1
|
Zhou Y, He Z, Li T, Choppavarapu L, Hu X, Cao R, Leone GW, Kahn M, Jin VX. 3D Chromatin Alteration by Disrupting β-Catenin/CBP Interaction Is Enriched with Insulin Signaling in Pancreatic Cancer. Cancers (Basel) 2024; 16:2202. [PMID: 38927910 PMCID: PMC11201718 DOI: 10.3390/cancers16122202] [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: 03/28/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
The therapeutic potential of targeting the β-catenin/CBP interaction has been demonstrated in a variety of preclinical tumor models with a small molecule inhibitor, ICG-001, characterized as a β-catenin/CBP antagonist. Despite the high binding specificity of ICG-001 for the N-terminus of CBP, this β-catenin/CBP antagonist exhibits pleiotropic effects. Our recent studies found global changes in three-dimensional (3D) chromatin architecture in response to disruption of the β-catenin/CBP interaction in pancreatic cancer cells. However, an understanding of how the functional crosstalk between the antagonist and the β-catenin/CBP interaction affects changes in 3D chromatin architecture and, thereby, gene expression and downstream effects remains to be elucidated. Here, we perform Hi-C analyses on canonical and patient-derived pancreatic cancer cells before and after treatment with ICG-001. In addition to global alteration of 3D chromatin domains, we unexpectedly identify insulin signaling genes enriched in the altered chromatin domains. We further demonstrate that the chromatin loops associated with insulin signaling genes are significantly weakened after ICG-001 treatment. We finally elicit the deletion of a looping of IRS1-a key insulin signaling gene-significantly impeding pancreatic cancer cell growth, indicating that looping-mediated insulin signaling might act as an oncogenic pathway to promote pancreatic cancer progression. Our work shows that targeting aberrant insulin chromatin looping in pancreatic cancer might provide a therapeutic benefit.
Collapse
Affiliation(s)
- Yufan Zhou
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Y.Z.); (Z.H.); (T.L.)
| | - Zhijing He
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Y.Z.); (Z.H.); (T.L.)
- Department of Stomatology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Tian Li
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Y.Z.); (Z.H.); (T.L.)
| | - Lavanya Choppavarapu
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Xiaohui Hu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China;
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ 08854, USA;
| | - Gustavo W. Leone
- MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael Kahn
- Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
| | - Victor X. Jin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
2
|
Zhou Y, Li T, He Z, Choppavarapu L, Hu X, Cao R, Leone GW, Kahn M, Jin VX. Reprogramming of 3D chromatin domains by antagonizing the β-catenin/CBP interaction attenuates insulin signaling in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566585. [PMID: 38013997 PMCID: PMC10680786 DOI: 10.1101/2023.11.10.566585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The therapeutic potential of targeting the β-catenin/CBP interaction has been demonstrated in a variety of preclinical tumor models with a small molecule inhibitor, ICG-001, characterized as a β-catenin/CBP antagonist. Despite the high binding specificity of ICG-001 for the N-terminus of CBP, this β-catenin/CBP antagonist exhibits pleiotropic effects. Our recent studies found global changes in three-dimensional (3D) chromatin architecture in response to disruption of the β-catenin/CBP interaction in pancreatic cancer cells. However, an understanding of the functional crosstalk between antagonizing the β-catenin/CBP interaction effect changes in 3D chromatin architecture and thereby gene expression and downstream effects remains to be elucidated. Here we perform Hi-C analyses on canonical and patient-derived pancreatic cancer cells before and after the treatment with ICG-001. In addition to global alteration of 3D chromatin domains, we unexpectedly identify insulin signaling genes enriched in the altered chromatin domains. We further demonstrate the chromatin loops associated with insulin signaling genes are significantly weakened after ICG-001 treatment. We finally elicit the deletion of a looping of IRS1, a key insulin signaling gene, significantly impede pancreatic cancer cell growth, indicating that looping-mediated insulin signaling might act as an oncogenic pathway to promote pancreatic cancer progression. Our work shows that targeting aberrant insulin chromatin looping in pancreatic cancer might provide a therapeutic benefit.
Collapse
|
3
|
Carcaboso AM. Results from the Children's Oncology Group phase III trial of a monoclonal antibody against the insulin-like growth factor-1 receptor in patients with newly diagnosed metastatic Ewing sarcoma. Transl Pediatr 2023; 12:1916-1919. [PMID: 37969125 PMCID: PMC10644028 DOI: 10.21037/tp-23-388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/14/2023] [Indexed: 11/17/2023] Open
Affiliation(s)
- Angel M. Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Pediatric Cancer Program, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| |
Collapse
|
4
|
Schmid P, Cortes J, Joaquim A, Jañez NM, Morales S, Díaz-Redondo T, Blau S, Neven P, Lemieux J, García-Sáenz JÁ, Hart L, Biyukov T, Baktash N, Massey D, Burris HA, Rugo HS. XENERA-1: a randomised double-blind Phase II trial of xentuzumab in combination with everolimus and exemestane versus everolimus and exemestane in patients with hormone receptor-positive/HER2-negative metastatic breast cancer and non-visceral disease. Breast Cancer Res 2023; 25:67. [PMID: 37308971 DOI: 10.1186/s13058-023-01649-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/20/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Xentuzumab is a humanised monoclonal antibody that binds to IGF-1 and IGF-2, neutralising their proliferative activity and restoring inhibition of AKT by everolimus. This study evaluated the addition of xentuzumab to everolimus and exemestane in patients with advanced breast cancer with non-visceral disease. METHODS This double-blind, randomised, Phase II study was undertaken in female patients with hormone-receptor (HR)-positive/human epidermal growth factor 2 (HER2)-negative advanced breast cancer with non-visceral disease who had received prior endocrine therapy with or without CDK4/6 inhibitors. Patients received a weekly intravenous infusion of xentuzumab (1000 mg) or placebo in combination with everolimus (10 mg/day orally) and exemestane (25 mg/day orally). The primary endpoint was progression-free survival (PFS) per independent review. RESULTS A total of 103 patients were randomised and 101 were treated (n = 50 in the xentuzumab arm and n = 51 in the placebo arm). The trial was unblinded early due to high rates of discordance between independent and investigator assessment of PFS. Per independent assessment, median PFS was 12.7 (95% CI 6.8-29.3) months with xentuzumab and 11.0 (7.7-19.5) months with placebo (hazard ratio 1.19; 95% CI 0.55-2.59; p = 0.6534). Per investigator assessment, median PFS was 7.4 (6.8-9.7) months with xentuzumab and 9.2 (5.6-14.4) months with placebo (hazard ratio 1.23; 95% CI 0.69-2.20; p = 0.4800). Tolerability was similar between the arms, with diarrhoea (33.3-56.0%), fatigue (33.3-44.0%) and headache (21.6-40.0%) being the most common treatment-emergent adverse events. The incidence of grade ≥ 3 hyperglycaemia was similar between the xentuzumab (2.0%) and placebo (5.9%) arms. CONCLUSIONS While this study demonstrated that xentuzumab could be safely combined with everolimus and exemestane in patients with HR-positive/HER2-negative advanced breast cancer with non-visceral disease, there was no PFS benefit with the addition of xentuzumab. Trial registration ClinicalTrials.gov, NCT03659136. Prospectively registered, September 6, 2018.
Collapse
Affiliation(s)
- Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - Ana Joaquim
- Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | | | | | - Tamara Díaz-Redondo
- Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Unidad de Gestión Clínica Intercentros de Oncología, Málaga, Spain
| | - Sibel Blau
- Northwest Medical Specialties, Tacoma, WA, USA
| | | | - Julie Lemieux
- Centre Hospitalier Universitaire de Québec-Université Laval Research Centre, Quebec, Canada
| | | | - Lowell Hart
- Florida Cancer Specialists, Fort Myers, FL, USA
| | | | - Navid Baktash
- Boehringer Ingelheim (Canada) Ltd, Burlington, ON, Canada
| | - Dan Massey
- Elderbrook Solutions GmbH on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Hope S Rugo
- University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Shackleford TJ, Hariharan S, Vaseva AV, Alagoa K, Espinoza M, Bid HK, Li F, Zhong H, Phelps DA, Roberts RD, Cam H, London CA, Guttridge DC, Chen Y, Rao M, Shiio Y, Houghton PJ. Redundant Signaling as the Predominant Mechanism for Resistance to Antibodies Targeting the Type-I Insulin-Like Growth Factor Receptor in Cells Derived from Childhood Sarcoma. Mol Cancer Ther 2023; 22:539-550. [PMID: 36696581 PMCID: PMC10073271 DOI: 10.1158/1535-7163.mct-20-0625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 07/12/2021] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Antibodies targeting insulin-like growth factor 1 receptor (IGF-1R) induce objective responses in only 5% to 15% of children with sarcoma. Understanding the mechanisms of resistance may identify combination therapies that optimize efficacy of IGF-1R-targeted antibodies. Sensitivity to the IGF-1R-targeting antibody TZ-1 was determined in rhabdomyosarcoma and Ewing sarcoma cell lines. Acquired resistance to TZ-1 was developed and characterized in sensitive Rh41 cells. The BRD4 inhibitor, JQ1, was evaluated as an agent to prevent acquired TZ-1 resistance in Rh41 cells. The phosphorylation status of receptor tyrosine kinases (RTK) was assessed. Sensitivity to TZ-1 in vivo was determined in Rh41 parental and TZ-1-resistant xenografts. Of 20 sarcoma cell lines, only Rh41 was sensitive to TZ-1. Cells intrinsically resistant to TZ-1 expressed multiple (>10) activated RTKs or a relatively less complex set of activated RTKs (∼5). TZ-1 decreased the phosphorylation of IGF-1R but had little effect on other phosphorylated RTKs in all resistant lines. TZ-1 rapidly induced activation of RTKs in Rh41 that was partially abrogated by knockdown of SOX18 and JQ1. Rh41/TZ-1 cells selected for acquired resistance to TZ-1 constitutively expressed multiple activated RTKs. TZ-1 treatment caused complete regressions in Rh41 xenografts and was significantly less effective against the Rh41/TZ-1 xenograft. Intrinsic resistance is a consequence of redundant signaling in pediatric sarcoma cell lines. Acquired resistance in Rh41 cells is associated with rapid induction of multiple RTKs, indicating a dynamic response to IGF-1R blockade and rapid development of resistance. The TZ-1 antibody had greater antitumor activity against Rh41 xenografts compared with other IGF-1R-targeted antibodies tested against this model.
Collapse
Affiliation(s)
- Terry J. Shackleford
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
- Saint Mary’s University, San Antonio, TX
| | | | - Angelina V. Vaseva
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| | | | | | - Hemant K. Bid
- Resonant Therapeutics, Inc. Life Sciences Institute (LSI) University of Michigan
| | - Fuyang Li
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| | | | - Doris A. Phelps
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| | | | - Hakan Cam
- Nationwide Children’s Hospital, Columbus, OH
| | - Cheryl A. London
- Cummings School of Veterinary Medicine, Tufts University, Boston
| | - Denis C. Guttridge
- Darby Children’s Research Institute, Medical College of South Carolina, Charleston
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| | - Manjeet Rao
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| | - Yuzuru Shiio
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| | - Peter J. Houghton
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX
| |
Collapse
|
6
|
Galambus J, Tsai KY. Molecular and immune targets in cutaneous squamous cell carcinoma. Mol Carcinog 2023; 62:38-51. [PMID: 36000298 DOI: 10.1002/mc.23451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/23/2022] [Accepted: 07/05/2022] [Indexed: 02/03/2023]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer and often confers a good prognosis. Though surgery is the gold standard of treatment, unresectable or metastatic disease can necessitate systemic therapy. Of systemic agents, there is increasing interest in the use of immunotherapies and targeted therapy. Further study into the driver mutations in cSCC has identified opportunities for targeted therapy. In this review, we discuss both current and investigational immune and molecular targets of therapy for cSCC.
Collapse
Affiliation(s)
- Justine Galambus
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Kenneth Y Tsai
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Donald A. Adam Melanoma and Skin Cancer Center of Excellence, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
7
|
Werner H, LeRoith D. Hallmarks of cancer: The insulin-like growth factors perspective. Front Oncol 2022; 12:1055589. [PMID: 36479090 PMCID: PMC9720135 DOI: 10.3389/fonc.2022.1055589] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/07/2022] [Indexed: 08/30/2023] Open
Abstract
The identification of a series of attributes or hallmarks that are shared by virtually all cancer cells constitutes a true milestone in cancer research. The conceptualization of a catalogue of common genetic, molecular, biochemical and cellular events under a unifying Hallmarks of Cancer idea had a major impact in oncology. Furthermore, the fact that different types of cancer, ranging from pediatric tumors and leukemias to adult epithelial cancers, share a large number of fundamental traits reflects the universal nature of the biological events involved in oncogenesis. The dissection of a complex disease like cancer into a finite directory of hallmarks is of major basic and translational relevance. The role of insulin-like growth factor-1 (IGF1) as a progression/survival factor required for normal cell cycle transition has been firmly established. Similarly well characterized are the biochemical and cellular activities of IGF1 and IGF2 in the chain of events leading from a phenotypically normal cell to a diseased one harboring neoplastic traits, including growth factor independence, loss of cell-cell contact inhibition, chromosomal abnormalities, accumulation of mutations, activation of oncogenes, etc. The purpose of the present review is to provide an in-depth evaluation of the biology of IGF1 at the light of paradigms that emerge from analysis of cancer hallmarks. Given the fact that the IGF1 axis emerged in recent years as a promising therapeutic target, we believe that a careful exploration of this signaling system might be of critical importance on our ability to design and optimize cancer therapies.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
8
|
Halisah H, Nur A, Taslim NA, Chalid SM, Juliaty A, Mappaware NA, Ahmad M, Hamid F, Alasiry E, Aminuddin A, Thalib KU, Hermanses SS, Fasiha F, Achmad IH, Mustardianto M. The Effect of Family Approach Model Education and Zinc Supplementation on Pregnant Women with Chronic Energy Lack from Poor Family. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Pregnant women’s health and nutritional status must be maintained because they are indicators of perinatal growth and neonatal welfare; efforts to prevent fetal growth disorders must begin with pregnant women, one of which is through increased knowledge and nutritional supplementation.
AIM: This study aimed to examine the effect of family-based education and zinc supplementation on knowledge, weight, upper arm circumference, nutritional intake, serum zinc levels, insulin-like growth factor-1 (IGF-1), hemoglobin, infant anthropometry, and placenta in stunted pregnant women.
MATERIALS AND METHODS: Quasi-experiment with a pre-post-test control design was carried out from September 2020 to June 1, 2021, in four areas of the Mamuju Regency City Health Center. The subject population is pregnant women aged 20–26 weeks gestation, experiencing chronic energy deficiency.
RESULTS: The mean knowledge increased in the intervention group with a rate of change of 133.57%, while the control group did not experience a significant increase. Unpaired t-test showed a significant difference in the mean serum zinc, IGF-1, and hemoglobin levels with p < 0.001.
CONCLUSION: Zinc supplementation is very beneficial for pregnant women, especially teenage pregnant women who have chronic energy deficiency, but the results of zinc supplementation are better accompanied by providing education based on a family approach, as a form of community support system for mothers, because family diet has a strong impact on pregnant women diet.
Collapse
|
9
|
Sair AT, Liu RH. Molecular regulation of phenolic compounds on IGF-1 signaling cascade in breast cancer. Food Funct 2022; 13:3170-3184. [PMID: 35253808 DOI: 10.1039/d1fo03283f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Breast cancer is a highly aggressive and heterogeneous disease with complex features that remains a major health problem and undermines the span and quality of life of women worldwide. Primary literature has shown the role of phenolic compounds in controlling the onset of breast cancer. The mechanism of action of phenolic compounds can be explained by their interaction with signal transduction pathways that regulate cell proliferation and induction of apoptosis. One of the targets of phenolic compounds is the insulin like growth factor 1 (IGF-1) signaling cascade, which plays a significant role in the growth and development of mammary tissues by leading proliferative and anti-apoptotic events. Increasing research evidence points to the function of the IGF-1 cascade system in the commencement, progression, and metastasis of breast tissue malignancy. In this review, we mainly discuss the function of the IGF-1 system, and the role of phenolic compounds in regulating the IGF-1 signaling cascade and curbing breast malignancies.
Collapse
Affiliation(s)
- Ali Tahir Sair
- Department of Food Science, Cornell University, 245 Stocking Hall, Ithaca, New York 14853, USA.
| | - Rui Hai Liu
- Department of Food Science, Cornell University, 245 Stocking Hall, Ithaca, New York 14853, USA.
| |
Collapse
|
10
|
Boichuk S, Dunaev P, Mustafin I, Mani S, Syuzov K, Valeeva E, Bikinieva F, Galembikova A. Infigratinib (BGJ 398), a Pan-FGFR Inhibitor, Targets P-Glycoprotein and Increases Chemotherapeutic-Induced Mortality of Multidrug-Resistant Tumor Cells. Biomedicines 2022; 10:biomedicines10030601. [PMID: 35327403 PMCID: PMC8945560 DOI: 10.3390/biomedicines10030601] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
The microtubule-targeting agents (MTAs) are well-known chemotherapeutic agents commonly used for therapy of a broad spectrum of human malignancies, exhibiting epithelial origin, including breast, lung, and prostate cancer. Despite the impressive response rates shortly after initiation of MTA-based therapy, the vast majority of human malignancies develop resistance to MTAs due to the different mechanisms. Here, we report that infigratinib (BGJ 398), a potent FGFR1-4 inhibitor, restores sensitivity of a broad spectrum of ABCB1-overexpressing cancer cells to certain chemotherapeutic agents, including paclitaxel (PTX) and doxorubicin (Dox). This was evidenced for the triple-negative breast cancer (TNBC), and gastrointestinal stromal tumor (GIST) cell lines, as well. Indeed, when MDR-overexpressing cancer cells were treated with a combination of BGJ 398 and PTX (or Dox), we observed a significant increase of apoptosis which was evidenced by an increased expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin V-positive cells, as well. Moreover, BGJ 398 used in combination with PTX significantly decreased the viability and proliferation of the resistant cancer cells. As expected, no apoptosis was found in ABCB1-overexpressing cancer cells treated with PTX, Dox, or BGJ 398 alone. Inhibition of FGFR-signaling by BGJ 398 was evidenced by the decreased expression of phosphorylated (i.e., activated) forms of FGFR and FRS-2, a well-known adaptor protein of FGFR signaling, and downstream signaling molecules (e.g., STAT-1, -3, and S6). In contrast, expression of MDR-related ABC-transporters did not change after BGJ 398 treatment, thereby suggesting an impaired function of MDR-related ABC-transporters. By using the fluorescent-labeled chemotherapeutic agent PTX-Alexa488 (Flutax-2) and doxorubicin, exhibiting an intrinsic fluorescence, we found that BGJ 398 substantially impairs their efflux from MDR-overexpressing TNBC cells. Moreover, the efflux of Calcein AM, a well-known substrate for ABCB1, was also significantly impaired in BGJ 398-treated cancer cells, thereby suggesting the ABCB1 as a novel molecular target for BGJ 398. Of note, PD 173074, a potent FGFR1 and VEGFR2 inhibitor failed to retain chemotherapeutic agents inside ABCB1-overexpressing cells. This was consistent with the inability of PD 173074 to sensitize Tx-R cancer cells to PTX and Dox. Collectively, we show here for the first time that BGJ 398 reverses the sensitivity of MDR-overexpressing cancer cells to certain chemotherapeutic agents due to inhibition of their efflux from cancer cells via ABCB1-mediated mechanism.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
- Сentral Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia;
- Department of Radiotherapy and Radiology, Faculty of Surgery, Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
- Correspondence: ; Tel.: +7-917-397-80-93; Fax: +7-843-236-06-52
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Ilshat Mustafin
- Department of Biochemistry, Kazan State Medical University, 420012 Kazan, Russia;
| | - Shinjit Mani
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Kirill Syuzov
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Elena Valeeva
- Сentral Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia;
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| |
Collapse
|
11
|
Douglas RS, Kahaly GJ, Ugradar S, Elflein H, Ponto KA, Fowler BT, Dailey R, Harris GJ, Schiffman J, Tang R, Wester S, Jain AP, Marcocci C, Marinò M, Antonelli A, Eckstein A, Führer-Sakel D, Salvi M, Sile S, Francis-Sedlak M, Holt RJ, Smith TJ. Teprotumumab Efficacy, Safety and Durability in Longer Duration Thyroid Eye Disease and Retreatment: Optic-X Study. Ophthalmology 2021; 129:438-449. [PMID: 34688699 DOI: 10.1016/j.ophtha.2021.10.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Evaluate teprotumumab safety and efficacy in patients with thyroid eye disease (TED) who previously did not respond or who had a disease flare. DESIGN OPTIC-X is an open-label (previous treatment masked) teprotumumab treatment and retreatment trial in patients from the randomized double-masked, multicenter, placebo-controlled OPTIC study. PARTICIPANTS OPTIC study patients who previously received placebo, 37 patients, or who previously received teprotumumab, 14 patients. INTERVENTION OPTIC non-responders and those who flared (≥2mm increase in proptosis, ≥2point increase in clinical activity score [CAS], or both) during follow-up were treated for the first time (previous placebo patients) or retreated with teprotumumab in OPTIC-X with 8 infusions over 24-weeks. MAIN OUTCOME MEASURES Proptosis responder rate and safety were examined. Secondary outcomes included proptosis, CAS, subjective diplopia, and quality of life responses. RESULTS Thirty-three of 37 (89.2%) placebo-treated OPTIC patients became proptosis responders (mean [standard deviation] -3.5mm [1.7]) when treated with teprotumumab in OPTIC-X. The magnitude of responses was equivalent to those in the OPTIC study. In these responders, proptosis, CAS 0 or 1, and diplopia responses were maintained in 29/32 (90.6%), 20/21 (95.2%), and 12/14 (85.7%), respectively, at week-48 of follow up. These patients had a median TED duration of 12.9 months versus 6.3 months in those treated with teprotumumab in the OPTIC study. Of the 5 OPTIC teprotumumab non-responders retreated in the OPTIC-X study, 2 responded, 1 had a proptosis reduction of 1.5mm from OPTIC baseline and 2 discontinued treatment early. Of the OPTIC teprotumumab responders who flared, 5/8 (62.5%) became responders when retreated (mean proptosis reduction of 1.9mm [1.2] from OPTIC-X baseline, 3.3mm [0.7] from OPTIC baseline). Compared to published double-masked trials and their integrated follow-up, no new safety signals were identified. Mild hearing impairment was reported with 4 events occurring during the first course of treatment and 2 events reoccurring following retreatment. CONCLUSION These data indicate that TED patients with longer disease duration respond similarly to those treated earlier in their disease. Patients with an insufficient initial response or flare may benefit from additional teprotumumab therapy. This analysis did not find any new safety risk; however additional post-marketing pharmacovigilance is ongoing.
Collapse
Affiliation(s)
| | - George J Kahaly
- Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
| | - Shoaib Ugradar
- The Jules Stein Eye Institute, UCLA, Los Angeles, California, USA
| | - Heike Elflein
- Department of Ophthalmology, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
| | - Katharina A Ponto
- Department of Ophthalmology and Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
| | - Brian T Fowler
- University of Tennessee, Health Science Center, Memphis, Tennessee, USA
| | - Roger Dailey
- Casey Eye Institute, Oregon Health & Sciences University, Portland, Oregon, USA
| | - Gerald J Harris
- The Medical College of Wisconsin Eye Institute, Milwaukee, Wisconsin, USA
| | - Jade Schiffman
- Eye Wellness Center- Neuro-Eye Clinical Trials, Inc., Houston, Texas, USA
| | - Rosa Tang
- Eye Wellness Center- Neuro-Eye Clinical Trials, Inc., Houston, Texas, USA
| | - Sara Wester
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Amy Patel Jain
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Claudio Marcocci
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University Hospital of Pisa, Pisa, Italy
| | - Michele Marinò
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University Hospital of Pisa, Pisa, Italy
| | - Alessandro Antonelli
- Department of Surgical, Medical and Molecular Pathology and Critical Care, University of Pisa, Pisa, Italy
| | - Anja Eckstein
- Department of Ophthalmology, EUGOGO Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dagmar Führer-Sakel
- Department of Endocrinology, Diabetes and Metabolism, EUGOGO Center Essen, University Hospital Essen, University of Duisburg-Essen
| | - Mario Salvi
- Endocrinology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Saba Sile
- Horizon Therapeutics plc, Deerfield, Illinois, USA
| | | | | | - Terry J Smith
- Department of Ophthalmology and Visual Sciences and Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Wittlinger F, Laufer SA. The pre-clinical discovery and development of osimertinib used to treat non-small cell lung cancer. Expert Opin Drug Discov 2021; 16:1091-1103. [PMID: 34053372 DOI: 10.1080/17460441.2021.1936496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Osimertinib is currently the only FDA- and EMA-approved third-generation small-molecule epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI). It was initially indicated for second-line treatment of patients with metastatic EGFR T790M mutation-positive non-small cell lung cancer (NSCLC) and got approved for first-line treatment of EGFR activation mutation-positive metastatic NSCLC in 2018. Most recently, the FDA granted approval for the adjuvant treatment of patients with early-stage mutated EGFR NSCLC after tumor resection.Areas covered: This drug discovery case history focuses on the key studies that led to the preclinical discovery and development of osimertinib. The authors focus on published preclinical studies by scientists from AstraZeneca and highlight key events in the clinical development.Expert opinion: Although eventually compromised by the cellular plasticity of the tumor and the inevitable acquisition of drug resistance through the use of osimertinib, its key role in the treatment of NSCLC with specific EGFR mutations will be maintained in the near future. As the genome of EGFR is highly labile and since the rapid development of new mutants remains an issue, there is still room for improvement for the next generation of inhibitors.
Collapse
Affiliation(s)
- Florian Wittlinger
- Tuebingen Center for Academic Drug Discovery & Development, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Stefan A Laufer
- Tuebingen Center for Academic Drug Discovery & Development, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
13
|
IGF1-mediated HOXA13 overexpression promotes colorectal cancer metastasis through upregulating ACLY and IGF1R. Cell Death Dis 2021; 12:564. [PMID: 34075028 PMCID: PMC8169856 DOI: 10.1038/s41419-021-03833-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022]
Abstract
Metastasis is the major reason for the high mortality of colorectal cancer (CRC) patients and its molecular mechanism remains unclear. Here, we report a novel role of Homeobox A13 (HOXA13), a member of the Homeobox (HOX) family, in promoting CRC metastasis. The elevated expression of HOXA13 was positively correlated with distant metastasis, higher AJCC stage, and poor prognosis in two independent CRC cohorts. Overexpression of HOXA13 promoted CRC metastasis whereas downregulation of HOXA13 suppressed CRC metastasis. Mechanistically, HOXA13 facilitated CRC metastasis by transactivating ATP-citrate lyase (ACLY) and insulin-like growth factor 1 receptor (IGF1R). Knockdown of ACLY and IGFIR inhibited HOXA13-medicated CRC metastasis, whereas ectopic overexpression of ACLY and IGFIR rescued the decreased CRC metastasis induced by HOXA13 knockdown. Furthermore, Insulin-like growth factor 1 (IGF1), the ligand of IGF1R, upregulated HOXA13 expression through the PI3K/AKT/HIF1α pathway. Knockdown of HOXA13 decreased IGF1-mediated CRC metastasis. In addition, the combined treatment of ACLY inhibitor ETC-1002 and IGF1R inhibitor Linsitinib dramatically suppressed HOXA13-mediated CRC metastasis. In conclusion, HOXA13 is a prognostic biomarker in CRC patients. Targeting the IGF1-HOXA13-IGF1R positive feedback loop may provide a potential therapeutic strategy for the treatment of HOXA13-driven CRC metastasis.
Collapse
|
14
|
LeRoith D, Holly JMP, Forbes BE. Insulin-like growth factors: Ligands, binding proteins, and receptors. Mol Metab 2021; 52:101245. [PMID: 33962049 PMCID: PMC8513159 DOI: 10.1016/j.molmet.2021.101245] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/09/2021] [Accepted: 04/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The insulin-like growth factor family of ligands (IGF-I, IGF-II, and insulin), receptors (IGF-IR, M6P/IGF-IIR, and insulin receptor [IR]), and IGF-binding proteins (IGFBP-1-6) play critical roles in normal human physiology and disease states. SCOPE OF REVIEW Insulin and insulin receptors are the focus of other chapters in this series and will therefore not be discussed further. Here we review the basic components of the IGF system, their role in normal physiology and in critical pathology's. While this review concentrates on the role of IGFs in human physiology, animal models have been essential in providing understanding of the IGF system, and its regulation, and are briefly described. MAJOR CONCLUSIONS IGF-I has effects via the circulation and locally within tissues to regulate cellular growth, differentiation, and survival, thereby controlling overall body growth. IGF-II levels are highest prenatally when it has important effects on growth. In adults, IGF-II plays important tissue-specific roles, including the maintenance of stem cell populations. Although the IGF-IR is closely related to the IR it has distinct physiological roles both on the cell surface and in the nucleus. The M6P/IGF-IIR, in contrast, is distinct and acts as a scavenger by mediating internalization and degradation of IGF-II. The IGFBPs bind IGF-I and IGF-II in the circulation to prolong their half-lives and modulate tissue access, thereby controlling IGF function. IGFBPs also have IGF ligand-independent cell effects.
Collapse
Affiliation(s)
- Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeff M P Holly
- Translational Health Sciences, Bristol Medical School, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Briony E Forbes
- Discipline of Medical Biochemistry, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, Australia
| |
Collapse
|
15
|
Rosendahl AH, Björner S, Ygland Rödström M, Jirström K, Borgquist S, Ingvar C, Pollak MN, Jernström H. Pre- and Postoperative Circulating IGF-I, IGFBP-3, and IGFBP-7 Levels in Relation to Endocrine Treatment and Breast Cancer Recurrence: A Nested Case-Control Study. Front Oncol 2021; 11:626058. [PMID: 33767994 PMCID: PMC7986849 DOI: 10.3389/fonc.2021.626058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/25/2021] [Indexed: 01/18/2023] Open
Abstract
Insulin-like growth factor-I (IGF-I) and its binding proteins (BPs) have been associated with breast cancer risk, especially high IGF-I concentrations and the biologically active fraction estimated as the IGF-I/IGFBP-3 molar ratio. The relation of circulating IGF-I and IGFBP-3 concentrations with risk of breast cancer recurrence has been less documented. In addition a new member to a sub-group of the IGFBP-superfamily was recently identified, the low affinity IGFBP-7. To date, the role of systemic IGFBP-7 in breast cancer progression has not been investigated. Our purpose was to establish whether circulating IGF-I, IGFBP-3, and IGFBP-7 levels are related to recurrence-risk in breast cancer. A case-control study was nested within the population-based BCBlood cohort of 853 breast cancer patients diagnosed 2002-2010 in Sweden and followed through 2012. In total, 95 patients with recurrence and 170 controls were matched on age and tumor characteristics. Plasma IGF analytes and tumor membrane IGF-I receptor (IGF-IRm) positivity were analyzed and recurrence-risk was evaluated with conditional logistic regression. Preoperative tertiles of IGF-I and IGFBP-3 were both positively associated with recurrence-risk, but not IGFBP-7. The trend was of borderline significance for IGF-I, T1:REF, T2 OR:1.6, T3 OR: 2.2 adjusted P trend=0.057 and significant for IGFBP-3 T1:REF, T2 OR:1.2, T3 OR: 2.1 adjusted P trend=0.042. The models were adjusted for age, anthropometric factors, smoking, and treatments. There was a significant interaction between IGFBP-7 and IGF-IRm positivity on recurrence, where the highest IGFBP-7 highest IGFBP-7 tertile conferred increased recurrence-risk in patients with IGF-IRm positive tumors but not in those with IGF-IRm negative tumors (P interaction=0.024). By the 1-year visit, age-adjusted IGF-I levels were reduced by 17% while IGFBP-3 and IGFBP-7 were stable. IGF-I levels were significantly reduced by radiotherapy in all patients and by tamoxifen in patients with ER+ tumors. Postoperative changes >10% (n=208) in IGF-I, IGFBP-3, IGFBP-7, or the IGF-I/IGFBP-3 ratio did not predict recurrence after adjustment for preoperative levels, age, anthropometric factors, smoking, and treatments. In conclusion, this study suggests that preoperative IGF-I and IGFBP-3 levels, but not postoperative changes, might provide independent prognostic information and influence breast cancer recurrence. The role of IGFBP-7 in breast cancer merits further study.
Collapse
Affiliation(s)
- Ann H Rosendahl
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Sofie Björner
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Maria Ygland Rödström
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Oncology and Therapeutic Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden.,Department of Oncology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Christian Ingvar
- Department of Clinical Sciences Lund, Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Michael N Pollak
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Oncology McGill University, Montreal, QC, Canada
| | - Helena Jernström
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| |
Collapse
|
16
|
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: 17] [Impact Index Per Article: 4.3] [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
|
17
|
Vella V, De Francesco EM, Lappano R, Muoio MG, Manzella L, Maggiolini M, Belfiore A. Microenvironmental Determinants of Breast Cancer Metastasis: Focus on the Crucial Interplay Between Estrogen and Insulin/Insulin-Like Growth Factor Signaling. Front Cell Dev Biol 2020; 8:608412. [PMID: 33364239 PMCID: PMC7753049 DOI: 10.3389/fcell.2020.608412] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
The development and progression of the great majority of breast cancers (BCs) are mainly dependent on the biological action elicited by estrogens through the classical estrogen receptor (ER), as well as the alternate receptor named G-protein–coupled estrogen receptor (GPER). In addition to estrogens, other hormones and growth factors, including the insulin and insulin-like growth factor system (IIGFs), play a role in BC. IIGFs cooperates with estrogen signaling to generate a multilevel cross-communication that ultimately facilitates the transition toward aggressive and life-threatening BC phenotypes. In this regard, the majority of BC deaths are correlated with the formation of metastatic lesions at distant sites. A thorough scrutiny of the biological and biochemical events orchestrating metastasis formation and dissemination has shown that virtually all cell types within the tumor microenvironment work closely with BC cells to seed cancerous units at distant sites. By establishing an intricate scheme of paracrine interactions that lead to the expression of genes involved in metastasis initiation, progression, and virulence, the cross-talk between BC cells and the surrounding microenvironmental components does dictate tumor fate and patients’ prognosis. Following (i) a description of the main microenvironmental events prompting BC metastases and (ii) a concise overview of estrogen and the IIGFs signaling and their major regulatory functions in BC, here we provide a comprehensive analysis of the most recent findings on the role of these transduction pathways toward metastatic dissemination. In particular, we focused our attention on the main microenvironmental targets of the estrogen-IIGFs interplay, and we recapitulated relevant molecular nodes that orientate shared biological responses fostering the metastatic program. On the basis of available studies, we propose that a functional cross-talk between estrogens and IIGFs, by affecting the BC microenvironment, may contribute to the metastatic process and may be regarded as a novel target for combination therapies aimed at preventing the metastatic evolution.
Collapse
Affiliation(s)
- Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Maria Grazia Muoio
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy.,Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Livia Manzella
- Center of Experimental Oncology and Hematology, Azienda Ospedaliera Universitaria (A.O.U.) Policlinico Vittorio Emanuele, Catania, Italy.,Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| |
Collapse
|
18
|
de Groot S, Röttgering B, Gelderblom H, Pijl H, Szuhai K, Kroep JR. Unraveling the Resistance of IGF-Pathway Inhibition in Ewing Sarcoma. Cancers (Basel) 2020; 12:cancers12123568. [PMID: 33260481 PMCID: PMC7759976 DOI: 10.3390/cancers12123568] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The insulin-like growth factor-1 receptor (IGF1R) is a receptor commonly overexpressed and overactivated in a variety of cancers, including Ewing sarcoma, and promotes cell growth and survival. After promising results with targeting and inhibiting the receptor in vitro, multiple different IGF1R targeting compounds have been clinically tried but showed limited efficacy. Here we discuss several possible resistance mechanisms which could explain why IGF1R targeting fails in the clinic and discuss possible ways to overcome these resistances. Abstract Insulin-like growth factor-1 receptor (IGF1R) inhibitors are effective in preclinical studies, but so far, no convincing benefit in clinical studies has been observed, except in some rare cases of sustained response in Ewing sarcoma patients. The mechanism of resistance is unknown, but several hypotheses are proposed. In this review, multiple possible mechanisms of resistance to IGF-targeted therapies are discussed, including activated insulin signaling, pituitary-driven feedback loops through growth hormone (GH) secretion and autocrine loops. Additionally, the outcomes of clinical trials of IGF1-targeted therapies are discussed, as well as strategies to overcome the possible resistance mechanisms. In conclusion, lowering the plasma insulin levels or blocking its activity could provide an additional target in cancer therapy in combination with IGF1 inhibition. Furthermore, because Ewing sarcoma cells predominantly express the insulin receptor A (IRA) and healthy tissue insulin receptor B (IRB), it may be possible to synthesize a specific IRA inhibitor.
Collapse
Affiliation(s)
- Stefanie de Groot
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.d.G.); (H.G.)
| | - Bas Röttgering
- Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.d.G.); (H.G.)
| | - Hanno Pijl
- Department of Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
| | - Karoly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
- Correspondence: (K.S.); (J.R.K.); Tel.: +31-715266922 (K.S.); +31-715263464 (J.R.K.)
| | - Judith R. Kroep
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.d.G.); (H.G.)
- Correspondence: (K.S.); (J.R.K.); Tel.: +31-715266922 (K.S.); +31-715263464 (J.R.K.)
| |
Collapse
|
19
|
Abstract
Elevated circulating insulin levels are frequently observed in the setting of obesity and early type 2 diabetes, as a result of insensitivity of metabolic tissues to the effects of insulin. Higher levels of circulating insulin have been associated with increased cancer risk and progression in epidemiology studies. Elevated circulating insulin is believed to be a major factor linking obesity, diabetes and cancer. With the development of targeted cancer therapies, insulin signalling has emerged as a mechanism of therapeutic resistance. Although metabolic tissues become insensitive to insulin in the setting of obesity, a number of mechanisms allow cancer cells to maintain their ability to respond to insulin. Significant progress has been made in the past decade in understanding the insulin receptor and its signalling pathways in cancer, and a number of lessons have been learnt from therapeutic failures. These discoveries have led to numerous clinical trials that have aimed to reduce the levels of circulating insulin and to abrogate insulin signalling in cancer cells. With the rising prevalence of obesity and diabetes worldwide, and the realization that hyperinsulinaemia may contribute to therapeutic failures, it is essential to understand how insulin and insulin receptor signalling promote cancer progression.
Collapse
Affiliation(s)
- Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
20
|
Marra A, Trapani D, Viale G, Criscitiello C, Curigliano G. Practical classification of triple-negative breast cancer: intratumoral heterogeneity, mechanisms of drug resistance, and novel therapies. NPJ Breast Cancer 2020; 6:54. [PMID: 33088912 PMCID: PMC7568552 DOI: 10.1038/s41523-020-00197-2] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is not a unique disease, encompassing multiple entities with marked histopathological, transcriptomic and genomic heterogeneity. Despite several efforts, transcriptomic and genomic classifications have remained merely theoretic and most of the patients are being treated with chemotherapy. Driver alterations in potentially targetable genes, including PIK3CA and AKT, have been identified across TNBC subtypes, prompting the implementation of biomarker-driven therapeutic approaches. However, biomarker-based treatments as well as immune checkpoint inhibitor-based immunotherapy have provided contrasting and limited results so far. Accordingly, a better characterization of the genomic and immune contexture underpinning TNBC, as well as the translation of the lessons learnt in the metastatic disease to the early setting would improve patients' outcomes. The application of multi-omics technologies, biocomputational algorithms, assays for minimal residual disease monitoring and novel clinical trial designs are strongly warranted to pave the way toward personalized anticancer treatment for patients with TNBC.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| |
Collapse
|
21
|
Huang G, Song C, Wang N, Qin T, Sui S, Obr A, Zeng L, Wood TL, Leroith D, Li M, Wu Y. RNA-binding protein CUGBP1 controls the differential INSR splicing in molecular subtypes of breast cancer cells and affects cell aggressiveness. Carcinogenesis 2020; 41:1294-1305. [PMID: 31958132 PMCID: PMC7513956 DOI: 10.1093/carcin/bgz141] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
The insulin receptor gene (INSR) undergoes alternative splicing to give rise to two functionally related, but also distinct, isoforms IR-A and IR-B, which dictate proliferative and metabolic regulations, respectively. Previous studies identified the RNA-binding protein CUGBP1 as a key regulator of INSR splicing. In this study, we show that the differential splicing of INSR occurs more frequently in breast cancer than in non-tumor breast tissues. In breast cancer cell lines, the IR-A:IR-B ratio varies in different molecular subtypes, knockdown or overexpression of CUGBP1 gene in breast cancer cells altered IR-A:IR-B ratio through modulation of IR-A expression, thereby reversed or enhanced the insulin-induced oncogenic behavior of breast cancer cells, respectively. Our data revealed the predominant mitogenic role of IR-A isoform in breast cancer and depicted a novel interplay between INSR and CUGBP1, implicating CUGBP1 and IR-A isoform as the potential therapeutic targets and biomarkers for breast cancer.
Collapse
Affiliation(s)
- Gena Huang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Chen Song
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Qin
- Department of Pathology, Dalian Medical University, Dalian, Liaoning, China
| | - Silei Sui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Alison Obr
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Li Zeng
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Derek Leroith
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
| | - Man Li
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
- College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
22
|
Kashyap S, Zeidler JD, Chini CCS, Chini EN. Implications of the PAPP-A-IGFBP-IGF-1 pathway in the pathogenesis and treatment of polycystic kidney disease. Cell Signal 2020; 73:109698. [PMID: 32569826 DOI: 10.1016/j.cellsig.2020.109698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common genetic diseases implicated in the development of end stage renal disease (ESRD). Although FDA has recently approved a drug against ADPKD, there is still a great need for development of alternative management strategies for ADPKD. Understanding the different mechanisms that lead to cystogenesis and cyst expansion in ADPKD is imperative to develop new therapies against ADPKD. Recently, we demonstrated that caloric restriction can prevent the development of cystic disease in animal models of ADPKD and through these studies identified a new role for pregnancy associated plasma protein-A (PAPP-A), a component of the insulin-like growth factors (IGF) pathway, in the pathogenesis of this disease. The PAPP-A-IGF pathway plays an important role in regulation of cell growth, differentiation, and transformation and dysregulation of this pathway has been implicated in many diseases. Several indirect studies support the involvement of IGF-1 in the pathogenesis of ADPKD. However, it was only recently that we described a direct role for a component of this pathway in pathogenesis of ADPKD, opening a new avenue for the therapeutic approaches for this cystic disease. The present literature review will critically discuss the evidence that supports the role of components of IGF pathway in the pathogenesis of ADPKD and discuss the pharmacological implications of PAPP-A-IGF axis in this disease.
Collapse
Affiliation(s)
- Sonu Kashyap
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Julianna D Zeidler
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Claudia C S Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Eduardo Nunes Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
23
|
Sinai-Livne T, Pasmanik-Chor M, Cohen Z, Tsarfaty I, Werner H, Berger R. Proteomic analysis of combined IGF1 receptor targeted therapy and chemotherapy identifies signatures associated with survival in breast cancer patients. Oncotarget 2020; 11:1515-1530. [PMID: 32391121 PMCID: PMC7197451 DOI: 10.18632/oncotarget.27566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/03/2020] [Indexed: 01/05/2023] Open
Abstract
Clinical, epidemiological and experimental data identified the insulin-like growth factor-1 receptor (IGF1R) as a candidate therapeutic target in oncology. While this paradigm is based on well-established biological facts, including the potent anti-apoptotic and cell survival capabilities of the receptor, most Phase III clinical trials designed to target the IGF1R led to disappointing results. The present study was aimed at evaluating the hypothesis that combined treatment composed of selective IGF1R inhibitor along with classical chemotherapy might be more effective than individual monotherapies in breast cancer treatment. Analyses included comprehensive measurements of the synergism achieved by various combination regimens using the CompuSyn software. In addition, proteomic analyses were conducted to identify the proteins involved in the synergistic killing effect at a global level. Data presented here demonstrates that co-treatment of IGF1R inhibitor along with chemotherapeutic drugs markedly improves the therapeutic efficiency in breast cancer cells. Of clinical relevance, our analyses indicate that high IGF1R baseline expression may serve as a predictive biomarker for IGF1R targeted therapy. In addition, we identified a ten-genes signature with potential predictive value. In conclusion, the use of a series of bioinformatics tools shed light on some of the biological pathways that might be responsible for synergysm in cancer therapy.
Collapse
Affiliation(s)
- Tali Sinai-Livne
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Zoya Cohen
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer 52620, Israel
| | - Ilan Tsarfaty
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.,Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv 69978, Israel
| | - Raanan Berger
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer 52620, Israel
| |
Collapse
|
24
|
de Bono J, Lin CC, Chen LT, Corral J, Michalarea V, Rihawi K, Ong M, Lee JH, Hsu CH, Yang JCH, Shiah HS, Yen CJ, Anthoney A, Jove M, Buschke S, Fuertig R, Schmid U, Goeldner RG, Strelkowa N, Huang DCL, Bogenrieder T, Twelves C, Cheng AL. Two first-in-human studies of xentuzumab, a humanised insulin-like growth factor (IGF)-neutralising antibody, in patients with advanced solid tumours. Br J Cancer 2020; 122:1324-1332. [PMID: 32161368 PMCID: PMC7188670 DOI: 10.1038/s41416-020-0774-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/05/2020] [Accepted: 02/17/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Xentuzumab, an insulin-like growth factor (IGF)-1/IGF-2-neutralising antibody, binds IGF-1 and IGF-2, inhibiting their growth-promoting signalling. Two first-in-human trials assessed the maximum-tolerated/relevant biological dose (MTD/RBD), safety, pharmacokinetics, pharmacodynamics, and activity of xentuzumab in advanced/metastatic solid cancers. METHODS These phase 1, open-label trials comprised dose-finding (part I; 3 + 3 design) and expansion cohorts (part II; selected tumours; RBD [weekly dosing]). Primary endpoints were MTD/RBD. RESULTS Study 1280.1 involved 61 patients (part I: xentuzumab 10-1800 mg weekly, n = 48; part II: 1000 mg weekly, n = 13); study 1280.2, 64 patients (part I: 10-3600 mg three-weekly, n = 33; part II: 1000 mg weekly, n = 31). One dose-limiting toxicity occurred; the MTD was not reached for either schedule. Adverse events were generally grade 1/2, mostly gastrointestinal. Xentuzumab showed dose-proportional pharmacokinetics. Total plasma IGF-1 increased dose dependently, plateauing at ~1000 mg/week; at ≥450 mg/week, IGF bioactivity was almost undetectable. Two partial responses occurred (poorly differentiated nasopharyngeal carcinoma and peripheral primitive neuroectodermal tumour). Integration of biomarker and response data by Bayesian Logistic Regression Modeling (BLRM) confirmed the RBD. CONCLUSIONS Xentuzumab was well tolerated; MTD was not reached. RBD was 1000 mg weekly, confirmed by BLRM. Xentuzumab showed preliminary anti-tumour activity. CLINICAL TRIAL REGISTRATION NCT01403974; NCT01317420.
Collapse
Affiliation(s)
- Johann de Bono
- Drug Development Unit, Royal Marsden Hospital & Institute of Cancer Research, Downs Road, Sutton, UK.
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - Li-Tzong Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138 Sheng Li Road, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, 100 Tzyou 1st Road, Kaohsiung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng Li Road, Tainan, Taiwan
| | - Jesus Corral
- Medical Oncology Department, Clinica Universidad de Navarra, Calle Marquesado de Sta. Marta 1, Madrid, Spain
| | - Vasiliki Michalarea
- Drug Development Unit, Royal Marsden Hospital & Institute of Cancer Research, Downs Road, Sutton, UK
| | - Karim Rihawi
- Drug Development Unit, Royal Marsden Hospital & Institute of Cancer Research, Downs Road, Sutton, UK
- Azienda Sanitaria Universitaria Integrata di Udine, Via Pozzuolo, 330, 33100, Udine, Italy
| | - Michael Ong
- The Ottawa Hospital Cancer Centre, 501 Smyth Road, Ottawa, ON, Canada
| | - Jih-Hsiang Lee
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - Chih-Hung Hsu
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
| | - Her-Shyong Shiah
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, 250 Wuxing Street, Taipei, Taiwan
| | - Chia-Jui Yen
- Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 138 Sheng Li Road, Tainan, Taiwan
| | - Alan Anthoney
- University of Leeds and Leeds Teaching Hospitals Trust, Beckett Street, Leeds, UK
| | - Maria Jove
- University of Leeds and Leeds Teaching Hospitals Trust, Beckett Street, Leeds, UK
| | - Susanne Buschke
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - René Fuertig
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Ulrike Schmid
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Rainer-Georg Goeldner
- Biostatistics and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Natalja Strelkowa
- Biostatistics and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, Biberach an der Riß, Germany
| | - Dennis Chin-Lun Huang
- Medical Department, Boehringer Ingelheim Taiwan Ltd, 12F, No. 2, Sec 3, Minsheng East Road, Taipei, Taiwan
| | - Thomas Bogenrieder
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, 1 Geschwister-Scholl-Platz, Munich, Germany
- Medicine and Translational Research, Boehringer Ingelheim RCV, 5-11 Doktor-Boehringer-Gasse, Vienna, Austria
| | - Chris Twelves
- University of Leeds and Leeds Teaching Hospitals Trust, Beckett Street, Leeds, UK
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, 7 Chung Shan S. Rd., Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
25
|
Sarfstein R, Yeheskel A, Sinai-Livne T, Pasmanik-Chor M, Werner H. Systems Analysis of Insulin and IGF1 Receptors Networks in Breast Cancer Cells Identifies Commonalities and Divergences in Expression Patterns. Front Endocrinol (Lausanne) 2020; 11:435. [PMID: 32733384 PMCID: PMC7359857 DOI: 10.3389/fendo.2020.00435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/02/2020] [Indexed: 11/13/2022] Open
Abstract
Insulin and insulin-like growth factor-1 (IGF1), acting respectively via the insulin (INSR) and IGF1 (IGF1R) receptors, play key developmental and metabolic roles throughout life. In addition, both signaling pathways fulfill important roles in cancer initiation and progression. The present study was aimed at identifying mechanistic differences between INSR and IGF1R using a recently developed bioinformatics tool, the Biological Network Simulator (BioNSi). This application allows to import and merge multiple pathways and interaction information from the KEGG database into a single network representation. The BioNsi network simulation tool allowed us to exploit the availability of gene expression data derived from breast cancer cell lines with specific disruptions of the INSR or IGF1R genes in order to investigate potential differences in protein expression that might be linked to biological attributes of the specific receptor networks. Modeling-generated information was corroborated by experimental and biological assays. BioNSi analyses revealed that the expression of 75 and 71 genes changed during simulation of IGF1R-KD and INSR-KD, compared to control cells, respectively. Out of 16 proteins that BioNSi analysis was based on, validated by Western blotting, nine were shown to be involved in DNA repair, eight in cell cycle checkpoints, six in proliferation, eight in apoptosis, seven in oxidative stress, six in cell migration, two in energy homeostasis, and three in senescence. Taken together, analyses identified a number of commonalities and, most importantly, dissimilarities between the IGF1R and INSR pathways that might help explain the basis for the biological differences between these networks.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Humans
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/antagonists & inhibitors
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Systems Analysis
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Rive Sarfstein
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adva Yeheskel
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tali Sinai-Livne
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Metsada Pasmanik-Chor
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv, Israel
- Haim Werner
| |
Collapse
|
26
|
Werner H, Sarfstein R, Bruchim I. Investigational IGF1R inhibitors in early stage clinical trials for cancer therapy. Expert Opin Investig Drugs 2019; 28:1101-1112. [PMID: 31731883 DOI: 10.1080/13543784.2019.1694660] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: The insulin-like growth factors (IGFs) are a family of secreted peptide hormones with important roles in different cellular and organism functions. The biological activities of the IGFs are mediated by the IGF1 receptor (IGF1R), a cell surface, tyrosine kinase-containing heterotetramer that is linked to numerous cytoplasmic signaling cascades. The IGF1R displays potent antiapoptotic, pro-survival capacities and plays a key role in malignant transformation. Research has identified the IGF1R as a candidate therapeutic target in cancer.Areas covered: We offer a synopsis of ongoing efforts to target the IGF axis for therapeutic purposes. Our review includes a digest of early experimental work that led to the identification of IGF1R as a candidate therapeutic target in oncology.Expert opinion: Targeting of the IGF axis has yielded disappointing results in phase III trials, but it is important to learn from this to improve future trials in a rational manner. The potential of anti-IGF1R antibodies and small molecular weight inhibitors, alone or in combination with chemotherapy or other biological agents, should be investigated further in randomized studies. Moreover, the implementation of predictive biomarkers for patient selection will improve the outcome of future trials. Emerging personalized medicine could have a major impact on IGF1R targeting.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv, Israel
| | - Rive Sarfstein
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Bruchim
- Gynecologic Oncology Division, Hillel Yaffe Medical Center, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
27
|
Nedeljković M, Damjanović A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer-How We Can Rise to the Challenge. Cells 2019; 8:E957. [PMID: 31443516 PMCID: PMC6770896 DOI: 10.3390/cells8090957] [Citation(s) in RCA: 459] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Triple-negative (TNBC) is the most lethal subtype of breast cancer owing to high heterogeneity, aggressive nature, and lack of treatment options. Chemotherapy remains the standard of care for TNBC treatment, but unfortunately, patients frequently develop resistance. Accordingly, in recent years, tremendous effort has been made into elucidating the mechanisms of TNBC chemoresistance with the goal of identifying new molecular targets. It has become evident that the development of TNBC chemoresistance is multifaceted and based on the elaborate interplay of the tumor microenvironment, drug efflux, cancer stem cells, and bulk tumor cells. Alterations of multiple signaling pathways govern these interactions. Moreover, TNBC's high heterogeneity, highlighted in the existence of several molecular signatures, presents a significant obstacle to successful treatment. In the present, in-depth review, we explore the contribution of key mechanisms to TNBC chemoresistance as well as emerging strategies to overcome them. We discuss novel anti-tumor agents that target the components of these mechanisms and pay special attention to their current clinical development while emphasizing the challenges still ahead of successful TNBC management. The evidence presented in this review outlines the role of crucial pathways in TNBC survival following chemotherapy treatment and highlights the importance of using combinatorial drug strategies and incorporating biomarkers in clinical studies.
Collapse
Affiliation(s)
- Milica Nedeljković
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia.
| | - Ana Damjanović
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
28
|
Sharma P, Kumar S. S961, a biosynthetic insulin receptor antagonist, downregulates insulin receptor expression & suppresses the growth of breast cancer cells. Indian J Med Res 2018; 147:545-551. [PMID: 30168485 PMCID: PMC6118145 DOI: 10.4103/ijmr.ijmr_403_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background & objectives Insulin resistance associated with hyperinsulinaemia and overexpression of insulin receptors (IRs) have been intricately linked to the pathogenesis and treatment outcomes of the breast carcinoma. Studies have revealed that upregulated expression of IRs in breast cancer pathogenesis regulates several aspects of the malignant phenotype, including cell proliferation and metastasis. This study was aimed to investigate the pivotal role of an IR antagonist S961 on IR signalling and other biological parameters in MCF-7, MDA-MB-231 and T47D cell lines. Methods The effect of human insulin and S961 on growth, proliferation rate and clonogenic potential of breast cancer cells was evaluated by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide] assay and clonogenic assay. The mRNA expression of IR isoforms (IR-A and IR-B) was measured in the breast carcinoma cells using quantitative PCR. Results The study revealed that breast cancer cells predominantly expressed IR-A isoform and showed extensive growth and proliferation owing to IR overexpression. It was found that S961 downregulated the IRs (IR-A and IR-B) with nanomolar dose and efficiently blocked expression of IRs even in the presence of insulin. IR mRNA expression levels were significantly downregulated in the continued presence of S961. S961 also inhibited cellular proliferation and colony formation in breast tumour cells. Interpretation & conclusions IR antagonist, S961 showed distinct antagonism in vitro and appeared to be a powerful therapeutic modality that might provide insight into the pathogenesis of impaired IR signalling.
Collapse
Affiliation(s)
- Prateek Sharma
- Centre for Biosciences, School of Basic & Applied Science, Central University of Punjab, Bathinda, India
| | - Sanjeev Kumar
- Centre for Biosciences, School of Basic & Applied Science, Central University of Punjab, Bathinda, India
| |
Collapse
|
29
|
Insulin Receptor Substrate Suppression by the Tyrphostin NT157 Inhibits Responses to Insulin-Like Growth Factor-I and Insulin in Breast Cancer Cells. Discov Oncol 2018; 9:371-382. [PMID: 30229539 DOI: 10.1007/s12672-018-0343-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022] Open
Abstract
Insulin and insulin-like growth factor (IGF) signaling systems regulate breast cancer growth, progression, and metastasis. The insulin receptor substrates 1 and 2 (IRS1/2) transduce signaling from the type I IGF receptor (IGF-IR) and insulin receptor (InR) to mediate the biological effects of receptor activation. In breast cancer, IRS-1 plays a critical role in cancer cell proliferation while IRS-2 is associated with motility and metastasis. NT157, a small-molecule tyrphostin, downregulates IRS proteins in several model systems. In breast cancer cells, NT157 treatment suppressed IRS protein expression in a dose-dependent manner. Exposure to NT157 inhibited the activation of downstream signaling mediated by the IRS proteins. NT157 induced a MAPK-dependent serine phosphorylation of IRS proteins which resulted in disassociation between IRS proteins and their receptors resulting in IRS degradation. In estrogen receptor-α-positive (ERα+) breast cancer cells (MCF-7 and T47D), NT157 also resulted in cytoplasmic ERα downregulation likely because of disruption of an IRS-1-IGF-IR/InR/ERα complex. NT157 decreased S phase fraction, monolayer, and anchorage-independent growth after IGF/insulin treatment in ERα+ breast cancer cells. NT157 downregulation of IRS protein expression also sensitized ERα+ breast cancer cells to rapamycin. Moreover, NT157 inhibited the growth of tamoxifen-resistant ERα+ breast cancer cells. Given that both IGF-IR and InR play a role in cancer biology, targeting of IRS adaptor proteins may be a more effective strategy to inhibit the function of these receptors.
Collapse
|
30
|
Allegra A, Innao V, Gerace D, Allegra AG, Vaddinelli D, Bianco O, Musolino C. The adipose organ and multiple myeloma: Impact of adipokines on tumor growth and potential sites for therapeutic intervention. Eur J Intern Med 2018; 53:12-20. [PMID: 29859797 DOI: 10.1016/j.ejim.2018.05.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/26/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023]
Abstract
In addition to its capacity to store lipids the adipose tissue is now identified as a real organ with both endocrine and metabolic roles. Preclinical results indicate that modifying adipose tissue and bone marrow adipose tissue (BMAT) could be a successful multiple myeloma (MM) therapy. BMAT interrelates with bone marrow cells and other immune cells, and may influence MM disease progression. The BM adipocytes may have a role in MM progression, bone homing, chemoresistance, and relapse, due to local endocrine, paracrine, or metabolic factors. BM adipocytes isolated from MM subjects have been shown to increase myeloma growth in vitro and may preserve cells from chemotherapy-induced apoptosis. By producing free fatty acids and emitting signaling molecules such as growth factors and adipokines, BM adipocytes are both an energy font and an endocrine signaling factory. This review should suggest future research approaches toward developing novel treatments to target MM by targeting BMAT and its products.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy.
| | - Vanessa Innao
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| | - Demetrio Gerace
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| | - Andrea Gaetano Allegra
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| | - Doriana Vaddinelli
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| | - Oriana Bianco
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| |
Collapse
|
31
|
Identification of a Novel Invasion-Promoting Region in Insulin Receptor Substrate 2. Mol Cell Biol 2018; 38:MCB.00590-17. [PMID: 29685905 DOI: 10.1128/mcb.00590-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/15/2018] [Indexed: 12/13/2022] Open
Abstract
Although the insulin receptor substrate (IRS) proteins IRS1 and IRS2 share considerable homology and activate common signaling pathways, their contributions to breast cancer are distinct. IRS1 has been implicated in the proliferation and survival of breast tumor cells. In contrast, IRS2 facilitates glycolysis, invasion, and metastasis. To determine the mechanistic basis for IRS2-dependent functions, we investigated unique structural features of IRS2 that are required for invasion. Our studies revealed that the ability of IRS2 to promote invasion is dependent upon upstream insulin-like growth factor 1 receptor (IGF-1R)/insulin receptor (IR) activation and the recruitment and activation of phosphatidylinositol 3-kinase (PI3K), functions shared with IRS1. In addition, a 174-amino-acid region in the IRS2 C-terminal tail, which is not conserved in IRS1, is also required for IRS2-mediated invasion. Importantly, this "invasion (INV) region" is sufficient to confer invasion-promoting ability when swapped into IRS1. However, the INV region is not required for the IRS2-dependent regulation of glucose uptake. Bone morphogenetic protein 2-inducible kinase (BMP2K) binds to the INV region and contributes to IRS2-dependent invasion. Taken together, our data advance the mechanistic understanding of how IRS2 regulates invasion and reveal that IRS2 functions important for cancer can be independently targeted without interfering with the metabolic activities of this adaptor protein.
Collapse
|
32
|
Björner S, Rosendahl AH, Simonsson M, Markkula A, Jirström K, Borgquist S, Rose C, Ingvar C, Jernström H. Combined and individual tumor-specific expression of insulin-like growth factor-I receptor, insulin receptor and phospho-insulin-like growth factor-I receptor/insulin receptor in primary breast cancer: Implications for prognosis in different treatment groups. Oncotarget 2018; 8:9093-9107. [PMID: 28030849 PMCID: PMC5354717 DOI: 10.18632/oncotarget.14082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022] Open
Abstract
Clinical trials examining insulin-like growth factor-I receptor (IGF1R)-targeting strategies have emphasized that better predictive biomarkers are required to improve patient selection. Immunohistochemical tumor-specific protein expression of IGF1R, insulin receptor (InsR), and phosphorylated IGF1R/InsR (pIGF1R/InsR) individually and combined in relation to breast cancer prognosis was evaluated in a population-based cohort of 1,026 primary invasive breast cancer patients without preoperative treatment diagnosed in Sweden. IGF1R (n = 923), InsR (n = 900), and pIGF1R/InsR (n = 904) combined cytoplasmic and membrane staining was dichotomized. IGF1Rstrong/InsRmod/strong/pIGF1R/InsRpos tumors were borderline associated with 2-fold risk for events, HRadj (2.00; 95%CI 0.96-4.18). Combined IGF1R and pIGF1R/InsR status only impacted prognosis in patients with InsRmod/strong expressing tumors (Pinteraction = 0.041). IGF1Rstrong expression impacted endocrine treatment response differently depending on patients’ age and type of endocrine therapy. Phospho-IGF1R/InsRpos was associated with lower risk for events among non-endocrine-treated patients irrespective of ER status, HRadj (0.32; 95%CI 0.16-0.63), but not among endocrine-treated patients (Pinteraction = 0.024). In non-endocrine-treated patients, pIGF1R/InsRpos was associated with lower risk for events after radiotherapy, HRadj (0.31; 95%CI 0.12-0.80), and chemotherapy, HRadj (0.29; 95%CI 0.09-0.99). This study highlights the complexity of IGF hetero-and homodimer signaling network and its interplay with endocrine treatment, suggesting that combinations of involved factors may improve patient selection for IGF1R-targeted therapy.
Collapse
Affiliation(s)
- Sofie Björner
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Maria Simonsson
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Andrea Markkula
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden.,Department of Oncology and Haematology, Skåne University Hospital, Sweden
| | - Carsten Rose
- CREATE Health and Department of Immunotechnology, Lund University, Medicon Village, Lund, Sweden
| | - Christian Ingvar
- Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Surgery, Lund, Sweden
| | - Helena Jernström
- Department of Clinical Sciences Lund, Lund University Faculty of Medicine, Oncology and Pathology, Lund, Sweden
| |
Collapse
|
33
|
Lee HJ, Pham PC, Hyun SY, Baek B, Kim B, Kim Y, Min HY, Lee J, Lee HY. Development of a 4-aminopyrazolo[3,4-d]pyrimidine-based dual IGF1R/Src inhibitor as a novel anticancer agent with minimal toxicity. Mol Cancer 2018; 17:50. [PMID: 29455661 PMCID: PMC5817804 DOI: 10.1186/s12943-018-0802-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Both the type I insulin-like growth factor receptor (IGF1R) and Src pathways are associated with the development and progression of numerous types of human cancer, and Src activation confers resistance to anti-IGF1R therapies. Hence, targeting both IGF1R and Src concurrently is one of the main challenges in combating resistance to the currently available anti-IGF1R-based anticancer therapies. However, the enhanced toxicity from this combinatorial treatment could be one of the main hurdles for this strategy, suggesting the necessity of developing a novel strategy for co-targeting IGF1R and Src to meet an urgent clinical need. METHODS We synthesized a series of 4-aminopyrazolo[3,4-d]pyrimidine-based dual IGF1R/Src inhibitors, selected LL28 as an active compound and evaluated its potential antitumor effects in vitro and in vivo using the MTT assay, colony formation assays, flow cytometric analysis, a tumor xenograft model, and the Kras G12D/+ -driven spontaneous lung tumorigenesis model. RESULTS LL28 markedly suppressed the activation of IGF1R and Src and significantly inhibited the viability of several NSCLC cell lines in vitro by inducing apoptosis. Administration of mice with LL28 significantly suppressed the growth of H1299 NSCLC xenograft tumors without overt toxicity and substantially reduced the multiplicity, volume, and load of lung tumors in the Kras G12D/+ -driven lung tumorigenesis model. CONCLUSIONS The present results suggest the potential of LL28 as a novel anticancer drug candidate targeting both IGF1R and Src, providing a new avenue to efficient anticancer therapies. Further investigation is warranted in advanced preclinical and clinical settings.
Collapse
Affiliation(s)
- Ho Jin Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Phuong Chi Pham
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Yeob Hyun
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byungyeob Baek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byungjin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yunha Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeeyeon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea. .,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
34
|
Năstase L, Cretoiu D, Stoicescu SM. Skeletal Muscle Damage in Intrauterine Growth Restriction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:93-106. [PMID: 30390249 DOI: 10.1007/978-981-13-1435-3_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Intrauterine growth restriction (IUGR) represents a rate of fetal growth that is less than average for the population and the growth potential of a specific infant. IUGR produces infants who are small for gestational age (SGA) but also appropriate for gestational age (AGA). It refers to growth less than expected for gestational age and is most often under 10th percentiles for age. It develops during the late second and third trimesters of gestation. The etiology of IUGR is multifactorial. One of the most important factors which leads to IUGR is a decrease of nutrients and oxygen delivered to the fetus by the placenta. The growth of adipose tissue and skeletal muscle is limited by the declined fetal nutrient supply later in gestation. IUGR affects about 24% of babies born in developing countries. Worldwide, IUGR is the second cause of perinatal morbidity and mortality behind the premature birth and a major predisposing factor to metabolic disorders throughout postnatal life, even at adult age. Skeletal muscle represents about 35-40% of the body mass and plays an essential role in metabolic homeostasis, being responsible for 65% of fetal glucose consumption. A reduction in skeletal muscle growth characterizes IUGR fetuses compared to normal weight neonates. The decrease in muscle mass is not compensated after birth and persists until adulthood. This is a review of the literature, a neonatological, clinical point of view on the effects of IUGR on striated muscles. The available studies on this subject are currently the results of experimental research on animals, and information about the human fetus and newborn are scarce.
Collapse
Affiliation(s)
- Leonard Năstase
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania. .,Alessandrescu-Rusescu National Institute for the Mother and Child Health, Polizu Maternity, Bucharest, Romania.
| | - Dragos Cretoiu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Alessandrescu-Rusescu National Institute for the Mother and Child Health, Polizu Maternity, Bucharest, Romania
| | - Silvia Maria Stoicescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Alessandrescu-Rusescu National Institute for the Mother and Child Health, Polizu Maternity, Bucharest, Romania
| |
Collapse
|
35
|
Beattie J, Al-Khafaji H, Noer PR, Alkharobi HE, Alhodhodi A, Meade J, El-Gendy R, Oxvig C. Insulin- like Growth Factor-Binding Protein Action in Bone Tissue: A Key Role for Pregnancy- Associated Plasma Protein-A. Front Endocrinol (Lausanne) 2018; 9:31. [PMID: 29503631 PMCID: PMC5820303 DOI: 10.3389/fendo.2018.00031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/23/2018] [Indexed: 11/13/2022] Open
Abstract
The insulin-like growth factor (IGF) axis is required for the differentiation, development, and maintenance of bone tissue. Accordingly, dysregulation of this axis is associated with various skeletal pathologies including growth abnormalities and compromised bone structure. It is becoming increasingly apparent that the action of the IGF axis must be viewed holistically taking into account not just the actions of the growth factors and receptors, but also the influence of soluble high affinity IGF binding proteins (IGFBPs).There is a recognition that IGFBPs exert IGF-dependent and IGF-independent effects in bone and other tissues and that an understanding of the mechanisms of action of IGFBPs and their regulation in the pericellular environment impact critically on tissue physiology. In this respect, a group of IGFBP proteinases (which may be considered as ancillary members of the IGF axis) play a crucial role in regulating IGFBP function. In this model, cleavage of IGFBPs by specific proteinases into fragments with lower affinity for growth factor(s) regulates the partition of IGFs between IGFBPs and cell surface IGF receptors. In this review, we examine the importance of IGFBP function in bone tissue with special emphasis on the role of pregnancy associated plasma protein-A (PAPP-A). We examine the function of PAPP-A primarily as an IGFBP-4 proteinase and present evidence that PAPP-A induced cleavage of IGFBP-4 is potentially a key regulatory step in bone metabolism. We also highlight some recent findings with regard to IGFBP-2 and IGFBP-5 (also PAPP-A substrates) function in bone tissue and briefly discuss the actions of the other three IGFBPs (-1, -3, and -6) in this tissue. Although our main focus will be in bone we will allude to IGFBP activity in other cells and tissues where appropriate.
Collapse
Affiliation(s)
- James Beattie
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
- *Correspondence: James Beattie,
| | - Hasanain Al-Khafaji
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
| | - Pernille R. Noer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Hanaa Esa Alkharobi
- Department of Oral Biology, Dental College, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Aishah Alhodhodi
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
| | - Josephine Meade
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
| | - Reem El-Gendy
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
36
|
Yang Y, Leonard M, Zhang Y, Zhao D, Mahmoud C, Khan S, Wang J, Lower EE, Zhang X. HER2-Driven Breast Tumorigenesis Relies upon Interactions of the Estrogen Receptor with Coactivator MED1. Cancer Res 2017; 78:422-435. [PMID: 29187405 DOI: 10.1158/0008-5472.can-17-1533] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/18/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
Studies of the estrogen receptor (ER) coactivator protein Mediator subunit 1 (MED1) have revealed its specific roles in pubertal mammary gland development and potential contributions to breast tumorigenesis, based on coamplification of MED1 and HER2 in certain breast cancers. In this study, we generated a mouse model of mammary tumorigenesis harboring the MMTV-HER2 oncogene and mutation of MED1 to evaluate its role in HER2-driven tumorigenesis. MED1 mutation in its ER-interacting LxxLL motifs was sufficient to delay tumor onset and to impair tumor growth, metastasis, and cancer stem-like cell formation in this model. Mechanistic investigations revealed that MED1 acted directly to regulate ER signaling through the downstream IGF1 pathway but not the AREG pathway. Our findings show that MED1 is critical for HER2-driven breast tumorigenesis, suggesting its candidacy as a disease-selective therapeutic target.Significance: These findings identify an estrogen receptor-binding protein as a critical mediator of HER2-driven breast tumorigenesis, suggesting its candidacy as a disease-selective therapeutic target. Cancer Res; 78(2); 422-35. ©2017 AACR.
Collapse
Affiliation(s)
- Yongguang Yang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marissa Leonard
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Graduate Program in Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yijuan Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Dan Zhao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Charif Mahmoud
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Shugufta Khan
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Elyse E Lower
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xiaoting Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio. .,Graduate Program in Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
37
|
May CD, Landers SM, Bolshakov S, Ma X, Ingram DR, Kivlin CM, Watson KL, Sannaa GAA, Bhalla AD, Wang WL, Lazar AJ, Torres KE. Co-targeting PI3K, mTOR, and IGF1R with small molecule inhibitors for treating undifferentiated pleomorphic sarcoma. Cancer Biol Ther 2017; 18:816-826. [PMID: 29099264 PMCID: PMC5678691 DOI: 10.1080/15384047.2017.1373230] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Undifferentiated pleomorphic sarcomas (UPSs) are aggressive mesenchymal malignancies with no definitive cell of origin or specific recurrent genetic hallmarks. These tumors are largely chemoresistant; thus, identification of potential therapeutic targets is necessary to improve patient outcome. Previous studies demonstrated that high expression of activated protein kinase B (AKT) in patients with UPS corresponds to poor disease-specific survival. Here, we demonstrate that inhibiting phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) signaling using a small molecule inhibitor reduced UPS cell proliferation and motility and xenograft growth; however, increased phosphorylation of insulin-like growth factor 1 receptor (IGF1R) indicated the potential for adaptive resistance following treatment through compensatory receptor activation. Co-treatment with a dual PI3K/mTOR inhibitor and an anti-IGF1R kinase inhibitor reduced in vivo tumor growth rates despite a lack of antiproliferative effects in vitro. Moreover, this combination treatment significantly decreased UPS cell migration and invasion, which is linked to changes in p27 subcellular localization. Our results demonstrate that targeted inhibition of multiple components of the IGF1R/PI3K/mTOR pathway was more efficacious than single-agent therapy and suggest that co-targeting this pathway could be a beneficial therapeutic strategy for patients with UPS.
Collapse
Affiliation(s)
- Caitlin D May
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA
| | - Sharon M Landers
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Svetlana Bolshakov
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - XiaoYan Ma
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Davis R Ingram
- c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Christine M Kivlin
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA
| | - Kelsey L Watson
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Ghadah A Al Sannaa
- c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Angela D Bhalla
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Wei-Lien Wang
- c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Alexander J Lazar
- b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA.,c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Keila E Torres
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA
| |
Collapse
|
38
|
Chen L, Fu W, Feng C, Qu R, Tong L, Zheng L, Fang B, Qiu Y, Hu J, Cai Y, Feng J, Xie H, Ding J, Liu Z, Liang G. Structure-based design and synthesis of 2,4-diaminopyrimidines as EGFR L858R/T790M selective inhibitors for NSCLC. Eur J Med Chem 2017; 140:510-527. [DOI: 10.1016/j.ejmech.2017.08.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/06/2023]
|
39
|
Sanderson MP, Hofmann MH, Garin-Chesa P, Schweifer N, Wernitznig A, Fischer S, Jeschko A, Meyer R, Moll J, Pecina T, Arnhof H, Weyer-Czernilofsky U, Zahn SK, Adolf GR, Kraut N. The IGF1R/INSR Inhibitor BI 885578 Selectively Inhibits Growth of IGF2-Overexpressing Colorectal Cancer Tumors and Potentiates the Efficacy of Anti-VEGF Therapy. Mol Cancer Ther 2017; 16:2223-2233. [PMID: 28729397 DOI: 10.1158/1535-7163.mct-17-0336] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/16/2017] [Accepted: 06/30/2017] [Indexed: 12/21/2022]
Abstract
Clinical studies of pharmacologic agents targeting the insulin-like growth factor (IGF) pathway in unselected cancer patients have so far demonstrated modest efficacy outcomes, with objective responses being rare. As such, the identification of selection biomarkers for enrichment of potential responders represents a high priority for future trials of these agents. Several reports have described high IGF2 expression in a subset of colorectal cancers, with focal IGF2 amplification being responsible for some of these cases. We defined a novel cut-off value for IGF2 overexpression based on differential expression between colorectal tumors and normal tissue samples. Analysis of two independent colorectal cancer datasets revealed IGF2 to be overexpressed at a frequency of 13% to 22%. An in vitro screen of 34 colorectal cancer cell lines revealed IGF2 expression to significantly correlate with sensitivity to the IGF1R/INSR inhibitor BI 885578. Furthermore, autocrine IGF2 constitutively activated IGF1R and Akt phosphorylation, which was inhibited by BI 885578 treatment. BI 885578 significantly delayed the growth of IGF2-high colorectal cancer xenograft tumors in mice, while combination with a VEGF-A antibody increased efficacy and induced tumor regression. Besides colorectal cancer, IGF2 overexpression was detected in more than 10% of bladder carcinoma, hepatocellular carcinoma and non-small cell lung cancer patient samples. Meanwhile, IGF2-high non-colorectal cancer cells lines displayed constitutive IGF1R phosphorylation and were sensitive to BI 885578. Our findings suggest that IGF2 may represent an attractive patient selection biomarker for IGF pathway inhibitors and that combination with VEGF-targeting agents may further improve clinical outcomes. Mol Cancer Ther; 16(10); 2223-33. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Reiner Meyer
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Jürgen Moll
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Thomas Pecina
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | | | | | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
40
|
Higgins GS, Krause M, McKenna WG, Baumann M. Personalized Radiation Oncology: Epidermal Growth Factor Receptor and Other Receptor Tyrosine Kinase Inhibitors. Recent Results Cancer Res 2017; 198:107-22. [PMID: 27318683 DOI: 10.1007/978-3-662-49651-0_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Molecular biomarkers are currently evaluated in preclinical and clinical studies in order to establish predictors for treatment decisions in radiation oncology. The receptor tyrosine kinases (RTK) are described in the following text. Among them, the most data are available for the epidermal growth factor receptor (EGFR) that plays a major role for prognosis of patients after radiotherapy, but seems also to be involved in mechanisms of radioresistance, specifically in repopulation of tumour cells between radiotherapy fractions. Monoclonal antibodies against the EGFR improve locoregional tumour control and survival when applied during radiotherapy, however, the effects are heterogeneous and biomarkers for patient selection are warranted. Also other RTK´s such as c-Met and IGF-1R seem to play important roles in tumour radioresistance. Beside the potential to select patients for molecular targeting approaches combined with radiotherapy, studies are also needed to evluate radiotherapy adaptation approaches for selected patients, i.e. adaptation of radiation dose, or, more sophisticated, of target volumes.
Collapse
Affiliation(s)
- Geoff S Higgins
- Gray Laboratories, Department of Oncology, Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- German Cancer Consortium (DKTK) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Insititute of Radiooncology, Dresden, Germany.
- Department of Radiation Oncology, Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - W Gillies McKenna
- Gray Laboratories, Department of Oncology, Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Insititute of Radiooncology, Dresden, Germany
- Department of Radiation Oncology, Carl Gustav Carus Faculty of Medicine, University Hospital, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
41
|
Solingapuram Sai KK, Prabhakaran J, Sattiraju A, Mann JJ, Mintz A, Kumar JD. Radiosynthesis and evaluation of IGF1R PET ligand [ 11 C]GSK1838705A. Bioorg Med Chem Lett 2017; 27:2895-2897. [DOI: 10.1016/j.bmcl.2017.04.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 12/19/2022]
|
42
|
Chan JY, Hackel BJ, Yee D. Targeting Insulin Receptor in Breast Cancer Using Small Engineered Protein Scaffolds. Mol Cancer Ther 2017; 16:1324-1334. [PMID: 28468775 DOI: 10.1158/1535-7163.mct-16-0685] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 02/15/2017] [Accepted: 04/20/2017] [Indexed: 11/16/2022]
Abstract
Insulin receptor (InsR) and the type I insulin-like growth factor (IGF1R) are homologous receptors necessary for signal transduction by their cognate ligands insulin, IGF-I and IGF-II. IGF1R mAbs, intended to inhibit malignant phenotypic signaling, failed to show benefit in patients with endocrine-resistant tumors in phase III clinical trials. Our previous work showed that in tamoxifen-resistant cells, IGF1R expression was lacking, but InsR inhibition effectively blocked growth. In endocrine-sensitive breast cancer cells, insulin was not growth stimulatory, likely due to the presence of hybrid InsR/IGF1R, which has high affinity for IGF-I, but not insulin. Combination inhibition of InsR and IGF1R showed complete suppression of the system in endocrine-sensitive breast cancer cells. To develop InsR-binding agents, we employed a small protein scaffold, T7 phage gene 2 protein (Gp2) with the long-term goal of creating effective InsR inhibitors and diagnostics. Using yeast display and directed evolution, we identified three Gp2 variants (Gp2 #1, #5, and #10) with low nanomolar affinity and specific binding to cell surface InsR. These Gp2 variants inhibited insulin-mediated monolayer proliferation in both endocrine-sensitive and resistant breast cancer, but did not downregulate InsR expression. Gp2 #5 and Gp2 #10 disrupted InsR function by inhibiting ligand-induced receptor activation. In contrast, Gp2 #1 did not block InsR phosphorylation. Notably, Gp2 #1 binding was enhanced by pretreatment of cells with insulin, suggesting a unique receptor-ligand-binding mode. These Gp2 variants are the first nonimmunoglobulin protein scaffolds to target insulin receptor and present compelling opportunity for modulation of InsR signaling. Mol Cancer Ther; 16(7); 1324-34. ©2017 AACR.
Collapse
Affiliation(s)
- Jie Ying Chan
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Douglas Yee
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota. .,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
43
|
Abstract
The type I insulin-like growth factor-1 receptor is a well-described target in breast cancer and multiple clinical trials examining insulin-like growth factor-1 receptor have been completed. Unfortunately, monoclonal antibodies and tyrosine kinase inhibitors targeting insulin-like growth factor-1 receptor failed in phase III breast clinical trials for several reasons. First, insulin-like growth factor-1 receptor antibody therapy resulted in hyperglycemia and metabolic syndrome most likely due to disruption of insulin-like growth factor-1 homeostasis and subsequent growth hormone elevation. Growth hormone elevation induces insulin resistance, hence a subsequent elevation of insulin and the potential for activation of insulin receptor. Second, the insulin-like growth factor-1 receptor and insulin receptor are highly homologous in amino acid sequence, structure, and function. These two receptors bind insulin, insulin-like growth factor-1 and insulin-like growth factor-2, to regulate glucose uptake and other cellular functions. Hybrid receptors composed of one chain of insulin-like growth factor-1 receptor and insulin receptor also participate in signaling. Third, since all the monoclonal antibodies were specific for insulin-like growth factor-1 receptor, any pathophysiologic role for insulin receptor was not inhibited. While the insulin-like growth factor-1 receptor tyrosine kinase inhibitors effectively inhibited both insulin-like growth factor-1 receptor and insulin receptor, these drugs are not being further developed likely due to their metabolic toxicities. Insulin-like growth factor-1/2 neutralizing antibodies are still being studied in early phase clinical trials. Perhaps a more comprehensive strategy of targeting the insulin-like growth factor-1 receptor network would be successful. For example, targeting receptor, ligand and downstream signaling molecules such as phosphatidylinositol 3′-kinase or particularly the insulin receptor substrate adapter proteins might result in a complete blockade of insulin-like growth factor-1 receptor/insulin receptor biological functions.
Collapse
Affiliation(s)
- Roudy Chiminch Ekyalongo
- Masonic Cancer Center, University of Minnesota, MMC 806, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, MMC 806, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
44
|
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: 14] [Impact Index Per Article: 2.0] [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
|
45
|
Huang D, Yang F, Wang Y, Guan X. Mechanisms of resistance to selective estrogen receptor down-regulator in metastatic breast cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:148-156. [PMID: 28344099 DOI: 10.1016/j.bbcan.2017.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/18/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023]
Abstract
Based on the prominent role estrogen receptor (ER) plays in breast cancer, endocrine therapy has been developed to block the ER pathway and has shown great effectiveness. Fulvestrant, the first selective ER down-regulator (SERD), was demonstrated to completely suppress ERα and notably efficient. However, resistance to fulvestrant occurs, either intrinsic or acquired during the treatment. Several potential mechanisms inducing fulvestrant resistance have been proposed, composed of activated ERα-independent compensatory growth factor signaling, stimulated downstream kinases, altered cell cycle mediators, etcetera. Experimentally, combinations of fulvestrant with targeted treatments were reported to eliminate the resistance and improve the effect of fulvestrant. Meanwhile, some clinical trials associated with the targeted combination therapies are in progress. This review focuses on the underlying mechanisms that contribute to fulvestrant resistance in ER-positive breast cancer and provides an overview of combined fulvestrant with targeted agents to shed light on optimal therapies for patients with ER-positive breast cancer.
Collapse
Affiliation(s)
- Doudou Huang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Yucai Wang
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China.
| |
Collapse
|
46
|
Cao H, Cui L, Ma W, Zhu L, Wang K, Ni Y, Wang Y, Du J. Adverse Events and Efficacy of Cixutumumab in Phase II Clinical Trials: A systematic Review and Meta-Analysis. Clin Drug Investig 2017; 37:135-153. [PMID: 27858328 DOI: 10.1007/s40261-016-0475-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Cixutumumab is a monoclonal antibody targeting insulin-like growth factor 1 receptor (IGF1R). We sought to evaluate the efficacy of cixutumumab in the treatment of cancer, and to comprehensively assess the associated adverse events in phase II clinical trials. METHODS Data were collected from PubMed, Embase, and Clinicaltrials.gov. The improvement on progression-free survival (PFS) was evaluated by hazard ratio (HR) and 95% confidence intervals (95% CIs). We also carried a meta-analysis to comprehensively evaluate the incidence of adverse events. RESULTS The adverse events that were mentioned most frequently were hyperglycemia, anemia, nausea, fatigue, and thrombocytopenia. The most frequent adverse events were hyponatremia (40.28%), fatigue (35.18%), and skin rash (35.11%). Results showed that cixutumumab treatments did not benefit PFS (HR 1.03, 95% CI 0.83-1.26, p = 0.979). The complete response (CR) was rarely seen in phase II trials. CONCLUSIONS Cixutumumab was well tolerated when used alone and in combination therapies, but its antitumor activity was low in the existing phase II clinical trials. An acceptable incidence of adverse effects supports further investigation of this drug, provided that it shows antitumor activity in combination with other drugs.
Collapse
Affiliation(s)
- Hongxin Cao
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China.,Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, 8, Jinan, Shandong, 250012, People's Republic of China
| | - Lixuan Cui
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Wei Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Linhai Zhu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Kai Wang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Yang Ni
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Yibing Wang
- Department of Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China. .,Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China.
| |
Collapse
|
47
|
THADA fusion is a mechanism of IGF2BP3 activation and IGF1R signaling in thyroid cancer. Proc Natl Acad Sci U S A 2017; 114:2307-2312. [PMID: 28193878 DOI: 10.1073/pnas.1614265114] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Thyroid cancer development is driven by known point mutations or gene fusions found in ∼90% of cases, whereas driver mutations in the remaining tumors are unknown. The insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) plays an important role in cancer, yet the mechanisms of its activation in cancer cells remain poorly understood. Using whole-transcriptome and whole-genome analyses, we identified a recurrent fusion between the thyroid adenoma-associated (THADA) gene on chromosome 2 and the LOC389473 gene on chromosome 7 located 12 kb upstream of the IGF2BP3 gene. We show that THADA fusion to LOC389473 and other regions in the vicinity does not result in the formation of a chimeric protein but instead leads to strong overexpression of the full-length IGF2BP3 mRNA and protein, increased IGF2 translation and IGF1 receptor (IGF1R) signaling via PI3K and MAPK cascades, and promotion of cell proliferation, invasion, and transformation. THADA fusions and IGF2BP3 overexpression are found in ∼5% of thyroid cancers that lack any other driver mutations. We also find that strong IGF2BP3 overexpression via gene fusion, amplification, or other mechanisms occurs in 5 to 15% of several other cancer types. Finally, we provide in vitro and in vivo evidence that growth of IGF2BP3-driven cells and tumors may be blocked by IGF1R inhibition, raising the possibility that IGF2BP3 overexpression in cancer cells may predict an anti-IGF1R benefit.
Collapse
|
48
|
Matsunaga Y, Adachi Y, Sasaki Y, Koide H, Motoya M, Nosho K, Takagi H, Yamamoto H, Sasaki S, Arimura Y, Tokino T, Carbone DP, Imai K, Shinomura Y. The effect of forced expression of mutated K-RASgene on gastrointestinal cancer cell lines and the IGF-1R targeting therapy. Mol Carcinog 2017; 56:515-526. [PMID: 27312358 DOI: 10.1002/mc.22513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Affiliation(s)
- Yasutaka Matsunaga
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Yasushi Adachi
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
- Sapporo Shirakaba-Dai Hospital; Sapporo Japan
| | - Yasushi Sasaki
- Medical Genome Sciences, Research Institute of Frontier Medicine; Sapporo Medical University; Sapporo Japan
| | - Hideyuki Koide
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Masayo Motoya
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Katsuhiko Nosho
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Hideyasu Takagi
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Hiroyuki Yamamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; St. Marianna University School of Medicine; Kawasaki Japan
| | - Shigeru Sasaki
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Yoshiaki Arimura
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| | - Takashi Tokino
- Medical Genome Sciences, Research Institute of Frontier Medicine; Sapporo Medical University; Sapporo Japan
| | - David P. Carbone
- James Cancer Center; The Ohio State University Medical Center; Columbus Ohio
| | - Kohzoh Imai
- The Institute of Medical Science Hospital; The University of Tokyo; Tokyo Japan
| | - Yasuhisa Shinomura
- Department of Gastroenterology, Rheumatology and Clinical Immunology; Sapporo Medical University; Sapporo Japan
| |
Collapse
|
49
|
Abstract
Growth hormone (GH) replacement in GH deficient (GHD) children secures normal linear growth, while in GHD adults it improves metabolic status, body composition and quality of life. Safety of GH treatment is an important issue in particular concerning the controversy of potential cancer risk. Unlike in congenital IGF-1 deficiency, there is no complete protection against cancer in GHD patients. Important modifiable risk factors in GHD patients are obesity, insulin resistance, sedentary behavior, circadian rhythm disruption, chronic low grade inflammation and concomitant sex hormone replacement. Age, family history, hereditary cancer predisposition syndromes or cranial irradiation may present non-modifiable risk factors. Quantifying the risk of cancer in relation to GH therapy in adult GHD patients is complex. There is evidence that links GH to cancer occurrence or promotion, but the evidence is progressively weaker when moving from in vitro studies to in vivo animal studies to epidemiological studies and finally to studies on GH treated patients. GH-IGF inhibition in experimental animals leads to decreased cancer incidence and progression. Epidemiological studies suggest an association of high normal circulating IGF-1 or GH to cancer incidence in general population. Data regarding cancer incidence in acromegaly are inconsistent but thyroid and colorectal neoplasias are the main source of concern. Replacement therapy with rhGH for GHD is generally safe. Overall the rate of de novo cancers was not increased in studies of GH-treated GHD patients. Additional caution is mandated in patients with history of cancer, strong family history of cancer and with advancing age. Childhood cancer survivors may be at increased risk for secondary neoplasms compared with general population. In this subgroup GH therapy should be used cautiously and with respect to other risk factors (cranial irradiation etc). We believe that the benefits of GH therapy against the morbidity of untreated GH deficiency outweigh the theoretical cancer risk. Proper monitoring of GH treatment with diligent cancer surveillance remains essential.
Collapse
Affiliation(s)
- Sandra Pekic
- University of Belgrade, School of Medicine, Dr Subotica 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, 11000 Belgrade, Serbia
| | - Marko Stojanovic
- University of Belgrade, School of Medicine, Dr Subotica 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, 11000 Belgrade, Serbia
| | - Vera Popovic
- University of Belgrade, School of Medicine, Dr Subotica 8, 11000 Belgrade, Serbia.
| |
Collapse
|
50
|
In vivo evaluation of IGF1R/IR PET ligand [ 18F]BMS-754807 in rodents. Bioorg Med Chem Lett 2017; 27:941-943. [PMID: 28094184 DOI: 10.1016/j.bmcl.2016.12.086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 11/21/2022]
Abstract
In vivo evaluation of [18F]BMS-754807 binding in mice and rats using microPET and biodistribution methods is described herein. The radioligand shows consistent binding characteristics, in vivo, in both species. Early time frames of the microPET images and time activity curves of brain indicate poor penetration of the tracer across the blood brain barrier (BBB) in both species. However, microPET experiments in mice and rats show high binding of the radioligand outside the brain to heart, pancreas and muscle, the organs known for higher expression of IGF1R/1R. Biodistribution analysis 2h after injection of [18F]BMS-754807 in rats show negligible [18F]defluorination as reflected by the low bone uptake and clearance from blood. Overall, the data indicate that [18F]BMS-754807 can potentially be a radiotracer for the quantification of IGF1R/IR outside the brain using PET.
Collapse
|