1
|
Tao Y, Wang Q, Guo S, Liu J, Cao Y. m6A related metabolic genes in breast cancer and their relationship with prognosis. Int Immunopharmacol 2025; 148:114121. [PMID: 39889412 DOI: 10.1016/j.intimp.2025.114121] [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: 02/18/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 02/03/2025]
Abstract
Breast cancer (BC) is the most prevalent malignancy among women, with incidence rates rising annually. N6-methyladenosine (m6A) modification has been recognized as a key regulator in the onset and progression of BC. Nevertheless, the role of m6A-associated metabolic genes (mMGs) in BC regulation remains insufficiently understood. In this study, we first analyzed and clustered single-cell transcriptomic (scRNA-seq) data from the peripheral blood of BC patients. Differentially expressed genes (DEGs) across various cell populations were intersected with mMGs to identify differentially expressed mMGs (DEmMGs). The AUCell algorithm was employed to score DEmMGs across cell populations, followed by subgroup clustering of high-scoring cell types. Additionally, DEGs from BC and control transcriptomic (RNA-seq) data in The Cancer Genome Atlas (TCGA) were intersected with DEmMGs. BC subtypes were identified based on the expression levels of overlapping genes, and differences in survival rates and immune microenvironment characteristics were examined across subtypes. A BC risk model was constructed using Lasso-Cox regression, and variations in prognosis, tumor mutational burden, immune cell infiltration, and drug sensitivity were explored. Finally, real-time quantitative PCR (qRT-PCR) and Western blot (WB) were used to validate the identified prognostic genes. NDUFAB1, VDAC1, TYMP, UGDH, ATP6AP1, and ALDH2 showed consistent and significant expression differences between the BC and control groups. This study's identification of key prognostic genes and the development of a risk model offer potential new targets for therapeutic intervention and clinical management of BC.
Collapse
Affiliation(s)
- Yong Tao
- Department of Colorectal Surgery, The First Affiliated Hospital of Ningbo University Ningbo Zhejiang China
| | - Qin Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Ningbo University Ningbo Zhejiang China
| | - Shenchao Guo
- Department of Colorectal Surgery, The First Affiliated Hospital of Ningbo University Ningbo Zhejiang China
| | - Jiang Liu
- Department of Colorectal Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China.
| | - Yuepeng Cao
- Department of Colorectal Surgery, The First Affiliated Hospital of Ningbo University Ningbo Zhejiang China.
| |
Collapse
|
2
|
Ye L, Long C, Xu B, Yao X, Yu J, Luo Y, Xu Y, Jiang Z, Nian Z, Zheng Y, Cai Y, Xue X, Guo G. Multi‑omics identification of a novel signature for serous ovarian carcinoma in the context of 3P medicine and based on twelve programmed cell death patterns: a multi-cohort machine learning study. Mol Med 2025; 31:5. [PMID: 39773329 PMCID: PMC11707953 DOI: 10.1186/s10020-024-01036-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Predictive, preventive, and personalized medicine (PPPM/3PM) is a strategy aimed at improving the prognosis of cancer, and programmed cell death (PCD) is increasingly recognized as a potential target in cancer therapy and prognosis. However, a PCD-based predictive model for serous ovarian carcinoma (SOC) is lacking. In the present study, we aimed to establish a cell death index (CDI)-based model using PCD-related genes. METHODS We included 1254 genes from 12 PCD patterns in our analysis. Differentially expressed genes (DEGs) from the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) were screened. Subsequently, 14 PCD-related genes were included in the PCD-gene-based CDI model. Genomics, single-cell transcriptomes, bulk transcriptomes, spatial transcriptomes, and clinical information from TCGA-OV, GSE26193, GSE63885, and GSE140082 were collected and analyzed to verify the prediction model. RESULTS The CDI was recognized as an independent prognostic risk factor for patients with SOC. Patients with SOC and a high CDI had lower survival rates and poorer prognoses than those with a low CDI. Specific clinical parameters and the CDI were combined to establish a nomogram that accurately assessed patient survival. We used the PCD-genes model to observe differences between high and low CDI groups. The results showed that patients with SOC and a high CDI showed immunosuppression and hardly benefited from immunotherapy; therefore, trametinib_1372 and BMS-754807 may be potential therapeutic agents for these patients. CONCLUSIONS The CDI-based model, which was established using 14 PCD-related genes, accurately predicted the tumor microenvironment, immunotherapy response, and drug sensitivity of patients with SOC. Thus this model may help improve the diagnostic and therapeutic efficacy of PPPM.
Collapse
Affiliation(s)
- Lele Ye
- Department of Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunhao Long
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Binbing Xu
- First Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuyang Yao
- First Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiaye Yu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunhui Luo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Xu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuofeng Jiang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zekai Nian
- Second Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yawen Zheng
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yaoyao Cai
- Department of Obstetrics, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Gangqiang Guo
- Department of Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Hassan MS, Johnson C, Ponna S, Scofield D, Awasthi N, von Holzen U. Inhibition of Insulin-like Growth Factor 1 Receptor/Insulin Receptor Signaling by Small-Molecule Inhibitor BMS-754807 Leads to Improved Survival in Experimental Esophageal Adenocarcinoma. Cancers (Basel) 2024; 16:3175. [PMID: 39335147 PMCID: PMC11430189 DOI: 10.3390/cancers16183175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The insulin-like growth factor-1 (IGF-1) and insulin axes are upregulated in obesity and obesity-associated esophageal adenocarcinoma (EAC). Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a contemporary nanotechnology-based paclitaxel (PT) bound to human albumin, ensuring its solubility in water rather than a toxic solvent. Here, we examined the benefits of inhibiting insulin-like growth factor-1 receptor/insulin receptor (IGF-1/IR) signaling and the enhancement of nab-paclitaxel effects by inclusion of the small-molecule inhibitor BMS-754807 using both in vitro and in vivo models of EAC. Using multiple EAC cell lines, BMS-754807 and nab-paclitaxel were evaluated as mono and combination therapies for in vitro effects on cell proliferation, cell death, and cell movement. We then analyzed the in vivo anticancer potency with survival improvement with BMS-754807 and nab-paclitaxel mono and combination therapies. BMS-754807 monotherapy suppressed in vitro cell proliferation and wound healing while increasing apoptosis. BMS-754807, when combined with nab-paclitaxel, enhanced those effects on the inhibition of cell proliferation, increment in cell apoptosis, and inhibition of wound healing. BMS-754807 with nab-paclitaxel produced substantially greater antitumor effects by increasing in vivo apoptosis, leading to increased mice survival compared to those of BMS-754807 or nab-paclitaxel monotherapy. Our outcomes support the use of BMS-754807, alone and in combination with nab-paclitaxel, as an efficient and innovative treatment choice for EAC.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Chloe Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Saisantosh Ponna
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Dimitri Scofield
- Department of Biology, Indiana University, South Bend, IN 47405, USA
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
- Goshen Center for Cancer Care, Goshen, IN 46526, USA
- School of Medicine, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
4
|
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
|
5
|
Li J, Huang G. Insulin receptor alternative splicing in breast and prostate cancer. Cancer Cell Int 2024; 24:62. [PMID: 38331804 PMCID: PMC10851471 DOI: 10.1186/s12935-024-03252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer etiology represents an intricate, multifactorial orchestration where metabolically associated insulin-like growth factors (IGFs) and insulin foster cellular proliferation and growth throughout tumorigenesis. The insulin receptor (IR) exhibits two splice variants arising from alternative mRNA processing, namely IR-A, and IR-B, with remarkable distribution and biological effects disparities. This insightful review elucidates the structural intricacies, widespread distribution, and functional significance of IR-A and IR-B. Additionally, it explores the regulatory mechanisms governing alternative splicing processes, intricate signal transduction pathways, and the intricate association linking IR-A and IR-B splicing variants to breast and prostate cancer tumorigenesis. Breast cancer and prostate cancer are the most common malignant tumors with the highest incidence rates among women and men, respectively. These findings provide a promising theoretical framework for advancing preventive strategies, diagnostic modalities, and therapeutic interventions targeting breast and prostate cancer.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China
| | - Gena Huang
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China.
| |
Collapse
|
6
|
Malvi P, Chava S, Cai G, Hu K, Zhu LJ, Edwards YJK, Green MR, Gupta R, Wajapeyee N. HOXC6 drives a therapeutically targetable pancreatic cancer growth and metastasis pathway by regulating MSK1 and PPP2R2B. Cell Rep Med 2023; 4:101285. [PMID: 37951219 PMCID: PMC10694669 DOI: 10.1016/j.xcrm.2023.101285] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 11/13/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, which lacks effective therapies. Here, we demonstrate that the transcription factor, homeobox C6 (HOXC6), is overexpressed in most PDACs, and its inhibition blocks PDAC tumor growth and metastasis. HOXC6 transcriptionally activates tumor-promoting kinase MSK1 and suppresses tumor-inhibitory protein PPP2R2B in PDAC. HOXC6-induced PPP2R2B suppression causes mammalian target of rapamycin (mTOR) pathway activation, which facilitates PDAC growth. Also, MSK1 upregulation by HOXC6 is necessary for PDAC growth because of its ability to suppress apoptosis via its substrate DDX17. Combinatorial pharmacological inhibition of MSK1 and mTOR potently suppressed PDAC tumor growth and metastasis in PDAC mouse models. PDAC cells with acquired resistance to MSK1/mTOR-inhibitors displayed activated insulin-like growth factor 1 receptor (IGF1R) signaling and were successfully eradicated by IGF1R inhibitor. Furthermore, MEK inhibitor trametinib enhanced the efficacy of dual MSK1 and mTOR inhibition. Collectively, these results identify therapeutic vulnerabilities of PDAC and an approach to overcome acquired drug resistance to prolong therapeutic benefit.
Collapse
Affiliation(s)
- Parmanand Malvi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Guoping Cai
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kai Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yvonne J K Edwards
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Michael R Green
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
7
|
Chen J, Sun M, Chen C, Kang M, Qian B, Sun J, Ma X, Zhou J, Huang L, Jiang B, Fang Y. Construction of a novel anoikis-related prognostic model and analysis of its correlation with infiltration of immune cells in neuroblastoma. Front Immunol 2023; 14:1135617. [PMID: 37081871 PMCID: PMC10111050 DOI: 10.3389/fimmu.2023.1135617] [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: 01/01/2023] [Accepted: 03/23/2023] [Indexed: 04/22/2023] Open
Abstract
Background Anoikis resistance (AR) plays an important role in the process of metastasis, which is an important factor affecting the risk stage of neuroblastoma (NB). This study aims to construct an anoikis-related prognostic model and analyze the characteristics of hub genes, important pathways and tumor microenvironment of anoikis-related subtypes of NB, so as to provide help for the clinical diagnosis, treatment and research of NB. Methods We combined transcriptome data of GSE49710 and E-MTAB-8248, screened anoikis-related genes (Args) closely related to the prognosis of NB by univariate cox regression analysis, and divided the samples into anoikis-related subtypes by consistent cluster analysis. WGCNA was used to screen hub genes, GSVA and GSEA were used to analyze the differentially enriched pathways between anoikis-related subtypes. We analyzed the infiltration levels of immune cells between different groups by SsGSEA and CIBERSORT. Lasso and multivariate regression analyses were used to construct a prognostic model. Finally, we analyzed drug sensitivity through the GDSC database. Results 721 cases and 283 Args were included in this study. All samples were grouped into two subtypes with different prognoses. The analyses of WGCNA, GSVA and GSEA suggested the existence of differentially expressed hub genes and important pathways in the two subtypes. We further constructed an anoikis-related prognostic model, in which 15 Args participated. This model had more advantages in evaluating the prognoses of NB than other commonly used clinical indicators. The infiltration levels of 9 immune cells were significantly different between different risk groups, and 13 Args involved in the model construction were correlated with the infiltration levels of immune cells. There was a relationship between the infiltration levels of 6 immune cells and riskscores. Finally, we screened 15 drugs with more obvious effects on NB in high-risk group. Conclusion There are two anoikis-related subtypes with different prognoses in the population of NB. The anoikis-related prognostic model constructed in this study can accurately predict the prognoses of children with NB, and has a good guiding significance for clinical diagnosis, treatment and research of NB.
Collapse
Affiliation(s)
- Ji Chen
- Department of General Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Mengjiao Sun
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Chuqin Chen
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Meiyun Kang
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Qian
- Department of Cardiothoracic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Sun
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaopeng Ma
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jianfeng Zhou
- Department of General Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Huang
- Department of General Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Lei Huang, ; Bin Jiang, ; Yongjun Fang,
| | - Bin Jiang
- Department of General Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Lei Huang, ; Bin Jiang, ; Yongjun Fang,
| | - Yongjun Fang
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Lei Huang, ; Bin Jiang, ; Yongjun Fang,
| |
Collapse
|
8
|
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
|
9
|
Zhang X, Zhu X, Bi X, Huang J, Zhou L. The Insulin Receptor: An Important Target for the Development of Novel Medicines and Pesticides. Int J Mol Sci 2022; 23:7793. [PMID: 35887136 PMCID: PMC9325136 DOI: 10.3390/ijms23147793] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
The insulin receptor (IR) is a transmembrane protein that is activated by ligands in insulin signaling pathways. The IR has been considered as a novel therapeutic target for clinical intervention, considering the overexpression of its protein and A-isoform in multiple cancers, Alzheimer's disease, and Type 2 diabetes mellitus in humans. Meanwhile, it may also serve as a potential target in pest management due to its multiple physiological influences in insects. In this review, we provide an overview of the structural and molecular biology of the IR, functions of IRs in humans and insects, physiological and nonpeptide small molecule modulators of the IR, and the regulating mechanisms of the IR. Xenobiotic compounds and the corresponding insecticidal chemicals functioning on the IR are also discussed. This review is expected to provide useful information for a better understanding of human IR-related diseases, as well as to facilitate the development of novel small-molecule activators and inhibitors of the IR for use as medicines or pesticides.
Collapse
Affiliation(s)
| | | | | | - Jiguang Huang
- Key Laboratory of Natural Pesticides & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.Z.); (X.B.)
| | - Lijuan Zhou
- Key Laboratory of Natural Pesticides & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.Z.); (X.B.)
| |
Collapse
|
10
|
Bulatowicz JJ, Wood TL. Activation Versus Inhibition of IGF1R: A Dual Role in Breast Tumorigenesis. Front Endocrinol (Lausanne) 2022; 13:911079. [PMID: 35784559 PMCID: PMC9247239 DOI: 10.3389/fendo.2022.911079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Historically, the body of literature surrounding the insulin-like growth factor type 1 receptor (IGF1R) has described a largely pro-tumorigenic role in breast cancer cells and in several transgenic or xenograft mouse models of breast cancer. Interestingly, however, more recent evidence has emerged that suggests an additional, previously undescribed, tumor and metastasis suppressive function for IGF1R in both human breast tumors and mammary oncogenesis in mice. These seemingly conflicting reports can be reconciled when considering what is currently known about IGF1R function in the context of tissue development and cancer as it relates to cellular growth, proliferation, and differentiation. In this mini review, we will summarize the currently existing data with a particular focus on mouse models that have been developed to study IGF1R function in mammary development, tumorigenesis, and metastasis in vivo and propose hypotheses for how both the tumor-promoting and tumor-suppressing schools of thought regarding IGF1R in these histological contexts are compatible.
Collapse
Affiliation(s)
| | - Teresa L. Wood
- Department of Pharmacology, Physiology, & Neuroscience, Center for Cell Signaling and Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| |
Collapse
|
11
|
Identification of Molecular Subtypes and Potential Small-Molecule Drugs for Esophagus Cancer Treatment Based on m 6A Regulators. JOURNAL OF ONCOLOGY 2022; 2022:5490461. [PMID: 35069736 PMCID: PMC8776445 DOI: 10.1155/2022/5490461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Esophagus cancer (ESCA) is the sixth most frequent cancer in males, with 5-year overall survival of 15%-25%. RNA modifications function critically in cancer progression, and m6A regulators are associated with ESCA prognosis. This study further revealed correlations between m6A and ESCA development. METHODS Univariate Cox regression analysis and consensus clustering were applied to determine molecular subtypes. Functional pathways and gene ontology terms were enriched by gene set enrichment analysis. Protein-protein interaction (PPI) analysis on differentially expressed genes (DEGs) was conducted for hub gene screening. Public drug databases were employed to study the interactions between hub genes and small molecules. RESULTS Three molecular subtypes related to ESCA prognosis were determined. Based on multiple analyses among molecular subtypes, 146 DEGs were screened, and a PPT network of 15 hub genes was visualized. Finally, 8 potential small-molecule drugs (BMS-754807, gefitinib, neratinib, zuclopenthixol, puromycin, sulfasalazine, and imatinib) were identified for treating ESCA. CONCLUSIONS This study applied a new approach to analyzing the relation between m6A and ESCA prognosis, providing a reference for exploring potential targets and drugs for ESCA treatment.
Collapse
|
12
|
Mansour MA, Caputo VS, Aleem E. Highlights on selected growth factors and their receptors as promising anticancer drug targets. Int J Biochem Cell Biol 2021; 140:106087. [PMID: 34563698 DOI: 10.1016/j.biocel.2021.106087] [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: 04/15/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Growth factor receptors (GFRs) and receptor tyrosine kinases (RTK) are groups of proteins mediating a plethora of physiological processes, including cell growth, proliferation, survival, differentiation and migration. Under certain circumstances, expression of GFRs and subsequently their downstream kinase signaling are deregulated by genetic, epigenetic, and somatic changes leading to uncontrolled cell division in many human diseases, most notably cancer. Cancer cells rely on growth factors to sustain the increasing need to cell division and metabolic reprogramming through cancer-associated activating mutations of their receptors (i.e., GFRs). In this review, we highlight the recent advances of selected GFRs and their ligands (growth factors) in cancer with emphasis on structural and functional differences. We also interrogate how overexpression and/or hyperactivation of GFRs contribute to cancer initiation, development, progression, and resistance to conventional chemo- and radiotherapies. Novel approaches are being developed as anticancer agents to target growth factor receptors and their signaling pathways in different cancers. Here, we illustrate how the current knowledge of GFRs biology, and their ligands lead to development of targeted therapies to inhibit and/or block the activity of growth factors, GFRs and downstream kinases to treat diseases such as cancer.
Collapse
Affiliation(s)
- Mohammed A Mansour
- Cancer Biology and Therapy Lab, Division of Human Sciences, School of Applied Sciences, London South Bank University, London, UK; Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Valentina S Caputo
- Cancer Biology and Therapy Lab, Division of Human Sciences, School of Applied Sciences, London South Bank University, London, UK
| | - Eiman Aleem
- Cancer Biology and Therapy Lab, Division of Human Sciences, School of Applied Sciences, London South Bank University, London, UK.
| |
Collapse
|
13
|
Lero MW, Shaw LM. Diversity of insulin and IGF signaling in breast cancer: Implications for therapy. Mol Cell Endocrinol 2021; 527:111213. [PMID: 33607269 PMCID: PMC8035314 DOI: 10.1016/j.mce.2021.111213] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
This review highlights the significance of the insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF-1R) signaling pathway in cancer and assesses its potential as a therapeutic target. Our emphasis is on breast cancer, but this pathway is central to the behavior of many cancers. An understanding of how IR/IGF-1R signaling contributes to the function of the normal mammary gland provides a foundation for understanding its aberrations in breast cancer. Specifically, dysregulation of the expression and function of ligands (insulin, IGF-1 and IGF-2), receptors and their downstream signaling effectors drive breast cancer initiation and progression, often in a subtype-dependent manner. Efforts to target this pathway for the treatment of cancer have been hindered by several factors including a lack of biomarkers to select patients that could respond to targeted therapy and adverse effects on normal metabolism. To this end, we discuss ongoing efforts aimed at overcoming such obstacles.
Collapse
Affiliation(s)
- Michael W Lero
- Department of Molecular, Cell & Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Leslie M Shaw
- Department of Molecular, Cell & Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
14
|
Ghafouri-Fard S, Abak A, Mohaqiq M, Shoorei H, Taheri M. The Interplay Between Non-coding RNAs and Insulin-Like Growth Factor Signaling in the Pathogenesis of Neoplasia. Front Cell Dev Biol 2021; 9:634512. [PMID: 33768092 PMCID: PMC7985092 DOI: 10.3389/fcell.2021.634512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
The insulin-like growth factors (IGFs) are polypeptides with similar sequences with insulin. These factors regulate cell growth, development, maturation, and aging via different processes including the interplay with MAPK, Akt, and PI3K. IGF signaling participates in the pathogenesis of neoplasia, insulin resistance, diabetes mellitus, polycystic ovarian syndrome, cerebral ischemic injury, fatty liver disease, and several other conditions. Recent investigations have demonstrated the interplay between non-coding RNAs and IGF signaling. This interplay has fundamental roles in the development of the mentioned disorders. We designed the current study to search the available data about the role of IGF-associated non-coding RNAs in the evolution of neoplasia and other conditions. As novel therapeutic strategies have been designed for modification of IGF signaling, identification of the impact of non-coding RNAs in this pathway is necessary for the prediction of response to these modalities.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mohaqiq
- School of Advancement, Centennial College, Ashtonbee Campus, Toronto, ON, Canada
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Biranjd University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Fuentes-Baile M, Ventero MP, Encinar JA, García-Morales P, Poveda-Deltell M, Pérez-Valenciano E, Barberá VM, Gallego-Plazas J, Rodríguez-Lescure Á, Martín-Nieto J, Saceda M. Differential Effects of IGF-1R Small Molecule Tyrosine Kinase Inhibitors BMS-754807 and OSI-906 on Human Cancer Cell Lines. Cancers (Basel) 2020; 12:cancers12123717. [PMID: 33322337 PMCID: PMC7763458 DOI: 10.3390/cancers12123717] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 01/30/2023] Open
Abstract
Simple Summary We have tested the effects of IGF-1R tyrosine kinase inhibitors BMS-754807 (BMS) and OSI-906 (OSI) on human colon, pancreatic carcinoma cell, and glioblastoma cell lines and primary cultures. Although OSI and BMS are able to inhibit IGF-1R activity at low doses, the differential effect on cell proliferation and cell-cycle phase distribution shown by both compounds probes that many effects observed are mediated by BMS off-target interactions. Using MAPKs ELISAs and phospho-RTK array analysis, we have identified several BMS regulated putative kinases able to mediate BMS off-target effects. Interestingly, molecular docking assays suggest that BMS could affect these kinases not only by blocking their ATP-binding domain, but also by means of allosteric interactions. Since BMS has an important antineoplastic effect on these poor prognosis types of cancer, these compounds could be taken in consideration for treatment independently of IGF-1R status. Abstract We have determined the effects of the IGF-1R tyrosine kinase inhibitors BMS-754807 (BMS) and OSI-906 (OSI) on cell proliferation and cell-cycle phase distribution in human colon, pancreatic carcinoma, and glioblastoma cell lines and primary cultures. IGF-1R signaling was blocked by BMS and OSI at equivalent doses, although both inhibitors exhibited differential antiproliferative effects. In all pancreatic carcinoma cell lines tested, BMS exerted a strong antiproliferative effect, whereas OSI had a minimal effect. Similar results were obtained on glioblastoma primary cultures, where HGUE-GB-15, -16 and -17 displayed resistance to OSI effects, whereas they were inhibited in their proliferation by BMS. Differential effects of BMS and OSI were also observed in colon carcinoma cell lines. Both inhibitors also showed different effects on cell cycle phase distribution, BMS induced G2/M arrest followed by cell death, while OSI induced G1 arrest with no cell death. Both inhibitors also showed different effects on other protein kinases activities. Taken together, our results are indicative that BMS mainly acts through off-target effects exerted on other protein kinases. Given that BMS exhibits a potent antiproliferative effect, we believe that this compound could be useful for the treatment of different types of tumors independently of their IGF-1R activation status.
Collapse
Affiliation(s)
- María Fuentes-Baile
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (M.F.-B.); (V.M.B.)
| | - María P. Ventero
- Unidad de Investigación, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario de Alicante, 03005 Alicante, Spain;
| | - José A. Encinar
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
- Correspondence: (J.A.E.); (M.S.); Tel.: +34-966658432 (M.S.)
| | - Pilar García-Morales
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
| | - María Poveda-Deltell
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
| | - Elizabeth Pérez-Valenciano
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
| | - Víctor M. Barberá
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (M.F.-B.); (V.M.B.)
- Unidad de Genética Molecular, Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain
| | - Javier Gallego-Plazas
- Servicio de Oncología, Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (J.G.-P.); (Á.R.-L.)
| | - Álvaro Rodríguez-Lescure
- Servicio de Oncología, Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (J.G.-P.); (Á.R.-L.)
| | - José Martín-Nieto
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, 03080 Alicante, Spain;
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (M.F.-B.); (V.M.B.)
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
- Correspondence: (J.A.E.); (M.S.); Tel.: +34-966658432 (M.S.)
| |
Collapse
|
16
|
Crudden C, Song D, Cismas S, Trocmé E, Pasca S, Calin GA, Girnita A, Girnita L. Below the Surface: IGF-1R Therapeutic Targeting and Its Endocytic Journey. Cells 2019; 8:cells8101223. [PMID: 31600876 PMCID: PMC6829878 DOI: 10.3390/cells8101223] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/15/2022] Open
Abstract
Ligand-activated plasma membrane receptors follow pathways of endocytosis through the endosomal sorting apparatus. Receptors cluster in clathrin-coated pits that bud inwards and enter the cell as clathrin-coated vesicles. These vesicles travel through the acidic endosome whereby receptors and ligands are sorted to be either recycled or degraded. The traditional paradigm postulated that the endocytosis role lay in signal termination through the removal of the receptor from the cell surface. It is now becoming clear that the internalization process governs more than receptor signal cessation and instead reigns over the entire spatial and temporal wiring of receptor signaling. Governing the localization, the post-translational modifications, and the scaffolding of receptors and downstream signal components established the endosomal platform as the master regulator of receptor function. Confinement of components within or between distinct organelles means that the endosome instructs the cell on how to interpret and translate the signal emanating from any given receptor complex into biological effects. This review explores this emerging paradigm with respect to the cancer-relevant insulin-like growth factor type 1 receptor (IGF-1R) and discusses how this perspective could inform future targeting strategies.
Collapse
Affiliation(s)
- Caitrin Crudden
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands.
| | - Dawei Song
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| | - Sonia Cismas
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| | - Eric Trocmé
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
- St. Erik Eye Hospital, 11282 Stockholm, Sweden.
| | - Sylvya Pasca
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Ada Girnita
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
- Dermatology Department, Karolinska University Hospital, 17176 Stockholm, Sweden.
| | - Leonard Girnita
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| |
Collapse
|
17
|
Lehman CE, Khalil AA, Axelrod MJ, Dougherty MI, Schoeff SS, Taniguchi LE, Mendez RE, David AP, McGarey PO, Hubbard MA, Donaldson L, Frierson HF, Stelow EB, Bekiranov S, Wulfkuhle JD, Petricoin EF, Gioeli DG, Jameson MJ. Antitumor effect of insulin-like growth factor-1 receptor inhibition in head and neck squamous cell carcinoma. Laryngoscope 2019; 130:1470-1478. [PMID: 31433065 DOI: 10.1002/lary.28236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 11/10/2022]
Abstract
OBJECTIVES The insulin-like growth factor-1 receptor (IGF1R) has been implicated in therapeutic resistance in head and neck squamous cell carcinoma (HNSCC), and small molecule tyrosine kinase inhibitors (TKIs) of IGF1R activity may have anticancer activity. Therefore, the relationship between survival and IGF1R expression was assessed for oral cavity (OC) cancer, and the antitumor effects of two IGF1R-TKIs, OSI-906 and BMS-754807, were evaluated in HNSCC cell lines in vitro. METHODS Clinical outcome data and tissue microarray immunohistochemistry were used to generate IGF1R expression-specific survival curves. Immunoblot, alamarBlue proliferation assay, trypan blue exclusion viability test, clonogenic assay, flow cytometry, and reverse phase protein array (RPPA) were used to evaluate in vitro responses to IGF1R-TKIs. RESULTS For patients with stage III/IV OCSCC, higher IGF1R expression was associated with poorer overall 5-year survival (P = 0.029). Both BMS-754807 and OSI-906 caused dose-dependent inhibition of IGF1R and Akt phosphorylation and inhibited proliferation; BMS-754807 was more potent than OSI-906. Both drugs reduced HNSCC cell viability; only OSI-906 was able to eliminate all viable cells at 10 μM. The two drugs similarly inhibited clonogenic cell survival. At 1 μM, only BMS-754807 caused a fourfold increase in the basal apoptotic rate. RPPA demonstrated broad effects of both drugs on canonical IGF1R signaling pathways and also inhibition of human epidermal growth factor receptor-3 (HER3), Src, paxillin, and ezrin phosphorylation. CONCLUSION OSI-906 and BMS-754807 inhibit IGF1R activity in HNSCC cell lines with reduction in prosurvival and proliferative signaling and with concomitant antiproliferative and proapoptotic effects. Such antagonists may have utility as adjuvants to existing therapies for HNSCC. LEVEL OF EVIDENCE NA Laryngoscope, 130:1470-1478, 2020.
Collapse
Affiliation(s)
- Christine E Lehman
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Ashraf A Khalil
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A.,Department of Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufiya University, Shebin El Kom, Egypt
| | - Mark J Axelrod
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A.,Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, U.S.A
| | - Michael I Dougherty
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Stephen S Schoeff
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Linnea E Taniguchi
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Rolando E Mendez
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Abel P David
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Patrick O McGarey
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Matthew A Hubbard
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Lane Donaldson
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Henry F Frierson
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Edward B Stelow
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Stefan Bekiranov
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, U.S.A
| | - Julia D Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, U.S.A
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, U.S.A
| | - Daniel G Gioeli
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, U.S.A
| | - Mark J Jameson
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| |
Collapse
|
18
|
Vella V, Malaguarnera R, Nicolosi ML, Morrione A, Belfiore A. Insulin/IGF signaling and discoidin domain receptors: An emerging functional connection. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118522. [PMID: 31394114 DOI: 10.1016/j.bbamcr.2019.118522] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022]
Abstract
The insulin/insulin-like growth factor system (IIGFs) plays a fundamental role in the regulation of prenatal and postnatal growth, metabolism and homeostasis. As a consequence, dysregulation of this axis is associated with growth disturbance, type 2 diabetes, chronic inflammation and tumor progression. A functional crosstalk between IIGFs and discoidin domain receptors (DDRs) has been recently discovered. DDRs are non-integrin collagen receptors that canonically undergo slow and long-lasting autophosphorylation after binding to fibrillar collagen. While both DDR1 and DDR2 functionally interact with IIGFs, the crosstalk with DDR1 is so far better characterized. Notably, the IIGFs-DDR1 crosstalk presents a feed-forward mechanism, which does not require collagen binding, thus identifying novel non-canonical action of DDR1. Further studies are needed to fully explore the role of this IIGFs-DDRs functional loop as potential target in the treatment of inflammatory and neoplastic disorders.
Collapse
Affiliation(s)
- Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | | | - Maria Luisa Nicolosi
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Andrea Morrione
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy.
| |
Collapse
|
19
|
Crosby P, Hamnett R, Putker M, Hoyle NP, Reed M, Karam CJ, Maywood ES, Stangherlin A, Chesham JE, Hayter EA, Rosenbrier-Ribeiro L, Newham P, Clevers H, Bechtold DA, O'Neill JS. Insulin/IGF-1 Drives PERIOD Synthesis to Entrain Circadian Rhythms with Feeding Time. Cell 2019; 177:896-909.e20. [PMID: 31030999 PMCID: PMC6506277 DOI: 10.1016/j.cell.2019.02.017] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 10/26/2018] [Accepted: 02/11/2019] [Indexed: 01/21/2023]
Abstract
In mammals, endogenous circadian clocks sense and respond to daily feeding and lighting cues, adjusting internal ∼24 h rhythms to resonate with, and anticipate, external cycles of day and night. The mechanism underlying circadian entrainment to feeding time is critical for understanding why mistimed feeding, as occurs during shift work, disrupts circadian physiology, a state that is associated with increased incidence of chronic diseases such as type 2 (T2) diabetes. We show that feeding-regulated hormones insulin and insulin-like growth factor 1 (IGF-1) reset circadian clocks in vivo and in vitro by induction of PERIOD proteins, and mistimed insulin signaling disrupts circadian organization of mouse behavior and clock gene expression. Insulin and IGF-1 receptor signaling is sufficient to determine essential circadian parameters, principally via increased PERIOD protein synthesis. This requires coincident mechanistic target of rapamycin (mTOR) activation, increased phosphoinositide signaling, and microRNA downregulation. Besides its well-known homeostatic functions, we propose insulin and IGF-1 are primary signals of feeding time to cellular clocks throughout the body. Insulin and IGF-1 are a systemic synchronizing cue for circadian rhythms in mammals Insulin and IGF-1 signaling rapidly upregulates translation of PERIOD clock proteins Coincident signaling facilitates selective induction of PERIOD synthesis Circadian disruption is recapitulated by mistimed insulin in cell and animal models
Collapse
Affiliation(s)
- Priya Crosby
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Ryan Hamnett
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Marrit Putker
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Hubrecht Institute, Utrecht 3584 CT, the Netherlands
| | | | - Martin Reed
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | | | | | | | - Edward A Hayter
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | | | - Peter Newham
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, UK
| | - Hans Clevers
- Hubrecht Institute, Utrecht 3584 CT, the Netherlands; Princess Máxima Centre, Utrecht 3584 CS, the Netherlands
| | - David A Bechtold
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - John S O'Neill
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
20
|
Wang M, Hu Y, Yu T, Ma X, Wei X, Wei Y. Pan-HER-targeted approach for cancer therapy: Mechanisms, recent advances and clinical prospect. Cancer Lett 2018; 439:113-130. [PMID: 30218688 DOI: 10.1016/j.canlet.2018.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 02/05/2023]
Abstract
The Human Epidermal Growth Factor Receptor family is composed of 4 structurally related receptor tyrosine kinases that are involved in many human cancers. The efficacy and safety of HER inhibitors have been compared in a wide range of clinical trials, suggesting the superior inhibitory ability of multiple- HER-targeting blockade compared with single receptor antagonists. However, many patients are currently resistant to current therapeutic treatment and novel strategies are warranted to conquer the resistance. Thus, we performed a critical review to summarize the molecular involvement of HER family receptors in tumour progression, recent anti-HER drug development based on clinical trials, and the potential resistance mechanisms of anti-HER therapy.
Collapse
Affiliation(s)
- Manni Wang
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Yuzhu Hu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Ting Yu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xuelei Ma
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xiawei Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China.
| | - Yuquan Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| |
Collapse
|
21
|
Du J, Yu Y, Zhan J, Zhang H. Targeted Therapies Against Growth Factor Signaling in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1026:125-146. [PMID: 29282682 DOI: 10.1007/978-981-10-6020-5_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Breast cancer is the most prevalent female malignancy throughout the world. Conventional treatment strategies for breast cancer consist of chemotherapy, radiation, surgery, chemoradiation, hormone therapy, and targeted therapies. Among them, targeted therapies show advantages to reduce cost and toxicity for being possible for individualized treatments based on the intrinsic subtypes of breast cancer. With deeper understanding of key signaling pathways concerning tumor growth and survival, growth factor-controlled signaling pathways are frequently dysregulated in the development and progression of breast cancer. Thus, targeted therapies against growth factor-mediated signaling pathways have been shown to have promising efficacy in both preclinical animal models and human clinical trials. In this chapter, we will briefly introduce inhibitors and monoclonal antibodies that target the main growth factor-modulated scenarios including epidermal growth factor receptor (EGFR), transforming growth factor beta (TGF-β), insulin-like growth factor 1 receptor (IGF1R), and fibroblast growth factor receptor (FGFR) signaling pathways in breast cancer therapy.
Collapse
Affiliation(s)
- Juan Du
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yu Yu
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jun Zhan
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hongquan Zhang
- Department of Anatomy, Histology and Embryology, Laboratory of Molecular Cell Biology and Tumor Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
22
|
Yohe ME, Gryder BE, Shern JF, Song YK, Chou HC, Sindiri S, Mendoza A, Patidar R, Zhang X, Guha R, Butcher D, Isanogle KA, Robinson CM, Luo X, Chen JQ, Walton A, Awasthi P, Edmondson EF, Difilippantonio S, Wei JS, Zhao K, Ferrer M, Thomas CJ, Khan J. MEK inhibition induces MYOG and remodels super-enhancers in RAS-driven rhabdomyosarcoma. Sci Transl Med 2018; 10:eaan4470. [PMID: 29973406 PMCID: PMC8054766 DOI: 10.1126/scitranslmed.aan4470] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022]
Abstract
The RAS isoforms are frequently mutated in many types of human cancers, including PAX3/PAX7 fusion-negative rhabdomyosarcoma. Pediatric RMS arises from skeletal muscle progenitor cells that have failed to differentiate normally. The role of mutant RAS in this differentiation blockade is incompletely understood. We demonstrate that oncogenic RAS, acting through the RAF-MEK [mitogen-activated protein kinase (MAPK) kinase]-ERK (extracellular signal-regulated kinase) MAPK effector pathway, inhibits myogenic differentiation in rhabdomyosarcoma by repressing the expression of the prodifferentiation myogenic transcription factor, MYOG. This repression is mediated by ERK2-dependent promoter-proximal stalling of RNA polymerase II at the MYOG locus. Small-molecule screening with a library of mechanistically defined inhibitors showed that RAS-driven RMS is vulnerable to MEK inhibition. MEK inhibition with trametinib leads to the loss of ERK2 at the MYOG promoter and releases the transcriptional stalling of MYOG expression. MYOG subsequently opens chromatin and establishes super-enhancers at genes required for late myogenic differentiation. Furthermore, trametinib, in combination with an inhibitor of IGF1R, potently decreases rhabdomyosarcoma cell viability and slows tumor growth in xenograft models. Therefore, this combination represents a potential therapeutic for RAS-mutated rhabdomyosarcoma.
Collapse
Affiliation(s)
- Marielle E Yohe
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Berkley E Gryder
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jack F Shern
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Young K Song
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Hsien-Chao Chou
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sivasish Sindiri
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Rajesh Patidar
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Rajarashi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Kristine A Isanogle
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Christina M Robinson
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ashley Walton
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Elijah F Edmondson
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Jun S Wei
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Keji Zhao
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
23
|
Xu DD, Wang Y, Zhou PJ, Qin SR, Zhang R, Zhang Y, Xue X, Wang J, Wang X, Chen HC, Wang X, Pan YW, Zhang L, Yan HZ, Liu QY, Liu Z, Chen SH, Chen HY, Wang YF. The IGF2/IGF1R/Nanog Signaling Pathway Regulates the Proliferation of Acute Myeloid Leukemia Stem Cells. Front Pharmacol 2018; 9:687. [PMID: 30013477 PMCID: PMC6036281 DOI: 10.3389/fphar.2018.00687] [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: 12/03/2017] [Accepted: 06/07/2018] [Indexed: 12/17/2022] Open
Abstract
Acute myeloid leukemia is an aggressive disease characterized by clonal proliferation and differentiation into immature hematopoietic cells of dysfunctional myeloid precursors. Accumulating evidence shows that CD34+CD38- leukemia stem cells (LSCs) are responsible for drug resistance, metastasis, and relapse of leukemia. In this study, we found that Nanog, a transcription factor in stem cells, is significantly overexpressed in CD34+ populations from patients with acute myeloid leukemia and in LSCs from leukemia cell lines. Our data demonstrate that the knockdown of Nanog inhibited proliferation and induced cell cycle arrest and cell apoptosis. Moreover, Nanog silencing suppressed the leukemogenesis of LSCs in mice. In addition, we found that these functions of Nanog were regulated by the insulin-like growth factor receptor (IGF1R) signaling pathway. Nanog overexpression rescued the colony formation ability of LSCs treated with picropodophyllin (PPP), an IGF1R inhibitor. By contrast, knockdown of Nanog abolished the effects of IGF2 on the colony formation ability of these LSCs. These findings suggest that the IGF2/IGF1R/Nanog signaling pathway plays a critical role in LSC proliferation.
Collapse
Affiliation(s)
- Dan-Dan Xu
- College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Ying Wang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Peng-Jun Zhou
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Shu-Rong Qin
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Rong Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Zhang
- Department of Biochemistry and Molecular Medicine, Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Xue Xue
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Jianping Wang
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Xia Wang
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Hong-Ce Chen
- Department of Pathogen Biology and Immunology, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao Wang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yu-Wei Pan
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Li Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Hai-Zhao Yan
- Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Kofu, Japan
| | - Qiu-Ying Liu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhong Liu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Su-Hong Chen
- College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Hong-Yuan Chen
- Department of Pathogen Biology and Immunology, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi-Fei Wang
- College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, Morrione A, Vigneri R. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocr Rev 2017; 38:379-431. [PMID: 28973479 PMCID: PMC5629070 DOI: 10.1210/er.2017-00073] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 02/08/2023]
Abstract
The insulin receptor (IR) gene undergoes differential splicing that generates two IR isoforms, IR-A and IR-B. The physiological roles of IR isoforms are incompletely understood and appear to be determined by their different binding affinities for insulin-like growth factors (IGFs), particularly for IGF-2. Predominant roles of IR-A in prenatal growth and development and of IR-B in metabolic regulation are well established. However, emerging evidence indicates that the differential expression of IR isoforms may also help explain the diversification of insulin and IGF signaling and actions in various organs and tissues by involving not only different ligand-binding affinities but also different membrane partitioning and trafficking and possibly different abilities to interact with a variety of molecular partners. Of note, dysregulation of the IR-A/IR-B ratio is associated with insulin resistance, aging, and increased proliferative activity of normal and neoplastic tissues and appears to sustain detrimental effects. This review discusses novel information that has generated remarkable progress in our understanding of the physiology of IR isoforms and their role in disease. We also focus on novel IR ligands and modulators that should now be considered as an important strategy for better and safer treatment of diabetes and cancer and possibly other IR-related diseases.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Veronica Vella
- School of Human and Social Sciences, University Kore of Enna, via della Cooperazione, 94100 Enna, Italy
| | - Michael C. Lawrence
- Structural Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Laura Sciacca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Francesco Frasca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Riccardo Vigneri
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| |
Collapse
|
25
|
Yan J, Xu Y, Wang H, Du T, Chen H. MicroRNA-503 inhibits the proliferation and invasion of breast cancer cells via targeting insulin-like growth factor 1 receptor. Mol Med Rep 2017; 16:1707-1714. [PMID: 28656281 PMCID: PMC5562074 DOI: 10.3892/mmr.2017.6816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 03/07/2017] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRs), a class of non-coding RNAs that are 18–25 nucleotides in length, serve as key regulators in the development and progression of human cancers. Previously, miR-503 has been implicated in breast cancer. However, the underlying mechanism of miR-503 in regulating the proliferation and invasion of breast cancer cells remains largely unknown. In the present study, reverse transcription-quantitative polymerase chain reaction analysis indicated that the expression of miR-503 was significantly reduced in breast cancer tissues compared with their matched adjacent normal tissues. Furthermore, miR-503 expression levels were markedly reduced in T2-T4 stage breast cancer, compared with T1 stage. Insulin-like growth factor 1 receptor (IGF-1R) was further identified as a novel target of miR-503. Overexpression of miR-503 significantly suppressed the protein expression levels of IGF-1R. Furthermore, it inhibited the proliferation and invasion of human breast cancer MCF-7 cells, as assessed by MTT and Transwell assays, respectively. However, restoration of IGF-1R expression markedly ameliorated the suppressive effects of miR-503 overexpression on MCF-7 cell proliferation and invasion, indicating that miR-503 inhibits breast cancer cell proliferation and invasion at least partially via directly targeting IGF-1R. Furthermore, the mRNA and protein expression levels of IGF-1R were demonstrated to be significantly increased in breast cancer tissues compared with their matched adjacent normal tissues. In addition, IGF-1R mRNA expression levels were reversely correlated with miR-503 expression levels in breast tumors, suggesting that the upregulation of IGF-1R may be due to downregulation of miR-503 in breast cancer. In conclusion, the present study expanded the understanding of the regulatory mechanism of miR-503 in breast cancer, and implicates the miR-503/IGF-1R axis as a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jingwang Yan
- Department of General Surgery, Xinxiang Center Hospital, Xinxiang, Henan 453000, P.R. China
| | - Yonghuan Xu
- Department of Oncology, People's Hospital of Xixia County, Nanyang, Henan 474550, P.R. China
| | - Haipeng Wang
- Department of General Surgery, Xinxiang Center Hospital, Xinxiang, Henan 453000, P.R. China
| | - Taiping Du
- Department of General Surgery, Xinxiang Center Hospital, Xinxiang, Henan 453000, P.R. China
| | - Hao Chen
- Department of General Surgery, Xinxiang Center Hospital, Xinxiang, Henan 453000, P.R. China
| |
Collapse
|
26
|
McDermott MSJ, Canonici A, Ivers L, Browne BC, Madden SF, O'Brien NA, Crown J, O'Donovan N. Dual inhibition of IGF1R and ER enhances response to trastuzumab in HER2 positive breast cancer cells. Int J Oncol 2017; 50:2221-2228. [PMID: 28498399 DOI: 10.3892/ijo.2017.3976] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/05/2017] [Indexed: 11/06/2022] Open
Abstract
Although HER2 targeted therapies have improved prognosis for HER2 positive breast cancer, HER2 positive cancers which co-express ER have poorer response rates to standard HER2 targeted therapies, combined with chemotherapy, than HER2 positive/ER negative breast cancer. Administration of hormone therapy concurrently with chemotherapy and HER2 targeted therapy is generally not recommended. Using publically available gene expression datasets we found that high expression of IGF1R is associated with shorter disease-free survival in patients whose tumors are ER positive and HER2 positive. IGF1R is frequently expressed in HER2 positive breast cancer and there is significant evidence for crosstalk between IGF1R and both HER2 and ER. Therefore, we evaluated the therapeutic potential of targeting ER and IGF1R in cell line models of HER2/ER/IGF1R positive breast cancer, using tamoxifen and two IGF1R targeted tyrosine kinase inhibitors (NVP-AEW541 and BMS-536924). Dual inhibition of ER and IGF1R enhanced growth inhibition in the four HER2 positive cell lines tested and caused an increase in cell cycle arrest in G1 in BT474 cells. In addition, combined treatment with trastuzumab, tamoxifen and either of the IGF1R TKIs enhanced response compared to dual targeting strategies in three of the four HER2 positive breast cancer cell lines tested. Furthermore, in a cell line model of trastuzumab-resistant HER2 positive breast cancer (BT474/Tr), tamoxifen combined with an IGF1R TKI produced a similar enhanced response as observed in the parental BT474 cells suggesting that this combination may overcome acquired trastuzumab resistance in this model. Combining ER and IGF1R targeting with HER2 targeted therapies may be an alternative to HER2 targeted therapy and chemotherapy for patients with HER2/ER/IGF1R positive breast cancer.
Collapse
Affiliation(s)
- Martina S J McDermott
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Alexandra Canonici
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Laura Ivers
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Brigid C Browne
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Stephen F Madden
- Population Health Sciences Division, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Neil A O'Brien
- Department of Medicine, Division of Haematology/Oncology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Norma O'Donovan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
27
|
Rodriguez-Barrueco R, Nekritz EA, Bertucci F, Yu J, Sanchez-Garcia F, Zeleke TZ, Gorbatenko A, Birnbaum D, Ezhkova E, Cordon-Cardo C, Finetti P, Llobet-Navas D, Silva JM. miR-424(322)/503 is a breast cancer tumor suppressor whose loss promotes resistance to chemotherapy. Genes Dev 2017; 31:553-566. [PMID: 28404630 PMCID: PMC5393051 DOI: 10.1101/gad.292318.116] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/06/2017] [Indexed: 01/08/2023]
Abstract
In this study, Rodriguez-Barrueco et al. analyzed ∼3000 primary tumors and show that miR-424(322)/503 is commonly lost in a subset of aggressive breast cancers; they then describe the genetic aberrations that inactivate its expression. Their data show that miR-424(322)/503 is a tumor suppressor in breast cancer and provide a link between mammary epithelial involution, tumorigenesis, and the phenomenon of chemoresistance. The female mammary gland is a very dynamic organ that undergoes continuous tissue remodeling during adulthood. Although it is well established that the number of menstrual cycles and pregnancy (in this case transiently) increase the risk of breast cancer, the reasons are unclear. Growing clinical and experimental evidence indicates that improper involution plays a role in the development of this malignancy. Recently, we described the miR-424(322)/503 cluster as an important regulator of mammary epithelial involution after pregnancy. Here, through the analysis of ∼3000 primary tumors, we show that miR-424(322)/503 is commonly lost in a subset of aggressive breast cancers and describe the genetic aberrations that inactivate its expression. Furthermore, through the use of a knockout mouse model, we demonstrate for the first time that loss of miR-424(322)/503 promotes breast tumorigenesis in vivo. Remarkably, we found that loss of miR-424(322)/503 promotes chemoresistance due to the up-regulation of two of its targets: BCL-2 and insulin-like growth factor-1 receptor (IGF1R). Importantly, targeted therapies blocking the aberrant activity of these targets restore sensitivity to chemotherapy. Overall, our studies reveal miR-424(322)/503 as a tumor suppressor in breast cancer and provide a link between mammary epithelial involution, tumorigenesis, and the phenomenon of chemoresistance.
Collapse
Affiliation(s)
- Ruth Rodriguez-Barrueco
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Institute of Genetic Medicine, Newcastle University, Newcastle-Upon-Tyne NE1 3BZ, United Kingdom
| | - Erin A Nekritz
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - François Bertucci
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille 13009, France
| | - Jiyang Yu
- St. Jude Children's Research Hospital, Kay Research and Care Center, IA6053, Memphis, Tennessee 38105, USA
| | - Felix Sanchez-Garcia
- Department of Systems Biology, Center for Computational Biology and Bioinformatics, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Tizita Z Zeleke
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Andrej Gorbatenko
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Daniel Birnbaum
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille 13009, France
| | - Elena Ezhkova
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Pascal Finetti
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille 13009, France
| | - David Llobet-Navas
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Institute of Genetic Medicine, Newcastle University, Newcastle-Upon-Tyne NE1 3BZ, United Kingdom
| | - Jose M Silva
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
28
|
Chakraborty A, Hatzis C, DiGiovanna MP. Co-targeting the HER and IGF/insulin receptor axis in breast cancer, with triple targeting with endocrine therapy for hormone-sensitive disease. Breast Cancer Res Treat 2017; 163:37-50. [PMID: 28236033 DOI: 10.1007/s10549-017-4169-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/17/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE Interactions between HER2, estrogen receptor (ER), and insulin-like growth factor I receptor (IGF1R) are implicated in resistance to monotherapies targeting these receptors. We have previously shown in pre-clinical studies synergistic anti-tumor effects for co-targeting each pairwise combination of HER2, IGF1R, and ER. Strikingly, synergy for HER2/IGF1R targeting occurred not only in a HER2+ model, but also in a HER2-normal model. The purpose of the current study was therefore to determine the generalizability of synergistic anti-tumor effects of co-targeting HER2/IGF1R, the anti-tumor activity of triple-targeting HER2/IGF1R/ER in hormone-dependent cell lines, and the effect of using the multi-targeting drugs neratinib (pan-HER) and BMS-754807 (dual IGF1R/insulin receptor). METHODS Proliferation and apoptosis assays were performed in a large panel of cell lines representing varying receptor expression levels. Mechanistic effects were studied using phospho-protein immunoblotting. Analyses of drug interaction effects were performed using linear mixed-effects regression models. RESULTS Enhanced anti-proliferative effects of HER/IGF-insulin co-targeting were seen in most, though not all, cell lines, including HER2-normal lines. For ER+ lines, triple targeting with inclusion of anti-estrogen generally resulted in the greatest anti-tumor effects. Double or triple targeting generally resulted in marked increases in apoptosis in the sensitive lines. Mechanistic studies demonstrated that the synergy between drugs was correlated with maximal inhibition of Akt and ERK pathway signaling. CONCLUSIONS Dual HER/IGF-insulin targeting, and triple targeting with inclusion of anti-estrogen drugs, shows striking anti-tumor activity across breast cancer types, and drugs with broader receptor specificity may be more effective than single receptor selective drugs, particularly for ER- cells.
Collapse
Affiliation(s)
- Ashok Chakraborty
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | - Christos Hatzis
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | - Michael P DiGiovanna
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA.
| |
Collapse
|
29
|
Cotargeting of CYP-19 (aromatase) and emerging, pivotal signalling pathways in metastatic breast cancer. Br J Cancer 2016; 116:10-20. [PMID: 27923036 PMCID: PMC5220158 DOI: 10.1038/bjc.2016.405] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/28/2016] [Accepted: 11/08/2016] [Indexed: 01/08/2023] Open
Abstract
Aromatase inhibition is one of the cornerstones of modern endocrine therapy of oestrogen receptor-positive (ER+) metastatic breast cancer (MBC). The nonsteroidal aromatase inhibitors anastrozole and letrozole, as well as the steroidal aromatase inactivator exemestane, are the preferred drugs and established worldwide in all clinical phases of the disease. However, although many patients suffering from MBC experience an initial stabilisation of their metastatic burden, drug resistance and disease progression occur frequently, following in general only a few months on treatment. Extensive translational research during the past two decades has elucidated the major pathways contributing to endocrine resistance and paved the way for clinical studies investigating the efficacy of novel drug combinations involving aromatase inhibitors and emerging drugable targets like mTOR, PI3K and CDK4/6. The present review summarises the basic research that provided the rationale for new drug combinations involving aromatase inhibitors and the main findings of pivotal clinical trials that have already started to change our way to treat hormone-sensitive MBC. The challenging situation of oestrogen receptor-positive and human epidermal growth factor receptor 2-positive (HER2+) MBC is also shortly reviewed to underline the complexity of the clinical scenario in the heterogeneous subgroups of hormone receptor-positive breast cancer patients and the increasing need for personalised medicine. Finally, we summarise some of the promising findings made with the combination of aromatase inhibitors with other potent endocrine treatment options like fulvestrant, a selective oestrogen receptor downregulator.
Collapse
|
30
|
Ochnik AM, Baxter RC. Combination therapy approaches to target insulin-like growth factor receptor signaling in breast cancer. Endocr Relat Cancer 2016; 23:R513-R536. [PMID: 27733416 DOI: 10.1530/erc-16-0218] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/09/2016] [Indexed: 12/19/2022]
Abstract
Insulin-like growth factor receptor (IGF1R) signaling as a therapeutic target has been widely studied and clinically tested. Despite the vast amount of literature supporting the biological role of IGF1R in breast cancer, effective clinical translation in targeting its activity as a cancer therapy has not been successful. The intrinsic complexity of cancer cell signaling mediated by many tyrosine kinase growth factor receptors that work together to modulate each other and intracellular downstream mediators in the cell highlights that studying IGF1R expression and activity as a prognostic factor and therapeutic target in isolation is certainly associated with problems. This review discusses the current literature and clinical trials associated with IGF-1 signaling and attempts to look at new ways of designing novel IGF1R-directed breast cancer therapy approaches to target its activity
and/or intracellular downstream signaling pathways in IGF1R-expressing breast cancers.
Collapse
Affiliation(s)
- Aleksandra M Ochnik
- Kolling Institute of Medical ResearchUniversity of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Robert C Baxter
- Kolling Institute of Medical ResearchUniversity of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
31
|
Becker MA, Hou X, Tienchaianada P, Haines BB, Harrington SC, Weroha SJ, Sathyanarayanan S, Haluska P. Ridaforolimus (MK-8669) synergizes with Dalotuzumab (MK-0646) in hormone-sensitive breast cancer. BMC Cancer 2016; 16:814. [PMID: 27765027 PMCID: PMC5073873 DOI: 10.1186/s12885-016-2847-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/07/2016] [Indexed: 11/16/2022] Open
Abstract
Background Mammalian target of rapamycin (mTOR) represents a key downstream intermediate for a myriad of oncogenic receptor tyrosine kinases. In the case of the insulin-like growth factor (IGF) pathway, the mTOR complex (mTORC1) mediates IGF-1 receptor (IGF-1R)-induced estrogen receptor alpha (ERα) phosphorylation/activation and leads to increased proliferation and growth in breast cancer cells. As a result, the prevalence of mTOR inhibitors combined with hormonal therapy has increased in recent years. Conversely, activated mTORC1 provides negative feedback regulation of IGF signaling via insulin receptor substrate (IRS)-1/2 serine phosphorylation and subsequent proteasomal degradation. Thus, the IGF pathway may provide escape (e.g. de novo or acquired resistance) from mTORC1 inhibitors. It is therefore plausible that combined inhibition of mTORC1 and IGF-1R for select subsets of ER-positive breast cancer patients presents as a viable therapeutic option. Methods Using hormone-sensitive breast cancer cells stably transfected with the aromatase gene (MCF-7/AC-1), works presented herein describe the in vitro and in vivo antitumor efficacy of the following compounds: dalotuzumab (DALO; “MK-0646”; anti-IGF-1R antibody), ridaforolimus (RIDA; “MK-8669”; mTORC1 small molecule inhibitor) and letrozole (“LET”, aromatase inhibitor). Results With the exception of MK-0646, all single agent and combination treatment arms effectively inhibited xenograft tumor growth, albeit to varying degrees. Correlative tissue analyses revealed MK-0646 alone and in combination with LET induced insulin receptor alpha A (InsR-A) isoform upregulation (both mRNA and protein expression), thereby further supporting a triple therapy approach. Conclusion These data provide preclinical rationalization towards the combined triple therapy of LET plus MK-0646 plus MK-8669 as an efficacious anti-tumor strategy for ER-positive breast tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2847-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marc A Becker
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA. .,Division of Medical Oncology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Piyawan Tienchaianada
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA.,Oncology unit, Department of Medicine, Rajavithi Hospital, Bangkok, 10400, Thailand
| | - Brian B Haines
- Molecular Oncology, Merck Research Laboratories, Boston, MA, 02115, USA
| | | | - S John Weroha
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Paul Haluska
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
32
|
Clinical studies in humans targeting the various components of the IGF system show lack of efficacy in the treatment of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 772:105-122. [PMID: 28528684 DOI: 10.1016/j.mrrev.2016.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 01/28/2023]
Abstract
The insulin-like growth factors (IGFs) system regulates cell growth, differentiation and energy metabolism and plays crucial role in the regulation of key aspects of tumor biology, such as cancer cell growth, survival, transformation and invasion. The current focus for cancer therapeutic approaches have shifted from the conventional treatments towards the targeted therapies and the IGF system has gained a great interest as anti-cancer therapy. The proliferative, anti-apoptotic and transformation effects of IGFs are mainly triggered by the ligation of the type I IGF receptor (IGF-IR). Thus, aiming at developing novel and effective cancer therapies, different strategies have been employed to target IGF system in human malignancies, including but not limited to ligand or receptor neutralizing antibodies and IGF-IR signaling inhibitors. In this review, we have focused on the clinical studies that have been conducted targeting the various components of the IGF system for the treatment of different types of cancer, providing a description and the challenges of each targeting strategy and the degree of success.
Collapse
|
33
|
The insulin-like growth factor-I receptor (IGF-IR) in breast cancer: biology and treatment strategies. Tumour Biol 2016; 37:11711-11721. [PMID: 27444280 DOI: 10.1007/s13277-016-5176-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/12/2016] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most common cancer and the second leading cause of cancer-related deaths among women worldwide. Although patients are often diagnosed in the early and curable stages, the treatment of metastatic breast cancer remains a major clinical challenge. The combination of chemotherapy with new targeting agents, such as bevacizumab, is helpful in improving patient survival; however, novel treatment strategies are required to improve clinical outcomes. The insulin-like growth factor-I receptor (IGF-IR) is a tyrosine kinase cell surface receptor which is involved in the regulation of cell growth and metabolism. Previous studies have shown that activation of the IGF-IR signaling pathway promotes proliferation, survival, and metastasis of breast cancer cells. Additionally, overexpression of IGF-IR is associated with breast cancer cell resistance to anticancer therapies. Recently, IGF-IR has been introduced as a marker of stemness in breast cancer cells and there is also accumulating evidence that IGF-IR contributes to the establishment and maintenance of breast cancer epithelial-mesenchymal transition (EMT). Therefore, pharmacological or molecular targeting of IGF-IR could be a promising strategy, in the treatment of patients with breast cancer, particularly in order to circumvent the therapeutic resistance and targeting breast cancer stem/progenitors. Currently, many strategies have been developed for targeting IGF-IR, some have entered clinical trials and some are in preclinical stages for breast cancer therapy. In this review, we will first discuss on the biology of IGF-IR in an attempt to find the role of this receptor in breast cancer and then discuss about therapeutic strategies to target this receptor.
Collapse
|
34
|
Iams WT, Lovly CM. Molecular Pathways: Clinical Applications and Future Direction of Insulin-like Growth Factor-1 Receptor Pathway Blockade. Clin Cancer Res 2016; 21:4270-7. [PMID: 26429980 DOI: 10.1158/1078-0432.ccr-14-2518] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The IGF1R signaling pathway is a complex and tightly regulated network that is critical for cell proliferation, growth, and survival. IGF1R is a potential therapeutic target for patients with many different malignancies. This brief review summarizes the results of clinical trials targeting the IGF1R pathway in patients with breast cancer, sarcoma, and non-small cell lung cancer (NSCLC). Therapeutic agents discussed include both monoclonal antibodies to IGF1R (dalotuzumab, figitumumab, cixutumumab, ganitumab, R1507, AVE1642) and newer IGF1R pathway targeting strategies, including monoclonal antibodies to IGF1 and IGF2 (MEDI-573 and BI 836845) and a small-molecule tyrosine kinase inhibitor of IGF1R (linsitinib). The pullback of trials in patients with breast cancer and NSCLC based on several large negative trials is noted and contrasted with the sustained success of IGF1R inhibitor monotherapy in a subset of patients with sarcoma. Several different biomarkers have been examined in these trials with varying levels of success, including tumor expression of IGF1R and its pathway components, serum IGF ligand levels, alternate pathway activation, and specific molecular signatures of IGF1R pathway dependence. However, there remains a critical need to define predictive biomarkers in order to identify patients who may benefit from IGF1R-directed therapies. Ongoing research focuses on uncovering such biomarkers and elucidating mechanisms of resistance, as this therapeutic target is currently being analyzed from the bedside to bench.
Collapse
Affiliation(s)
- Wade T Iams
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M Lovly
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee. Vanderbilt-Ingram Cancer Center, Nashville, Tennessee.
| |
Collapse
|
35
|
A comprehensive pharmacokinetic/pharmacodynamics analysis of the novel IGF1R/INSR inhibitor BI 893923 applying in vitro, in vivo and in silico modeling techniques. Cancer Chemother Pharmacol 2016; 77:1303-14. [DOI: 10.1007/s00280-016-3049-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/27/2016] [Indexed: 01/12/2023]
|
36
|
Franks SE, Jones RA, Briah R, Murray P, Moorehead RA. BMS-754807 is cytotoxic to non-small cell lung cancer cells and enhances the effects of platinum chemotherapeutics in the human lung cancer cell line A549. BMC Res Notes 2016; 9:134. [PMID: 26928578 PMCID: PMC4772483 DOI: 10.1186/s13104-016-1919-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/08/2016] [Indexed: 12/11/2022] Open
Abstract
Background Despite advances in targeted therapy for lung cancer, survival for patients remains poor and lung cancer remains the leading cause of cancer-related deaths worldwide. The type I insulin-like growth factor receptor (IGF-IR) has emerged as a potential target for lung cancer treatment, however, clinical trials to date have provided disappointing results. Further research is needed to identify if certain patients would benefit from IGF-IR targeted therapies and the ideal approach to incorporate IGF-IR targeted agents with current therapies. Methods The dual IGF-IR/insulin receptor inhibitor, BMS-754807, was evaluated alone and in combination with platinum-based chemotherapeutics in two human non-small cell lung cancer (NSCLC) cell lines. Cell survival was determined using WST-1 assays and drug interaction was evaluated using Calcusyn software. Proliferation and apoptosis were determined using immunofluorescence for phospho-histone H3 and cleaved caspase 3, respectively. Results Treatment with BMS-754807 alone reduced cell survival and wound closure while enhancing apoptosis in both human lung cancer cell lines. These effects appear to be mediated through IGF-IR/IR signaling and, at least in part, through the PI3K/AKT pathway as administration of BMS-754807 to A549 or NCI-H358 cells significantly suppressed IGF-IR/IR and AKT phosphorylation. In addition of BMS-754807 enhanced the cytotoxic effects of carboplatin or cisplatin in a synergistic manner when given simultaneously to A549 cells. Conclusions BMS-754807 may be an effective therapeutic agent for the treatment of NSCLC, particularly in lung cancer cells expressing high levels of IGF-IR.
Collapse
Affiliation(s)
- S Elizabeth Franks
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Robert A Jones
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Ritesh Briah
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Payton Murray
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Roger A Moorehead
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| |
Collapse
|
37
|
Sanderson MP, Apgar J, Garin-Chesa P, Hofmann MH, Kessler D, Quant J, Savchenko A, Schaaf O, Treu M, Tye H, Zahn SK, Zoephel A, Haaksma E, Adolf GR, Kraut N. BI 885578, a Novel IGF1R/INSR Tyrosine Kinase Inhibitor with Pharmacokinetic Properties That Dissociate Antitumor Efficacy and Perturbation of Glucose Homeostasis. Mol Cancer Ther 2015; 14:2762-72. [DOI: 10.1158/1535-7163.mct-15-0539] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/14/2015] [Indexed: 11/16/2022]
|
38
|
Zhao Q, Tran H, Dimitrov DS, Cheung NKV. A dual-specific anti-IGF-1/IGF-2 human monoclonal antibody alone and in combination with temsirolimus for therapy of neuroblastoma. Int J Cancer 2015; 137:2243-52. [PMID: 25924852 DOI: 10.1002/ijc.29588] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/14/2015] [Indexed: 12/16/2022]
Abstract
The insulin-like growth factors (IGFs), IGF-1 and IGF-2, have been implicated in the growth, survival and metastasis of a broad range of malignancies including pediatric tumors. They bind to the IGF receptor type 1 (IGF-1R) and the insulin receptor (IR) which are overexpressed in many types of solid malignancies. Activation of the IR by IGF-2 results in increased survival of tumor cells. We have previously identified a novel human monoclonal antibody, m708.5, which binds with high (pM) affinity to both human IGF-1 and IGF-2, and potently inhibits phosphorylation of the IGF-1R and the IR in tumor cells. m708.5 exhibited strong antitumor activity as a single agent against most cell lines derived from neuroblastoma, Ewing family of tumor, rhabdomyosarcoma and osteosarcoma. When tested in neuroblastoma cell lines, it showed strong synergy with temsirolimus and synergy with chemotherapeutic agents in vitro. In xenograft models, the combination of m708.5 and temsirolimus significantly inhibited neuroblastoma growth and prolonged mouse survival. Taken together, these results support the clinical development of m708.5 for pediatric solid tumors with potential for synergy with chemotherapy and mTOR inhibitors.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY.,Laboratory of Fully Human Antibody Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Guangdong, China
| | - Hoa Tran
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Dimiter S Dimitrov
- Protein Interaction Section, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, MD, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
39
|
Glaser G, Weroha SJ, Becker MA, Hou X, Enderica-Gonzalez S, Harrington SC, Haluska P. Conventional chemotherapy and oncogenic pathway targeting in ovarian carcinosarcoma using a patient-derived tumorgraft. PLoS One 2015; 10:e0126867. [PMID: 25962155 PMCID: PMC4427104 DOI: 10.1371/journal.pone.0126867] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 04/08/2015] [Indexed: 11/18/2022] Open
Abstract
Ovarian carcinosarcoma is a rare subtype of ovarian cancer with poor clinical outcomes. The low incidence of this disease makes accrual to large clinical trials challenging. However, studies have shown that treatment responses in patient-derived xenograft (PDX) models correlate with matched-patient responses in the clinic, supporting their use for preclinical testing of standard and novel therapies. An ovarian carcinosarcoma PDX is presented herein and showed resistance to carboplatin and paclitaxel (similar to the patient) but exhibited significant sensitivity to ifosfamide and paclitaxel. The PDX demonstrated overexpression of EGFR mRNA and gene amplification by array comparative genomic hybridization (log2 ratio 0.399). EGFR phosphorylation was also detected. Angiogensis and insulin-like growth factor pathways were also implicated by overexpression of VEGFC and IRS1. In order to improve response to chemotherapy, the PDX was treated with carboplatin/paclitaxel with or without a pan-HER and VEGF inhibitor (BMS-690514) but there was no tumor growth inhibition or improved animal survival, which may be explained by a KRAS mutation. Resistance was also observed when the IGF-1R inhibitor BMS-754807 was combined with carboplatin/paclitaxel. Because poly (ADP-ribose) polymerase inhibitors have activity in ovarian cancer patients, with and without BRCA mutations, ABT-888 was also tested but found to have no activity. Pathogenic mutations were also detected in TP53 and PIK3CA. In conclusion, ifosfamide/paclitaxel was superior to carboplatin/paclitaxel in this ovarian carcinosarcoma PDX and gene overexpression or amplification alone was not sufficient to predict response to targeted therapy. Better predictive markers of response are needed.
Collapse
Affiliation(s)
- Gretchen Glaser
- Division of Gynecologic Surgery, Mayo Clinic, Rochester, MN, United States of America
| | - S. John Weroha
- Department of Oncology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| | - Marc A. Becker
- Department of Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, MN, United States of America
| | | | - Sean C. Harrington
- Department of Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Paul Haluska
- Department of Oncology, Mayo Clinic, Rochester, MN, United States of America
| |
Collapse
|
40
|
A high-throughput in vitro drug screen in a genetically engineered mouse model of diffuse intrinsic pontine glioma identifies BMS-754807 as a promising therapeutic agent. PLoS One 2015; 10:e0118926. [PMID: 25748921 PMCID: PMC4352073 DOI: 10.1371/journal.pone.0118926] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 01/20/2015] [Indexed: 11/24/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) represent a particularly lethal type of pediatric brain cancer with no effective therapeutic options. Our laboratory has previously reported the development of genetically engineered DIPG mouse models using the RCAS/tv-a system, including a model driven by PDGF-B, H3.3K27M, and p53 loss. These models can serve as a platform in which to test novel therapeutics prior to the initiation of human clinical trials. In this study, an in vitro high-throughput drug screen as part of the DIPG preclinical consortium using cell-lines derived from our DIPG models identified BMS-754807 as a drug of interest in DIPG. BMS-754807 is a potent and reversible small molecule multi-kinase inhibitor with many targets including IGF-1R, IR, MET, TRKA, TRKB, AURKA, AURKB. In vitro evaluation showed significant cytotoxic effects with an IC50 of 0.13 μM, significant inhibition of proliferation at a concentration of 1.5 μM, as well as inhibition of AKT activation. Interestingly, IGF-1R signaling was absent in serum-free cultures from the PDGF-B; H3.3K27M; p53 deficient model suggesting that the antitumor activity of BMS-754807 in this model is independent of IGF-1R. In vivo, systemic administration of BMS-754807 to DIPG-bearing mice did not prolong survival. Pharmacokinetic analysis demonstrated that tumor tissue drug concentrations of BMS-754807 were well below the identified IC50, suggesting that inadequate drug delivery may limit in vivo efficacy. In summary, an unbiased in vitro drug screen identified BMS-754807 as a potential therapeutic agent in DIPG, but BMS-754807 treatment in vivo by systemic delivery did not significantly prolong survival of DIPG-bearing mice.
Collapse
|
41
|
Huzell L, Persson M, Simonsson M, Markkula A, Ingvar C, Rose C, Jernström H. History of oral contraceptive use in breast cancer patients: impact on prognosis and endocrine treatment response. Breast Cancer Res Treat 2015; 149:505-15. [DOI: 10.1007/s10549-014-3252-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/17/2014] [Indexed: 01/01/2023]
|
42
|
Bowers LW, Rossi EL, O’Flanagan CH, deGraffenried LA, Hursting SD. The Role of the Insulin/IGF System in Cancer: Lessons Learned from Clinical Trials and the Energy Balance-Cancer Link. Front Endocrinol (Lausanne) 2015; 6:77. [PMID: 26029167 PMCID: PMC4432799 DOI: 10.3389/fendo.2015.00077] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/29/2015] [Indexed: 02/06/2023] Open
Abstract
Numerous epidemiological and pre-clinical studies have demonstrated that the insulin/insulin-like growth factor (IGF) system plays a key role in the development and progression of several types of cancer. Insulin/IGF signaling, in cooperation with chronic low-grade inflammation, is also an important contributor to the cancer-promoting effects of obesity. However, clinical trials for drugs targeting different components of this system have produced largely disappointing results, possibly due to the lack of predictive biomarker use and problems with the design of combination therapy regimens. With careful attention to the identification of likely patient responders and optimal drug combinations, the outcome of future trials may be improved. Given that insulin/IGF signaling is known to contribute to obesity-associated cancer, further investigation regarding the efficacy of drugs targeting this system and its downstream effectors in the obese patient population is warranted.
Collapse
Affiliation(s)
- Laura W. Bowers
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily L. Rossi
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ciara H. O’Flanagan
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- *Correspondence: Stephen D. Hursting, Department of Nutrition, University of North Carolina at Chapel Hill, 135 Dauer Drive, McGavran-Greenberg Hall, Chapel Hill, NC 27599, USA,
| |
Collapse
|
43
|
Huang F, Chang H, Greer A, Hillerman S, Reeves KA, Hurlburt W, Cogswell J, Patel D, Qi Z, Fairchild C, Ryseck RP, Wong TW, Finckenstein FG, Jackson J, Carboni JM. IRS2 copy number gain, KRAS and BRAF mutation status as predictive biomarkers for response to the IGF-1R/IR inhibitor BMS-754807 in colorectal cancer cell lines. Mol Cancer Ther 2014; 14:620-30. [PMID: 25527633 DOI: 10.1158/1535-7163.mct-14-0794-t] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin-like growth factor receptor 1 (IGF-1R)-targeting therapies are currently at an important crossroad given the low clinical response rates seen in unselected patients. Predictive biomarkers for patient selection are critical for improving clinical benefit. Coupling in vitro sensitivity testing of BMS-754807, a dual IGF-1R/IR inhibitor, with genomic interrogations in 60 human colorectal cancer cell lines, we identified biomarkers correlated with response to BMS-754807. The results showed that cell lines with BRAF(V600E) or KRAS(G13D) mutation were resistant, whereas cell lines with wild-type of both KRAS and BRAF were particularly sensitive to BMS-754807 if they have either higher RNA expression levels of IR-A or lower levels of IGFBP6. In addition, the cell lines with KRAS mutations, those with either insulin receptor substrate 2 (IRS2) copy number gain (CNG) or higher IGF-1R expression levels, were more sensitive to the drug. Furthermore, cell lines with IRS2 CNG had higher levels of ligand-stimulated activation of IGF-1R and AKT, suggesting that these cell lines with IGF-IR signaling pathways more actively coupled to AKT signaling are more responsive to IGF-1R/IR inhibition. IRS2 siRNA knockdown reduced IRS2 protein expression levels and decreased sensitivity to BMS-754807, providing evidence for the functional involvement of IRS2 in mediating the drug response. The prevalence of IRS2 CNG in colorectal cancer tumors as measured by qPCR-CNV is approximately 35%. In summary, we identified IRS2 CNG, IGF-1R, IR-A, and IGFBP6 RNA expression levels, and KRAS and BRAF mutational status as candidate predictive biomarkers for response to BMS-754807. This work proposed clinical development opportunities for BMS-754807 in colorectal cancer with patient selection to improve clinical benefit.
Collapse
Affiliation(s)
- Fei Huang
- Bristol-Myers Squibb Company, Princeton, New Jersey.
| | - Han Chang
- Bristol-Myers Squibb Company, Princeton, New Jersey.
| | - Ann Greer
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | | | | | | | | | | | - Zhenhao Qi
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | | | | | - Tai W Wong
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | | | | | | |
Collapse
|
44
|
Beckwith H, Yee D. Insulin-like growth factors, insulin, and growth hormone signaling in breast cancer: implications for targeted therapy. Endocr Pract 2014; 20:1214-21. [PMID: 25297664 DOI: 10.4158/ep14208.ra] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In recent decades, multiple therapeutics targeting the estrogen and human epidermal growth factor-2 (HER2) receptors have been approved for the treatment of breast cancer. METHODS This review discusses a number of growth factor pathways that have been implicated in resistance to both anti-estrogen and HER2-targeted therapies. The association between growth factors and breast cancer is well established. Over decades, numerous laboratories have studied the link between insulin-like growth factor (IGF), insulin, and growth hormone (GH) to the development and progression of breast cancer. RESULTS Although preclinical data demonstrates that blockade of these receptors inhibits breast cancer growth, progression, and drug resistance, therapies targeting the IGF, insulin, and GH receptors (GHRs) have not been successful in producing significant increases in progression-free, disease-free, or overall survival for patients with breast cancer. The failure to demonstrate a benefit of growth factor blockade in clinical trials can be attributed to redundancy in IGF, insulin, and GHR signaling pathways. All 3 receptors are able to activate oncogenic phosphoinositide-3 kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. CONCLUSION Consequently, multitargeted blockade of growth factor receptors and their common downstream kinases will be necessary for the successful treatment of breast cancer.
Collapse
Affiliation(s)
- Heather Beckwith
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota Department of Medicine, University of Minnesota, Minneapolis, Minnesota Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
45
|
King H, Aleksic T, Haluska P, Macaulay VM. Can we unlock the potential of IGF-1R inhibition in cancer therapy? Cancer Treat Rev 2014; 40:1096-105. [PMID: 25123819 DOI: 10.1016/j.ctrv.2014.07.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 12/20/2022]
Abstract
IGF-1R inhibitors arrived in the clinic accompanied by optimism based on preclinical activity of IGF-1R targeting, and recognition that low IGF bioactivity protects from cancer. This was tempered by concerns about toxicity to normal tissue IGF-1R and cross-reactivity with insulin receptor (InsR). In fact, toxicity is not a show-stopper; the key issue is efficacy. While IGF-1R inhibition induces responses as monotherapy in sarcomas and with chemotherapy or targeted agents in common cancers, negative Phase 2/3 trials in unselected patients prompted the cessation of several Pharma programs. Here, we review completed and on-going trials of IGF-1R antibodies, kinase inhibitors and ligand antibodies. We assess candidate biomarkers for patient selection, highlighting the potential predictive value of circulating IGFs/IGFBPs, the need for standardized assays for IGF-1R, and preclinical evidence that variant InsRs mediate resistance to IGF-1R antibodies. We review hypothesis-led and unbiased approaches to evaluate IGF-1R inhibitors with other agents, and stress the need to consider sequencing with chemotherapy. The last few years were a tough time for IGF-1R therapeutics, but also brought progress in understanding IGF biology. Even failed studies include patients who derived benefit; they should be investigated to identify features distinguishing the tumors and host environment of responders from non-responders. We emphasize the importance of incorporating biospecimen collection into trial design, and wording patient consents to allow post hoc analysis of trial material as new data become available. Such information represents the key to unlocking the potential of this approach, to inform the next generation of trials of IGF signalling inhibitors.
Collapse
Affiliation(s)
- Helen King
- St Catherine's College, University of Oxford, Manor Road, Oxford OX1 3UJ, UK.
| | - Tamara Aleksic
- Department of Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, UK.
| | - Paul Haluska
- Division of Medical Oncology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN 55905, USA.
| | - Valentine M Macaulay
- Department of Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, UK; Oxford Cancer Centre, Churchill Hospital, Oxford OX3 7LE, UK.
| |
Collapse
|
46
|
Ades F, Zardavas D, Bozovic-Spasojevic I, Pugliano L, Fumagalli D, de Azambuja E, Viale G, Sotiriou C, Piccart M. Luminal B breast cancer: molecular characterization, clinical management, and future perspectives. J Clin Oncol 2014; 32:2794-803. [PMID: 25049332 DOI: 10.1200/jco.2013.54.1870] [Citation(s) in RCA: 263] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gene expression profiling has reshaped our understanding of breast cancer by defining and characterizing four main intrinsic molecular subtypes: human epidermal growth factor receptor 2-enriched, basal-like, luminal A, and luminal B subtypes. Luminal B breast cancer has been reported to have lower expression of hormone receptors, higher expression of proliferation markers, and higher histologic grade than luminal A. It also exhibits worse prognosis and has a distinct profile of response to hormone therapy and chemotherapy. Although luminal cancers share similarities, the studies conducted in recent years using next-generation sequencing technology show that luminal A and B breast cancers should be perceived as distinct entities, with specific oncogenic drivers, rather than more proliferative varieties of luminal tumors. This review discusses the definition and molecular characterization of luminal B breast cancer and presents the available clinical evidence for chemotherapy and endocrine therapy patterns of response. It also provides an overview of ongoing research on molecularly targeted agents for this disease.
Collapse
Affiliation(s)
- Felipe Ades
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Dimitrios Zardavas
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Ivana Bozovic-Spasojevic
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Lina Pugliano
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Debora Fumagalli
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Evandro de Azambuja
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Giuseppe Viale
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Christos Sotiriou
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy
| | - Martine Piccart
- Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Christos Sotiriou, and Martine Piccart, Institut Jules Bordet, Université Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
47
|
Haluska P, Menefee M, Plimack ER, Rosenberg J, Northfelt D, LaVallee T, Shi L, Yu XQ, Burke P, Huang J, Viner J, McDevitt J, LoRusso P. Phase I dose-escalation study of MEDI-573, a bispecific, antiligand monoclonal antibody against IGFI and IGFII, in patients with advanced solid tumors. Clin Cancer Res 2014; 20:4747-57. [PMID: 25024259 DOI: 10.1158/1078-0432.ccr-14-0114] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE This phase I, multicenter, open-label, single-arm, dose-escalation, and dose-expansion study evaluated the safety, tolerability, and antitumor activity of MEDI-573 in adults with advanced solid tumors refractory to standard therapy or for which no standard therapy exists. EXPERIMENTAL DESIGN Patients received MEDI-573 in 1 of 5 cohorts (0.5, 1.5, 5, 10, or 15 mg/kg) dosed weekly or 1 of 2 cohorts (30 or 45 mg/kg) dosed every 3 weeks. Primary end points included the MEDI-573 safety profile, maximum tolerated dose (MTD), and optimal biologic dose (OBD). Secondary end points included MEDI-573 pharmacokinetics (PK), pharmacodynamics, immunogenicity, and antitumor activity. RESULTS In total, 43 patients (20 with urothelial cancer) received MEDI-573. No dose-limiting toxicities were identified, and only 1 patient experienced hyperglycemia related to treatment. Elevations in levels of insulin and/or growth hormone were not observed. Adverse events observed in >10% of patients included fatigue, anorexia, nausea, diarrhea, and anemia. PK evaluation demonstrated that levels of MEDI-573 increased with dose at all dose levels tested. At doses >5 mg/kg, circulating levels of insulin-like growth factor (IGF)-I and IGFII were fully suppressed. Of 39 patients evaluable for response, none experienced partial or complete response and 13 had stable disease as best response. CONCLUSIONS The MTD of MEDI-573 was not reached. The OBD was 5 mg/kg weekly or 30 or 45 mg/kg every 3 weeks. MEDI-573 showed preliminary antitumor activity in a heavily pretreated population and had a favorable tolerability profile, with no notable perturbations in metabolic homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Shi
- MedImmune, Gaithersburg, Maryland
| | | | | | | | | | | | | |
Collapse
|
48
|
Austreid E, Lonning PE, Eikesdal HP. The emergence of targeted drugs in breast cancer to prevent resistance to endocrine treatment and chemotherapy. Expert Opin Pharmacother 2014; 15:681-700. [PMID: 24579888 DOI: 10.1517/14656566.2014.885952] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Deregulated signaling pathways are associated with resistance to chemotherapy and endocrine treatment, providing a rationale for the implementation of novel targeted therapies in breast cancer therapy. Key molecules targeted therapeutically in ongoing clinical breast cancer trials are phosphoinositide 3-kinase-Akt-mammalian target of rapamycin (mTOR), Src, insulin-like growth factor 1 receptor, heat shock protein-90, histone deacetylases, cyclin-dependent kinases (CDKs), Notch and human epidermal growth factor receptors (HERs). AREAS COVERED This review provides an overview of novel targeted agents currently explored in clinical breast cancer trials and registered in ClinicalTrials.gov. The main focus will be on their ability to prevent or reverse endocrine resistance and chemoresistance in breast cancer. EXPERT OPINION HER2 targeted agents have extended survival substantially, both in the adjuvant and metastatic setting, pointing to a crucial dependency on this pathway in HER2-amplified breast cancer, including drug resistance reversal. While data on mTOR inhibitors are encouraging and preliminary results on CDK4/6 and Src inhibitors exciting, so far other targeted agents have been of limited benefit when added in concert with conventional therapies. Future clinical trials should systematically explore biomarkers and defects in functional gene cascades to identify relevant biological mechanisms to be targeted therapeutically in breast cancer.
Collapse
Affiliation(s)
- Eilin Austreid
- University of Bergen, Department of Clinical Science, Section of Oncology , Bergen , Norway
| | | | | |
Collapse
|
49
|
Palmieri C, Patten DK, Januszewski A, Zucchini G, Howell SJ. Breast cancer: current and future endocrine therapies. Mol Cell Endocrinol 2014; 382:695-723. [PMID: 23933149 DOI: 10.1016/j.mce.2013.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 12/29/2022]
Abstract
Endocrine therapy forms a central modality in the treatment of estrogen receptor positive breast cancer. The routine use of 5 years of adjuvant tamoxifen has improved survival rates for early breast cancer, and more recently has evolved in the postmenopausal setting to include aromatase inhibitors. The optimal duration of adjuvant endocrine therapy remains an active area of clinical study with recent data supporting 10 years rather than 5 years of adjuvant tamoxifen. However, endocrine therapy is limited by the development of resistance, this can occur by a number of possible mechanisms and numerous studies have been performed which combine endocrine therapy with agents that modulate these mechanisms with the aim of preventing or delaying the emergence of resistance. Recent trial data regarding the combination of the mammalian target of rapamycin (mTOR) inhibitor, everolimus with endocrine therapy have resulted in a redefinition of the clinical treatment pathway in the metastatic setting. This review details the current endocrine therapy utilized in both early and advanced disease, as well as exploring potential new targets which modulate pathways of resistance, as well as agents which aim to modulate adrenal derived steroidogenic hormones.
Collapse
Affiliation(s)
- Carlo Palmieri
- The University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, Liverpool L69 3GA, UK; Liverpool & Merseyside Breast Academic Unit, The Linda McCartney Centre, Royal Liverpool University Hospital, Liverpool L7 8XP, UK; Academic Department of Medical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Wiral CH63 4JY, UK.
| | - Darren K Patten
- Department of Surgery, Imperial College Healthcare NHS Trust, Fulham Palace Road, London W6 8RF, UK
| | - Adam Januszewski
- Department of Medical Oncology, Imperial College Healthcare NHS Trust, Fulham Palace Road, London W6 8RF, UK
| | - Giorgia Zucchini
- The University of Manchester, Institute of Cancer Studies, Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Sacha J Howell
- The University of Manchester, Institute of Cancer Studies, Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
50
|
Singh P, Alex JM, Bast F. Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: novel treatment strategies for cancer. Med Oncol 2013; 31:805. [PMID: 24338270 DOI: 10.1007/s12032-013-0805-3] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/04/2013] [Indexed: 02/07/2023]
Abstract
Insulin and insulin-like growth factor (IGF) signaling system, commonly known for fine-tuning numerous biological processes, has lately made its mark as a much sought-after therapeutic targets for diabetes and cancer. These receptors make an attractive anticancer target owing to their overexpression in variety of cancer especially in prostate and breast cancer. Inhibitors of IGF signaling were subjected to clinical cancer trials with the main objective to confirm the effectiveness of these receptors as a therapeutic target. However, the results that these trials produced proved to be disappointing as the role played by the cross talk between IGF and insulin receptor (IR) signaling pathways at the receptor level or at downstream signaling level became more lucid. Therapeutic strategy for IGF-1R and IR inhibition mainly encompasses three main approaches namely receptor blockade with monoclonal antibodies, tyrosine kinase inhibition (ATP antagonist and non-ATP antagonist), and ligand neutralization via monoclonal antibodies targeted to ligand or recombinant IGF-binding proteins. Other drug-discovery approaches are employed to target IGF-1R, and IR includes antisense oligonucleotides and recombinant IGF-binding proteins. However, therapies with monoclonal antibodies and tyrosine kinase inhibition targeting the IGF-1R are not evidenced to be satisfactory as expected. Factors that are duly held responsible for the unsuccessfulness of these therapies include (a) the existence of the IR isoform A overexpressed on a variety of cancers, enhancing the mitogenic signals to the nucleus leading to the endorsement of cell growth, (b) IGF-1R and IR that form hybrid receptors sensitive to the stimulation of all three IGF axis ligands, and (c) IGF-1R and IR that also have the potential to form hybrid receptors with other tyrosine kinase to potentiate the cellular transformation, tumorigenesis, and tumor vascularization. This mini review is a concerted effort to explore and fathom the well-recognized roles of the IRA signaling system in human cancer phenotype and the main strategies that have been so far evaluated to target the IR and IGF-1R.
Collapse
Affiliation(s)
- Pushpendra Singh
- Centre for Biosciences, School of Basic and Applied Science, Central University of Punjab, Bathinda, 151001, Punjab, India
| | | | | |
Collapse
|