have been largely disappointing, with only a few but notable exceptions, such as trials targeting sarcomas, especially Ewing sarcoma. This review highlights key studies focusing on IGF signaling in sarcomas, specifically studies underscoring the properties that make this system an attractive therapeutic target and identifies new relationships that may be exploited. This review discusses the potential roles of IGF2 mRNA-binding proteins (IGF2BPs), discoidin domain receptors (DDRs) and metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) in regulating the IGF system. Deeper investigation of these novel regulators of the IGF system may help us to further elucidate the spatial and temporal control of the IGF axis, as understanding the control of this axis is essential for future clinical studies.
Collapse
Affiliation(s)
- Caterina Mancarella
- Experimental Oncology Lab, CRS Development of Biomolecular Therapies, Orthopaedic Rizzoli Institute, Bologna, Italy
| | - Katia Scotlandi
- Experimental Oncology Lab, CRS Development of Biomolecular Therapies, Orthopaedic Rizzoli Institute, Bologna, Italy
| |
Collapse
|
39
|
Aleksic T, Gray N, Wu X, Rieunier G, Osher E, Mills J, Verrill C, Bryant RJ, Han C, Hutchinson K, Lambert AG, Kumar R, Hamdy FC, Weyer-Czernilofsky U, Sanderson MP, Bogenrieder T, Taylor S, Macaulay VM. Nuclear IGF1R Interacts with Regulatory Regions of Chromatin to Promote RNA Polymerase II Recruitment and Gene Expression Associated with Advanced Tumor Stage. Cancer Res 2018; 78:3497-3509. [PMID: 29735545 PMCID: PMC6031306 DOI: 10.1158/0008-5472.can-17-3498] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/28/2018] [Accepted: 04/26/2018] [Indexed: 01/02/2023]
Abstract
Internalization of ligand-activated type I IGF receptor (IGF1R) is followed by recycling to the plasma membrane, degradation or nuclear translocation. Nuclear IGF1R reportedly associates with clinical response to IGF1R inhibitory drugs, yet its role in the nucleus is poorly characterized. Here, we investigated the significance of nuclear IGF1R in clinical cancers and cell line models. In prostate cancers, IGF1R was predominantly membrane localized in benign glands, while malignant epithelium contained prominent internalized (nuclear/cytoplasmic) IGF1R, and nuclear IGF1R associated significantly with advanced tumor stage. Using ChIP-seq to assess global chromatin occupancy, we identified IGF1R-binding sites at or near transcription start sites of genes including JUN and FAM21, most sites coinciding with occupancy by RNA polymerase II (RNAPol2) and histone marks of active enhancers/promoters. IGF1R was inducibly recruited to chromatin, directly binding DNA and interacting with RNAPol2 to upregulate expression of JUN and FAM21, shown to mediate tumor cell survival and IGF-induced migration. IGF1 also enriched RNAPol2 on promoters containing IGF1R-binding sites. These functions were inhibited by IGF1/II-neutralizing antibody xentuzumab (BI 836845), or by blocking receptor internalization. We detected IGF1R on JUN and FAM21 promoters in fresh prostate cancers that contained abundant nuclear IGF1R, with evidence of correlation between nuclear IGF1R content and JUN expression in malignant prostatic epithelium. Taken together, these data reveal previously unrecognized molecular mechanisms through which IGFs promote tumorigenesis, with implications for therapeutic evaluation of anti-IGF drugs.Significance: These findings reveal a noncanonical nuclear role for IGF1R in tumorigenesis, with implications for therapeutic evaluation of IGF inhibitory drugs. Cancer Res; 78(13); 3497-509. ©2018 AACR.
Collapse
Affiliation(s)
- Tamara Aleksic
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nicki Gray
- Computational Biology Research Group, University of Oxford, Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
| | - Xiaoning Wu
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Eliot Osher
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Jack Mills
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Clare Verrill
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Richard J Bryant
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Cheng Han
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| | | | - Adam G Lambert
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rajeev Kumar
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Marchioninistrasse, Munich, Germany
| | - Stephen Taylor
- Computational Biology Research Group, University of Oxford, Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
| | - Valentine M Macaulay
- Department of Oncology, University of Oxford, Oxford, United Kingdom.
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
40
|
Himes BE, Obraztsova K, Lian L, Shumyatcher M, Rue R, Atochina-Vasserman EN, Hur SK, Bartolomei MS, Evans JF, Krymskaya VP. Rapamycin-independent IGF2 expression in Tsc2-null mouse embryo fibroblasts and human lymphangioleiomyomatosis cells. PLoS One 2018; 13:e0197105. [PMID: 29758070 PMCID: PMC5951544 DOI: 10.1371/journal.pone.0197105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/26/2018] [Indexed: 11/19/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, almost exclusively female lung disease linked to inactivating mutations in tuberous sclerosis complex 2 (TSC2), a tumor suppressor gene that controls cell metabolic state and growth via regulation of the mechanistic target of rapamycin (mTORC1) signaling. mTORC1 is frequently activated in human cancers and, although the mTORC1 inhibitor rapamycin has a cytostatic effect, it is, in general, unable to elicit a robust curative effect or tumor regression. Using RNA-Seq, we identified (1) Insulin-like Growth Factor (IGF2) as one of the genes with the highest fold-change difference between human TSC2-null and TSC2-expressing angiomyolipoma cells from a patient with LAM, and (2) the mouse IGF2 homolog Igf2, as a top-ranking gene according to fold change between Tsc2-/- and Tsc2+/+ mouse embryo fibroblasts (MEFs). We extended transcript-level findings to protein level, observing increased Igf2 protein expression and Igf2 secretion by Tsc2-/- MEFs. Increased Igf2 expression was not due to epigenetic imprinting, but was partially mediated through the Stat3 pathway and was completely insensitive to rapamycin treatment. An siRNA-mediated decrease of Igf2 resulted in decreased Stat3 phosphorylation, suggesting presence of an autocrine Igf2/Stat3 amplification cycle in Tsc2-/- MEFs. In human pulmonary LAM lesions and metastatic cell clusters, high levels of IGF2 were associated with mTORC1 activation. In addition, treatment of three primary IGF2-expressing LAM lung cell lines with rapamycin did not result in IGF2 level changes. Thus, targeting of IGF2 signaling may be of therapeutic value to LAM patients, particularly those who are unresponsive to rapamycin.
Collapse
Affiliation(s)
- Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lurong Lian
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ryan Rue
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elena N. Atochina-Vasserman
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Stella K. Hur
- Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marisa S. Bartolomei
- Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jilly F. Evans
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Vera P. Krymskaya
- Penn Center for Pulmonary Biology and Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
41
|
Li T, Wang J, Liu P, Chi J, Yan H, Lei L, Li Z, Yang B, Wang X. Insulin-like growth factor 2 axis supports the serum-independent growth of malignant rhabdoid tumor and is activated by microenvironment stress. Oncotarget 2018; 8:47269-47283. [PMID: 28521298 PMCID: PMC5564563 DOI: 10.18632/oncotarget.17617] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/18/2017] [Indexed: 11/30/2022] Open
Abstract
Malignant rhabdoid tumors (MRTs) are rare, lethal, pediatric tumors predominantly found in the kidney, brain and soft tissues. MRTs are driven by loss of tumor suppressor SNF5/INI1/SMARCB1/BAF47. The prognosis of MRT is poor using currently available treatments, so new treatment targets need to be identified to expand treatment options for patients experiencing chemotherapy resistance. The growth hormone insulin-like growth factor 2 (IGF2) signaling pathway is a promising target to overcome drug resistance in many cancers. Here, we evaluated the role of IGF2 axis in MRT cell proliferation. We showed that microenvironment stress, including starvation treatment and chemotherapy exposure, lead to elevated expression of IGF2 in the SNF5-deficient MRT cell line. The autocrine IGF2, in turn, activated insulin-like growth factor 1 receptor (IGF1R), insulin receptor (INSR), followed by PI3K/AKT pathway and RAS/ERK pathway to promote cancer cell proliferation and survival. We further demonstrated that impairment of IGF2 signaling by IGF2 neutralizing antibody, IGF1R inhibitor NVP-AEW541 or AKT inhibitor MK-2206 2HCl treatment prevented MRT cell growth in vitro. Taken together, our characterization of this axis defines a novel mechanism for MRT cell growth in the microenvironment of stress. Our results also demonstrated the necessity to test the treatment effect targeting this axis in future research.
Collapse
Affiliation(s)
- Ting Li
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Jin Wang
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Pengfei Liu
- Department of Lymphoma, Sino-Us Center of Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jiadong Chi
- Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Han Yan
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Lei Lei
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Zexing Li
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Bing Yang
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Xi Wang
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
42
|
Rayes RF, Milette S, Fernandez MC, Ham B, Wang N, Bourdeau F, Perrino S, Yakar S, Brodt P. Loss of neutrophil polarization in colon carcinoma liver metastases of mice with an inducible, liver-specific IGF-I deficiency. Oncotarget 2018; 9:15691-15704. [PMID: 29644002 PMCID: PMC5884657 DOI: 10.18632/oncotarget.24593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/19/2018] [Indexed: 11/25/2022] Open
Abstract
The growth of cancer metastases in the liver depends on a permissive interaction with the hepatic microenvironment and neutrophils can contribute to this interaction, either positively or negatively, depending on their phenotype. Here we investigated the role of IGF-I in the control of the tumor microenvironment in the liver, using mice with a conditional, liver-specific, IGF-I deficiency (iLID) induced by a single tamoxifen injection. In mice that had a sustained (3 weeks) IGF-I deficiency prior to the intrasplenic/portal inoculation of colon carcinoma MC-38 cells, we observed an increase in neutrophil accumulation in the liver relative to controls. However, unlike controls, these neutrophils did not acquire the (anti-inflammatory) tumor-promoting phenotype, as evidenced by retention of high ICAM-1 expression and nitric oxide production and low CXCR4, CCL5, and VEGF expression and arginase production, all characteristic of the (pro-inflammatory) phenotype. This coincided with an increase in apoptotic tumor cells and reduced metastasis. Neutrophils isolated from these mice also had reduced IGF-IR expression levels. These changes were not observed in iLID mice with a short-term (2 days) IGF-I depletion, despite a 70% reduction in their circulating IGF-I levels, indicating that a sustained IGF-I deficiency was necessary to alter the neutrophil phenotype. Similar results were obtained with the highly metastatic Lewis lung carcinoma subline H-59 cells and in mice injected with an IGF-Trap that blocks IGF-IR signaling by reducing ligand bioavailability. Our results implicate the IGF axis in neutrophil polarization and the induction of a pro-metastatic microenvironment in the liver.
Collapse
Affiliation(s)
- Roni F. Rayes
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - Simon Milette
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - Maria Celia Fernandez
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - Boram Ham
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - Ni Wang
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - France Bourdeau
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - Stephanie Perrino
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| | - Shoshana Yakar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Pnina Brodt
- Departments of Surgery, McGill University and the McGill University Health Centre, Montréal, QC, Canada
- Department of Medicine, McGill University and the McGill University Health Centre, Montréal, QC, Canada
- Department of Oncology, McGill University and the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
43
|
Guo XF, Zhu XF, Cao HY, Zhong GS, Li L, Deng BG, Chen P, Wang PZ, Miao QF, Zhen YS. A bispecific enediyne-energized fusion protein targeting both epidermal growth factor receptor and insulin-like growth factor 1 receptor showing enhanced antitumor efficacy against non-small cell lung cancer. Oncotarget 2018; 8:27286-27299. [PMID: 28460483 PMCID: PMC5432335 DOI: 10.18632/oncotarget.15933] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/20/2017] [Indexed: 12/11/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and insulin-like growth factor 1 receptor (IGF-1R) both overexpressed on non-small cell lung cancer (NSCLC) and are known cooperatively to promote tumor progression and drug resistance. This study was to construct a novel bispecific fusion protein EGF-IGF-LDP-AE consisting of EGFR and IGF-IR specific ligands (EGF and IGF-1) and lidamycin, an enediyne antibiotic with potent antitumor activity, and investigate its antitumor efficacy against NSCLC. Binding and internalization assays showed that EGF-IGF-LDP protein could bind to NSCLC cells with high affinity and then internalized into cells with higher efficiency than that of monospecific proteins. In vitro, the enediyne-energized analogue of bispecific fusion protein (EGF-IGF-LDP-AE) displayed extremely potent cytotoxicity to NSCLC cell lines with IC50<10−11 mol/L. Moreover, the bispecific protein EGF-IGF-LDP-AE was more cytotoxic than monospecific proteins (EGF-LDP-AE and LDP-IGF-AE) and lidamycin. In vivo, EGF-IGF-LDP-AE markedly inhibited the growth of A549 xenografts, and the efficacy was more potent than that of lidamycin and monospecific counterparts. EGF-IGF-LDP-AE caused significant cell cycle arrest and it also induced cell apoptosis in a dosage-dependent manner. Pretreatment with EGF-IGF-LDP-AE inhibited EGF-, IGF-stimulated EGFR and IGF-1R phosphorylation, and blocked two main downstream signaling molecules AKT and ERK activation. These data suggested that EGF-LDP-IGF-AE protein would be a promising targeted agent for NSCLC patients with EGFR and/or IGF-1R overexpression.
Collapse
Affiliation(s)
- Xiao-Fang Guo
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiao-Fei Zhu
- Department of Clinical Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, China
| | - Hai-Ying Cao
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Gen-Shen Zhong
- Laboratory of Cancer Biotherapy, Institute of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Liang Li
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Perking Union Medical College, Beijing, China
| | - Bao-Guo Deng
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ping Chen
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Pei-Zhen Wang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Qing-Fang Miao
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Perking Union Medical College, Beijing, China
| | - Yong-Su Zhen
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Perking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Björner S, Rosendahl AH, Simonsson M, Markkula A, Jirström K, Borgquist S, Rose C, Ingvar C, Jernström H. Combined and individual tumor-specific expression of insulin-like growth factor-I receptor, insulin receptor and phospho-insulin-like growth factor-I receptor/insulin receptor in primary breast cancer: Implications for prognosis in different treatment groups. Oncotarget 2018; 8:9093-9107. [PMID: 28030849 PMCID: PMC5354717 DOI: 10.18632/oncotarget.14082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022] Open
Abstract
Clinical trials examining insulin-like growth factor-I receptor (IGF1R)-targeting strategies have emphasized that better predictive biomarkers are required to improve patient selection. Immunohistochemical tumor-specific protein expression of IGF1R, insulin receptor (InsR), and phosphorylated IGF1R/InsR (pIGF1R/InsR) individually and combined in relation to breast cancer prognosis was evaluated in a population-based cohort of 1,026 primary invasive breast cancer patients without preoperative treatment diagnosed in Sweden. IGF1R (n = 923), InsR (n = 900), and pIGF1R/InsR (n = 904) combined cytoplasmic and membrane staining was dichotomized. IGF1Rstrong/InsRmod/strong/pIGF1R/InsRpos tumors were borderline associated with 2-fold risk for events, HRadj (2.00; 95%CI 0.96-4.18). Combined IGF1R and pIGF1R/InsR status only impacted prognosis in patients with InsRmod/strong expressing tumors (Pinteraction = 0.041). IGF1Rstrong expression impacted endocrine treatment response differently depending on patients’ age and type of endocrine therapy. Phospho-IGF1R/InsRpos was associated with lower risk for events among non-endocrine-treated patients irrespective of ER status, HRadj (0.32; 95%CI 0.16-0.63), but not among endocrine-treated patients (Pinteraction = 0.024). In non-endocrine-treated patients, pIGF1R/InsRpos was associated with lower risk for events after radiotherapy, HRadj (0.31; 95%CI 0.12-0.80), and chemotherapy, HRadj (0.29; 95%CI 0.09-0.99). This study highlights the complexity of IGF hetero-and homodimer signaling network and its interplay with endocrine treatment, suggesting that combinations of involved factors may improve patient selection for IGF1R-targeted therapy.
Collapse
Affiliation(s)
- Sofie Björner
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Maria Simonsson
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Andrea Markkula
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden.,Department of Oncology and Haematology, Skåne University Hospital, Sweden
| | - Carsten Rose
- CREATE Health and Department of Immunotechnology, Lund University, Medicon Village, Lund, Sweden
| | - Christian Ingvar
- Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Surgery, Lund, Sweden
| | - Helena Jernström
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| |
Collapse
|
45
|
Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE. Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer 2018; 17:66. [PMID: 29475434 PMCID: PMC5824531 DOI: 10.1186/s12943-018-0806-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 02/01/2018] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is unrivalled the deadliest gastrointestinal cancer in the western world. There is substantial evidence implying that insulin and insulin-like growth factor (IGF) signaling axis prompt PDAC into an advanced stage by enhancing tumor growth, metastasis and by driving therapy resistance. Numerous efforts have been made to block Insulin/IGF signaling pathway in cancer therapy. However, therapies that target the IGF1 receptor (IGF-1R) and IGF subtypes (IGF-1 and IGF-2) have been repeatedly unsuccessful. This failure may not only be due to the complexity and homology that is shared by Insulin and IGF receptors, but also due to the complex stroma-cancer interactions in the pancreas. Shedding light on the interactions between the endocrine/exocrine pancreas and the stroma in PDAC is likely to steer us toward the development of novel treatments. In this review, we highlight the stroma-derived IGF signaling and IGF-binding proteins as potential novel therapeutic targets in PDAC.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - H Erdinc Besikcioglu
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, München, Germany.,Department of Histology and Embryology, Gazi University Institute of Health Sciences, Ankara, Turkey
| | - Shenghan Wang
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, München, Germany.
| |
Collapse
|
46
|
Ireland L, Santos A, Campbell F, Figueiredo C, Hammond D, Ellies LG, Weyer-Czernilofsky U, Bogenrieder T, Schmid M, Mielgo A. Blockade of insulin-like growth factors increases efficacy of paclitaxel in metastatic breast cancer. Oncogene 2018; 37:2022-2036. [PMID: 29367764 PMCID: PMC5895608 DOI: 10.1038/s41388-017-0115-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 11/15/2017] [Accepted: 12/14/2017] [Indexed: 12/27/2022]
Abstract
Breast cancer remains the leading cause of cancer death in women owing to metastasis and the development of resistance to established therapies. Macrophages are the most abundant immune cells in the breast tumor microenvironment and can both inhibit and support cancer progression. Thus, gaining a better understanding of how macrophages support cancer could lead to the development of more effective therapies. In this study, we find that breast cancer-associated macrophages express high levels of insulin-like growth factors 1 and 2 (IGFs) and are the main source of IGFs within both primary and metastatic tumors. In total, 75% of breast cancer patients show activation of insulin/IGF-1 receptor signaling and this correlates with increased macrophage infiltration and advanced tumor stage. In patients with invasive breast cancer, activation of Insulin/IGF-1 receptors increased to 87%. Blocking IGF in combination with paclitaxel, a chemotherapeutic agent commonly used to treat breast cancer, showed a significant reduction in tumor cell proliferation and lung metastasis in pre-clinical breast cancer models compared to paclitaxel monotherapy. Our findings provide the rationale for further developing the combination of paclitaxel with IGF blockers for the treatment of invasive breast cancer, and Insulin/IGF1R activation and IGF+ stroma cells as potential biomarker candidates for further evaluation.
Collapse
Affiliation(s)
- Lucy Ireland
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Almudena Santos
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Fiona Campbell
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Carlos Figueiredo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Dean Hammond
- Department of Physiology, University of Liverpool, Liverpool, UK
| | - Lesley G Ellies
- Department of Pathology, University of California San Diego, La Jolla, USA
| | | | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co KG Medicine and Translational Research, Vienna, Austria.,Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Michael Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
47
|
Christopoulos PF, Corthay A, Koutsilieris M. Aiming for the Insulin-like Growth Factor-1 system in breast cancer therapeutics. Cancer Treat Rev 2017; 63:79-95. [PMID: 29253837 DOI: 10.1016/j.ctrv.2017.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/23/2022]
Abstract
Despite the major discoveries occurred in oncology the recent years, breast malignancies remain one of the most common causes of cancer-related deaths for women in developed countries. Development of HER2-targeting drugs has been considered a breakthrough in anti-cancer approaches and alluded to the potential of targeting growth factors in breast cancer (BrCa) therapeutics. More than twenty-five years have passed since the Insulin-like Growth Factor-1 (IGF-1) system was initially recognized as a potential target candidate in BrCa therapy. To date, a growing body of studies have implicated the IGF-1 signaling with the BrCa biology. Despite the promising experimental evidence, the impression from clinical trials is rather disappointing. Several reasons may account for this and the last word regarding the efficacy of this system as a target candidate in BrCa therapeutics is probably not written yet. Herein, we provide the theoretical basis, as well as, a comprehensive overview of the current literature, regarding the different strategies targeting the various components of the IGF-1/IGF-1R axis in several pathophysiological aspects of BrCa, including the tumor micro-environment and cancer stemness. In addition, we review the rationale for targeting the IGF-1 system in the different BrCa molecular subtypes and in treatment resistant breast tumors with a focus on both the molecular mechanisms and on the clinical perspectives of such approaches in specific population subgroups. We also discuss the future challenges, as well as, the development of novel molecules and strategies targeting the system and suggest potential improvements in the field.
Collapse
Affiliation(s)
- Panagiotis F Christopoulos
- Department of Experimental Physiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece; Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway; Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway.
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Michael Koutsilieris
- Department of Experimental Physiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
48
|
Simpson A, Petnga W, Macaulay VM, Weyer-Czernilofsky U, Bogenrieder T. Insulin-Like Growth Factor (IGF) Pathway Targeting in Cancer: Role of the IGF Axis and Opportunities for Future Combination Studies. Target Oncol 2017; 12:571-597. [PMID: 28815409 PMCID: PMC5610669 DOI: 10.1007/s11523-017-0514-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite a strong preclinical rationale for targeting the insulin-like growth factor (IGF) axis in cancer, clinical studies of IGF-1 receptor (IGF-1R)-targeted monotherapies have been largely disappointing, and any potential success has been limited by the lack of validated predictive biomarkers for patient enrichment. A large body of preclinical evidence suggests that the key role of the IGF axis in cancer is in driving treatment resistance, via general proliferative/survival mechanisms, interactions with other mitogenic signaling networks, and class-specific mechanisms such as DNA damage repair. Consequently, combining IGF-targeted agents with standard cytotoxic agents, other targeted agents, endocrine therapies, or immunotherapies represents an attractive therapeutic approach. Anti-IGF-1R monoclonal antibodies (mAbs) do not inhibit IGF ligand 2 (IGF-2) activation of the insulin receptor isoform-A (INSR-A), which may limit their anti-proliferative activity. In addition, due to their lack of specificity, IGF-1R tyrosine kinase inhibitors are associated with hyperglycemia as a result of interference with signaling through the classical metabolic INSR-B isoform; this may preclude their use at clinically effective doses. Conversely, IGF-1/IGF-2 ligand-neutralizing mAbs inhibit proliferative/anti-apoptotic signaling via IGF-1R and INSR-A, without compromising the metabolic function of INSR-B. Therefore, combination regimens that include these agents may be more efficacious and tolerable versus IGF-1R-targeted combinations. Herein, we review the preclinical and clinical experience with IGF-targeted therapies to-date, and discuss the rationale for future combination approaches as a means to overcome treatment resistance.
Collapse
Affiliation(s)
- Aaron Simpson
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | - Thomas Bogenrieder
- Boehringer Ingelheim RCV, Dr. Boehringer Gasse 5-11, 1121, Vienna, Austria.
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
49
|
Liefers-Visser JAL, Meijering RAM, Reyners AKL, van der Zee AGJ, de Jong S. IGF system targeted therapy: Therapeutic opportunities for ovarian cancer. Cancer Treat Rev 2017; 60:90-99. [PMID: 28934637 DOI: 10.1016/j.ctrv.2017.08.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
The insulin-like growth factor (IGF) system comprises multiple growth factor receptors, including insulin-like growth factor 1 receptor (IGF-1R), insulin receptor (IR) -A and -B. These receptors are activated upon binding to their respective growth factor ligands, IGF-I, IGF-II and insulin, and play an important role in development, maintenance, progression, survival and chemotherapeutic response of ovarian cancer. In many pre-clinical studies anti-IGF-1R/IR targeted strategies proved effective in reducing growth of ovarian cancer models. In addition, anti-IGF-1R targeted strategies potentiated the efficacy of platinum based chemotherapy. Despite the vast amount of encouraging and promising pre-clinical data, anti-IGF-1R/IR targeted strategies lacked efficacy in the clinic. The question is whether targeting the IGF-1R/IR signaling pathway still holds therapeutic potential. In this review we address the complexity of the IGF-1R/IR signaling pathway, including receptor heterodimerization within and outside the IGF system and downstream signaling. Further, we discuss the implications of this complexity on current targeted strategies and indicate therapeutic opportunities for successful targeting of the IGF-1R/IR signaling pathway in ovarian cancer. Multiple-targeted approaches circumventing bidirectional receptor tyrosine kinase (RTK) compensation and prevention of system rewiring are expected to have more therapeutic potential.
Collapse
Affiliation(s)
- J A L Liefers-Visser
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - R A M Meijering
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A K L Reyners
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A G J van der Zee
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - S de Jong
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
50
|
Sanderson MP, Hofmann MH, Garin-Chesa P, Schweifer N, Wernitznig A, Fischer S, Jeschko A, Meyer R, Moll J, Pecina T, Arnhof H, Weyer-Czernilofsky U, Zahn SK, Adolf GR, Kraut N. The IGF1R/INSR Inhibitor BI 885578 Selectively Inhibits Growth of IGF2-Overexpressing Colorectal Cancer Tumors and Potentiates the Efficacy of Anti-VEGF Therapy. Mol Cancer Ther 2017; 16:2223-2233. [PMID: 28729397 DOI: 10.1158/1535-7163.mct-17-0336] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/16/2017] [Accepted: 06/30/2017] [Indexed: 12/21/2022]
Abstract
Clinical studies of pharmacologic agents targeting the insulin-like growth factor (IGF) pathway in unselected cancer patients have so far demonstrated modest efficacy outcomes, with objective responses being rare. As such, the identification of selection biomarkers for enrichment of potential responders represents a high priority for future trials of these agents. Several reports have described high IGF2 expression in a subset of colorectal cancers, with focal IGF2 amplification being responsible for some of these cases. We defined a novel cut-off value for IGF2 overexpression based on differential expression between colorectal tumors and normal tissue samples. Analysis of two independent colorectal cancer datasets revealed IGF2 to be overexpressed at a frequency of 13% to 22%. An in vitro screen of 34 colorectal cancer cell lines revealed IGF2 expression to significantly correlate with sensitivity to the IGF1R/INSR inhibitor BI 885578. Furthermore, autocrine IGF2 constitutively activated IGF1R and Akt phosphorylation, which was inhibited by BI 885578 treatment. BI 885578 significantly delayed the growth of IGF2-high colorectal cancer xenograft tumors in mice, while combination with a VEGF-A antibody increased efficacy and induced tumor regression. Besides colorectal cancer, IGF2 overexpression was detected in more than 10% of bladder carcinoma, hepatocellular carcinoma and non-small cell lung cancer patient samples. Meanwhile, IGF2-high non-colorectal cancer cells lines displayed constitutive IGF1R phosphorylation and were sensitive to BI 885578. Our findings suggest that IGF2 may represent an attractive patient selection biomarker for IGF pathway inhibitors and that combination with VEGF-targeting agents may further improve clinical outcomes. Mol Cancer Ther; 16(10); 2223-33. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Reiner Meyer
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Jürgen Moll
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Thomas Pecina
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | | | | | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|