1
|
Dunston K, Hunter MI, Johannesen E, Jung JS, Kim TH, Yoo JY, Jeong JW. ERBB2 Targeting Reveals a Significant Suppression of Tumorigenesis in Murine Endometrial Cancer with Pten Mutation. Reprod Sci 2024; 31:2458-2467. [PMID: 38637476 DOI: 10.1007/s43032-024-01546-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Endometrial cancer is the most common gynecologic malignancy. PTEN is a negative regulator of PI3K signaling and is deficient in > 50% of primary human endometrial cancer. Amplification of ERBB2 promotes tumorigenesis and pathogenesis of several human cancers. However, the effect of ERBB2 targeting has not been studied in endometrial cancer with PTEN mutations. The murine model Pgrcre/+Erbb2f/fPtenf/f (Erbb2d/d Ptend/d) was developed to evaluate the effect of ERBB2 targeted therapy in endometrial cancer with PTEN deficiency. Histopathological and molecular analysis was performed for Ptend/d and Erbb2d/dPtend/d mice. Histopathological analysis revealed that Erbb2d/dPtend/d mice significantly reduced development and progression of endometrial cancer compared to Ptend/d mice. Furthermore, percentage of proliferative cells in Erbb2d/dPtend/d mice revealed anti-tumorigenic effect of Erbb2 ablation compared to Ptend/d mice. Our results demonstrate that Erbb2 ablation reveals a significant suppression of tumorigenesis on endometrial cancer of Ptend/d mice. Our results suggest that Erbb2 functions as an oncogene in endometrial cancer of Ptend/d mice implying that Erbb2 targeting can be used as an effective therapeutic approach for treatment of endometrial cancer with PTEN deficiency to hinder cancer development.
Collapse
Affiliation(s)
- Krystina Dunston
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Mark I Hunter
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Eric Johannesen
- Department of Pathology Medical Science Building (MSB), University of Missouri, Columbia, MO, USA
| | - Jin-Seok Jung
- Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, 1 Yonseidae-gil, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, 1 Yonseidae-gil, Wonju, Gangwon-do, 26493, Republic of Korea.
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA.
| |
Collapse
|
2
|
Yalçıntaş YM, Duman H, López JMM, Portocarrero ACM, Lombardo M, Khallouki F, Koch W, Bordiga M, El-Seedi H, Raposo A, Alves JLDB, Karav S. Revealing the Potency of Growth Factors in Bovine Colostrum. Nutrients 2024; 16:2359. [PMID: 39064802 PMCID: PMC11279796 DOI: 10.3390/nu16142359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Colostrum is a nutritious milk synthesized by mammals during the postpartum period, and its rich bioactive components has led to a global increase in the consumption of bovine colostrum as a supplement. Bovine colostrum contains key components such as immunoglobulins, oligosaccharides, lactoferrin and lysozyme. It is a special supplement source due to its natural, high bioavailability and high concentrations of growth factors. Growth factors are critical to many physiological functions, and considering its presence in the colostrum, further research must be conducted on its safe application in many bodily disorders. Growth factors contribute to wound healing, muscle and bone development, and supporting growth in children. Additionally, the molecular mechanisms have been explored, highlighting the growth factors roles in cell proliferation, tissue regeneration, and the regulation of immune responses. These findings are crucial for understanding the potential health effects of bovine colostrum, ensuring its safe use, and forming a basis for future clinical applications. This review article examines the growth factors concentration in bovine colostrum, their benefits, clinical studies, and molecular mechanisms.
Collapse
Affiliation(s)
- Yalçın Mert Yalçıntaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (Y.M.Y.); (H.D.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (Y.M.Y.); (H.D.)
| | - Jose M. Miranda López
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, 27002 Lugo, Spain; (J.M.M.L.); (A.C.M.P.)
| | - Alicia C. Mondragón Portocarrero
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, 27002 Lugo, Spain; (J.M.M.L.); (A.C.M.P.)
| | - Mauro Lombardo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy;
| | - Farid Khallouki
- Team of Ethnopharmacology and Pharmacognosy, Biology Department, Faculty of Sciences and Techniques, Moulay Ismail University of Meknes, Errachidia 50050, Morocco;
| | - Wojciech Koch
- Chair and Department of Food and Nutrition, Faculty of Pharmacy, Medical University of Lublin, 4a Chodźki Str., 20-093 Lublin, Poland;
| | - Matteo Bordiga
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy;
| | - Hesham El-Seedi
- Chemistry Department, Faculty of Science, Islamic University of Madinah, P.O. Box 170, Madinah 42351, Saudi Arabia;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal;
| | - Jose Luiz de Brito Alves
- Department of Nutrition, Health Science Center, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye; (Y.M.Y.); (H.D.)
| |
Collapse
|
3
|
Abrahams B, Gerber A, Hiss DC. Combination Treatment with EGFR Inhibitor and Doxorubicin Synergistically Inhibits Proliferation of MCF-7 Cells and MDA-MB-231 Triple-Negative Breast Cancer Cells In Vitro. Int J Mol Sci 2024; 25:3066. [PMID: 38474312 DOI: 10.3390/ijms25053066] [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: 02/19/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The role of the epidermal growth factor receptor (EGFR) in tumor progression and survival is often underplayed. Its expression and/or dysregulation is associated with disease advancement and poor patient outcome as well as drug resistance in breast cancer. EGFR is often overexpressed in breast cancer and particularly triple-negative breast cancer (TNBC), which currently lacks molecular targets. We examined the synergistic potential of an EGFR inhibitor (EGFRi) in combination with doxorubicin (Dox) in estrogen-positive (ER+) MCF-7 and MDA-MB-231 TNBC cell lines. The exposure of MDA-MB-231 and MCF-7 to EGFRi produced an IC50s of 6.03 µM and 3.96 µM, respectively. Dox induced MDA-MB-231 (IC50 9.67 µM) and MCF-7 (IC50 1.4 µM) cytotoxicity. Combinations of EGFRi-Dox significantly reduced the IC50 in MCF-7 (0.46 µM) and MBA-MB 231 (0.01 µM). Synergistic drug interactions in both cell lines were confirmed using the Bliss independence model. Pro-apoptotic Caspase-3/7 activation occurred in MCF-7 at 0.1-10 µM of EGFRi and Dox single treatments, whilst 1 μM Dox yielded a more potent effect on MDA-MB-231. EGFRi and Dox individually and in combination downregulated the EGFR gene expression in MCF-7 and MDA-MB-231 (p < 0.001). This study demonstrates EGFRi's potential for eliciting synergistic interactions with Dox, causing enhanced growth inhibition, apoptosis induction, and downregulation of EGFR in both cell lines.
Collapse
Affiliation(s)
- Beynon Abrahams
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Anthonie Gerber
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Donavon Charles Hiss
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Bellville 7535, South Africa
| |
Collapse
|
4
|
Lu T, Freytag L, Narayana VK, Moore Z, Oliver SJ, Valkovic A, Nijagal B, Peterson AL, de Souza DP, McConville MJ, Whittle JR, Best SA, Freytag S. Matrix Selection for the Visualization of Small Molecules and Lipids in Brain Tumors Using Untargeted MALDI-TOF Mass Spectrometry Imaging. Metabolites 2023; 13:1139. [PMID: 37999235 PMCID: PMC10673325 DOI: 10.3390/metabo13111139] [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: 09/26/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging allows for the study of metabolic activity in the tumor microenvironment of brain cancers. The detectable metabolites within these tumors are contingent upon the choice of matrix, deposition technique, and polarity setting. In this study, we compared the performance of three different matrices, two deposition techniques, and the use of positive and negative polarity in two different brain cancer types and across two species. Optimal combinations were confirmed by a comparative analysis of lipid and small-molecule abundance by using liquid chromatography-mass spectrometry and RNA sequencing to assess differential metabolites and enzymes between normal and tumor regions. Our findings indicate that in the tumor-bearing brain, the recrystallized α-cyano-4-hydroxycinnamic acid matrix with positive polarity offered superior performance for both detected metabolites and consistency with other techniques. Beyond these implications for brain cancer, our work establishes a workflow to identify optimal matrices for spatial metabolomics studies.
Collapse
Affiliation(s)
- Tianyao Lu
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne 3052, Australia
| | - Lutz Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
| | - Vinod K. Narayana
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne 3010, Australia
| | - Zachery Moore
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne 3052, Australia
| | - Shannon J. Oliver
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
| | - Adam Valkovic
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
| | - Brunda Nijagal
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne 3010, Australia
| | - Amanda L. Peterson
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne 3010, Australia
| | - David P. de Souza
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne 3010, Australia
| | - Malcolm J. McConville
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne 3010, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne 3010, Australia
| | - James R. Whittle
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne 3052, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne 3052, Australia
| | - Sarah A. Best
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne 3052, Australia
| | - Saskia Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne 3052, Australia
| |
Collapse
|
5
|
Terceiro LEL, Ikeogu NM, Lima MF, Edechi CA, Nickel BE, Fischer G, Leygue E, McManus KJ, Myal Y. Navigating the Blood-Brain Barrier: Challenges and Therapeutic Strategies in Breast Cancer Brain Metastases. Int J Mol Sci 2023; 24:12034. [PMID: 37569410 PMCID: PMC10418424 DOI: 10.3390/ijms241512034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer (BC) is the most common cancer in women, with metastatic BC being responsible for the highest number of deaths. A frequent site for BC metastasis is the brain. Brain metastasis derived from BC involves the cooperation of multiple genetic, epigenetic, angiogenic, and tumor-stroma interactions. Most of these interactions provide a unique opportunity for development of new therapeutic targets. Potentially targetable signaling pathways are Notch, Wnt, and the epidermal growth factor receptors signaling pathways, all of which are linked to driving BC brain metastasis (BCBM). However, a major challenge in treating brain metastasis remains the blood-brain barrier (BBB). This barrier restricts the access of unwanted molecules, cells, and targeted therapies to the brain parenchyma. Moreover, current therapies to treat brain metastases, such as stereotactic radiosurgery and whole-brain radiotherapy, have limited efficacy. Promising new drugs like phosphatase and kinase modulators, as well as BBB disruptors and immunotherapeutic strategies, have shown the potential to ease the disease in preclinical studies, but remain limited by multiple resistance mechanisms. This review summarizes some of the current understanding of the mechanisms involved in BC brain metastasis and highlights current challenges as well as opportunities in strategic designs of potentially successful future therapies.
Collapse
Affiliation(s)
- Lucas E. L. Terceiro
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Nnamdi M. Ikeogu
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada;
| | - Matheus F. Lima
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Chidalu A. Edechi
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Barbara E. Nickel
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Gabor Fischer
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Etienne Leygue
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (E.L.); (K.J.M.)
| | - Kirk J. McManus
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (E.L.); (K.J.M.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Yvonne Myal
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
6
|
Geoerger B, Marshall LV, Nysom K, Makin G, Bouffet E, Defachelles AS, Amoroso L, Aerts I, Leblond P, Barahona P, Van-Vlerken K, Fu E, Solca F, Lorence RM, Ziegler DS. Afatinib in paediatric patients with recurrent/refractory ErbB-dysregulated tumours: Results of a phase I/expansion trial. Eur J Cancer 2023; 188:8-19. [PMID: 37178647 DOI: 10.1016/j.ejca.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 05/15/2023]
Abstract
AIM This phase I/expansion study assessed the safety, pharmacokinetics and preliminary antitumor activity of afatinib in paediatric patients with cancer. METHODS The dose-finding part enroled patients (2-<18 years) with recurrent/refractory tumours. Patients received 18 or 23 mg/m2/d afatinib orally (tablet or solution) in 28-d cycles. In the maximum tolerated dose (MTD) expansion, eligible patients (1-<18 years) had tumours fulfilling ≥2 of the following criteria in the pre-screening: EGFR amplification; HER2 amplification; EGFR membrane staining (H-score>150); HER2 membrane staining (H-score>0). The primary end-points were dose-limiting toxicities (DLTs), afatinib exposure, and objective response. RESULTS Of 564 patients pre-screened, 536 patients had biomarker data and 63 (12%) fulfilled ≥2 EGFR/HER2 criteria required for inclusion in the expansion part. A total of 56 patients were treated (17 in the dose-finding and 39 in the expansion part). DLTs were observed in one of six MTD-evaluable patients receiving 18 mg/m²/d and in two of five MTD-evaluable patients receiving 23 mg/m²/d; 18 mg/m²/d was defined as the MTD. There were no new safety signals. Pharmacokinetics confirmed exposure consistent with the approved dose in adults. One partial response (-81% per Response Assessment in Neuro-Oncology) was observed in a patient with a glioneuronal tumour harbouring a CLIP2::EGFR fusion; unconfirmed partial responses were observed in two patients. In total, 25% of patients experienced objective response or stable disease (95% confidence interval: 14-38). CONCLUSION Targetable EGFR/HER2 drivers are rare in paediatric cancers. Treatment with afatinib led to a durable response (>3 years) in one patient with a glioneuronal tumour with CLIP2::EGFR fusion.
Collapse
Affiliation(s)
- Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Pediatric and Adolescent Oncology, INSERM U1015, Université Paris-Saclay, Villejuif, France.
| | - Lynley V Marshall
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, UK
| | - Karsten Nysom
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Guy Makin
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK; Royal Manchester Children's Hospital, Manchester, UK
| | - Eric Bouffet
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | - Isabelle Aerts
- Institut Curie, PSL Research University, Oncology Center SIREDO, Paris, France
| | - Pierre Leblond
- Institute of Pediatric Hematology and Oncology, Centre Léon Bérard, Lyon, France
| | | | | | - Eric Fu
- Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, CT, USA
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co.KG Vienna, Austria
| | | | - David S Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia; Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
7
|
Valadan R, Dabiri M, Tehrani M, Hashemi Tabar G, Rafiei A. A cell-based subtractive panning strategy for selection of conformation-specific single-chain variable-fragment (scFv) against dimerization domain of EGFR. J Immunol Methods 2023; 515:113456. [PMID: 36898519 DOI: 10.1016/j.jim.2023.113456] [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: 12/28/2022] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND AND OBJECTIVE Overexpression of EGFR, a member of the ErbB receptor family, has been observed in several cancers and causes resistance to therapeutic antibodies, such as Herceptin. In this study, we produced a recombinant single-chain variable fragment (scFv) antibody against the EGFR dimerization domain. METHODS The recombinant scFv was generated using a cell-based subtractive panning strategy. Subtractive panning was performed on a genetically engineered, VERO/EGFR, cells as well as a triple-negative breast cancer, MDA-MB-468, cells. Phage cell-ELISA was used to monitor the binding of the selected scFvs to the dimerization domain of EGFR. Inhibition of EGFR and HER2 dimerization by the produced scFvs were finally evaluated using the dimerization inhibition test and the expression of apoptosis-related genes were measured using the quantitative RT-PCR. RESULTS PCR fingerprinting results showed a uniform digestion pattern following the third round of panning that confirmed the success of subtractive panning. Moreover, cell-ELISA validated the reactivity of the produced scFvs to EGFR following stimulation with EGF. Dimerization inhibition test showed the capacity of the scFvs to inhibit EGFR and HER2 dimerization. Investigation of apoptosis-related genes showed that treatment with the scFv antibody caused increased Bax and decreased Bcl2 expression. CONCLUSIONS Directed HER2 targeting was shown to be effective enough to block the functional domain of the cell receptor and its intracellular signaling pathway. The subtractive panning strategy used in this study could control the process of directed selection of specific antibodies against the dimerization domain of EGFR. Selected antibodies might then be functionally tested for antitumor effects in both in vitro and in vivo studies.
Collapse
Affiliation(s)
- Reza Valadan
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mina Dabiri
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Gholamreza Hashemi Tabar
- Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Pathobiology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Alireza Rafiei
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran..
| |
Collapse
|
8
|
Xia T, Liang X, Liu CS, Hu YN, Luo ZY, Tan XM. Network Pharmacology Integrated with Transcriptomics Analysis Reveals Ermiao Wan Alleviates Atopic Dermatitis via Suppressing MAPK and Activating the EGFR/AKT Signaling. Drug Des Devel Ther 2022; 16:4325-4341. [PMID: 36578822 PMCID: PMC9790806 DOI: 10.2147/dddt.s384927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background Ermiao Wan (EMW) is commonly used to treat atopic dermatitis (AD) in China. However, the pharmacological mechanisms underlying the action of EMW against AD remain unclear. Purpose We aimed to determine the mechanisms underlying the effectiveness of EMW in the treatment of AD. Methods We evaluated the effect of EMW on AD induced by dinitrochlorobenzene (DNCB) in BALB/C mice. To clarify the key components of EMW in AD treatment, the main components of EMW were identified using HPLC. Serum pharmacochemistry was used to analyze the absorbed ingredients from blood. Based on the phytochemical results, network pharmacology and molecular docking were used to predict the action of EMW. Skin transcriptomic analysis was used to validate the network pharmacology results. RT-qPCR,ELISA, and immunohistochemical were performed to validate the results of skin transcriptomics. Results EMW improved the symptoms of AD, with less rashes, less spontaneous scratching, less inflammatory cell infiltration, and fewer allergic reactions. The established HPLC method is simple and reliable. Chlorogenic acid, phellodendrine, magnoflorine, jatrorrhizine, palmatine, berberine, and atractylodin were the key effective ingredients with a high blood concentration. Fifty-seven primary causal targets of EMW against AD were identified. These targets are mainly involved in ErbB signaling pathways including EGFR, AKT1, MAPK8, JUN, MAPK1. Molecular docking showed that EGFR, AKT1, MAPK8, JUN, MAPK1 had good binding force with EMW. In AD mice, EMW regulated the EGFR/AKT signaling through upregulation of Grb2, GAB1, Raf-1, EGFR, and AKT, and downregulation of MAPK1 and JUN, compared to that in the MD group. Conclusion EMW could alleviate AD through activating EGFR/AKT signaling and suppressing MAPK. This study provides a theoretical basis for the clinical use of EMW.
Collapse
Affiliation(s)
- Ting Xia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Xiao Liang
- School of Pharmaceutical Sciences, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Chang-Shun Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Yan-Nan Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People’s Republic of China,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, People’s Republic of China,Correspondence: Xiao-Mei Tan, Tel/Fax + 86-020-61648265, Email
| |
Collapse
|
9
|
An oncogene addiction phosphorylation signature and its derived scores inform tumor responsiveness to targeted therapies. Cell Mol Life Sci 2022; 80:6. [PMID: 36494469 PMCID: PMC9734221 DOI: 10.1007/s00018-022-04634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Oncogene addiction provides important therapeutic opportunities for precision oncology treatment strategies. To date the cellular circuitries associated with driving oncoproteins, which eventually establish the phenotypic manifestation of oncogene addiction, remain largely unexplored. Data suggest the DNA damage response (DDR) as a central signaling network that intersects with pathways associated with deregulated addicting oncoproteins with kinase activity in cancer cells. EXPERIMENTAL DESIGN: We employed a targeted mass spectrometry approach to systematically explore alterations in 116 phosphosites related to oncogene signaling and its intersection with the DDR following inhibition of the addicting oncogene alone or in combination with irradiation in MET-, EGFR-, ALK- or BRAF (V600)-positive cancer models. An NSCLC tissue pipeline combining patient-derived xenografts (PDXs) and ex vivo patient organotypic cultures has been established for treatment responsiveness assessment. RESULTS We identified an 'oncogene addiction phosphorylation signature' (OAPS) consisting of 8 protein phosphorylations (ACLY S455, IF4B S422, IF4G1 S1231, LIMA1 S490, MYCN S62, NCBP1 S22, P3C2A S259 and TERF2 S365) that are significantly suppressed upon targeted oncogene inhibition solely in addicted cell line models and patient tissues. We show that the OAPS is present in patient tissues and the OAPS-derived score strongly correlates with the ex vivo responses to targeted treatments. CONCLUSIONS We propose a score derived from OAPS as a quantitative measure to evaluate oncogene addiction of cancer cell samples. This work underlines the importance of protein phosphorylation assessment for patient stratification in precision oncology and corresponding identification of tumor subtypes sensitive to inhibition of a particular oncogene.
Collapse
|
10
|
Bioimaging Nucleic-Acid Aptamers with Different Specificities in Human Glioblastoma Tissues Highlights Tumoral Heterogeneity. Pharmaceutics 2022; 14:pharmaceutics14101980. [PMID: 36297416 PMCID: PMC9609998 DOI: 10.3390/pharmaceutics14101980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Nucleic-acid aptamers are of strong interest for diagnosis and therapy. Compared with antibodies, they are smaller, stable upon variations in temperature, easy to modify, and have higher tissue-penetration abilities. However, they have been little described as detection probes in histology studies of human tissue sections. In this study, we performed fluorescence imaging with two aptamers targeting cell-surface receptors EGFR and integrin α5β1, both involved in the aggressiveness of glioblastoma. The aptamers’ cell-binding specificities were confirmed using confocal imaging. The affinities of aptamers for glioblastoma cells expressing these receptors were in the 100–300 nM range. The two aptamers were then used to detect EGFR and integrin α5β1 in human glioblastoma tissues and compared with antibody labeling. Our aptafluorescence assays proved to be able to very easily reveal, in a one-step process, not only inter-tumoral glioblastoma heterogeneity (differences observed at the population level) but also intra-tumoral heterogeneity (differences among cells within individual tumors) when aptamers with different specificities were used simultaneously in multiplexing labeling experiments. The discussion also addresses the strengths and limitations of nucleic-acid aptamers for biomarker detection in histology.
Collapse
|
11
|
Yuan Y, Zhou S, Li C, Zhang X, Mao H, Chen W, Jiang X. Cascade Downregulation of the HER Family by a Dual-Targeted Recombinant Protein-Drug Conjugate to Inhibit Tumor Growth and Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201558. [PMID: 35365900 DOI: 10.1002/adma.202201558] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Human epidermal growth factor receptor type 2 (HER2)-targeted therapy can significantly improve the outcome of patients with HER2 positive cancer. However, relapse after this treatment remains a great challenge in the clinic due to tumor resistance, in which the HER network induces constitutive signal transduction. In addition, integrin receptors in the tumor extracellular matrix can mitigate the therapeutic effect of inhibitors to the growth factors receptors and tyrosine kinases. Here, the development of a recombinant protein (RP-HI) and its drug conjugates (RPDC-HI) to target both HER2 and integrin is reported. When simultaneously blocking HER2 and integrin by RP-HI, functions of the HER family and their interactions with the integrin are disrupted by downregulated expressions of HER family members, leading to inhibition of several downstream signal pathways. In combination with targeted delivery of the anticancer agent, doxorubicin (DOX), RPDC-HI significantly improves the tumor inhibition efficacy to 97.5% in treating HER2-positive breast cancer, comparing to 34.3% for free DOX. RPDC-HI shows even better antitumor efficiency than a monoclonal antibody, trastuzumab, when treating larger tumors. The developed dual-targeted RPDC platform offers a new and promising strategy for treating HER2-positive patients with synergistic therapeutic effects against tumor resistance to the conventional HER2-targeted treatment.
Collapse
Affiliation(s)
- Yang Yuan
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Sensen Zhou
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Cheng Li
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Xiaoke Zhang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
12
|
Cranberry Polyphenols in Esophageal Cancer Inhibition: New Insights. Nutrients 2022; 14:nu14050969. [PMID: 35267943 PMCID: PMC8912450 DOI: 10.3390/nu14050969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/31/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is a cancer characterized by rapidly rising incidence and poor survival, resulting in the need for new prevention and treatment options. We utilized two cranberry polyphenol extracts, one proanthocyanidin enriched (C-PAC) and a combination of anthocyanins, flavonoids, and glycosides (AFG) to assess inhibitory mechanisms utilizing premalignant Barrett’s esophagus (BE) and EAC derived cell lines. We employed reverse phase protein arrays (RPPA) and Western blots to examine cancer-associated pathways and specific signaling cascades modulated by C-PAC or AFG. Viability results show that C-PAC is more potent than AFG at inducing cell death in BE and EAC cell lines. Based on the RPPA results, C-PAC significantly modulated 37 and 69 proteins in JH-EsoAd1 (JHAD1) and OE19 EAC cells, respectively. AFG treatment significantly altered 49 proteins in both JHAD1 and OE19 cells. Bioinformatic analysis of RPPA results revealed many previously unidentified pathways as modulated by cranberry polyphenols including NOTCH signaling, immune response, and epithelial to mesenchymal transition. Collectively, these results provide new insight regarding mechanisms by which cranberry polyphenols exert cancer inhibitory effects targeting EAC, with implications for potential use of cranberry constituents as cancer preventive agents.
Collapse
|
13
|
Varlet P, Bouffet E, Casanova M, Giangaspero F, Antonelli M, Hargrave D, Ladenstein R, Pearson A, Hawkins C, König FB, Rüschoff J, Schmauch C, Bühnemann C, Garin-Chesa P, Schweifer N, Uttenreuther-Fischer M, Gibson N, Ittrich C, Krämer N, Solca F, Stolze B, Geoerger B. Comprehensive analysis of the ErbB receptor family in pediatric nervous system tumors and rhabdomyosarcoma. Pediatr Blood Cancer 2022; 69:e29316. [PMID: 34546642 DOI: 10.1002/pbc.29316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/05/2021] [Accepted: 08/01/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND There is a paucity of knowledge regarding pediatric biomarkers, including the relevance of ErbB pathway aberrations in pediatric tumors. We investigated the occurrence of ErbB receptor aberrations across different pediatric malignancies, to identify patterns of ErbB dysregulation and define biomarkers suitable for patient enrichment in clinical studies. PROCEDURE Tissue samples from 297 patients with nervous system tumors and rhabdomyosarcoma were analyzed for immunohistochemical expression or gene amplification of epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2). Exploratory analyses of HER3/HER4 expression, and mRNA expression of ErbB receptors/ligands (NanoString) were performed. Assay validation followed general procedures, with additional validation to address Clinical Laboratory Improvement Amendments (CLIA) requirements. RESULTS In most tumor types, samples with high ErbB receptor expression were found with heterogeneous distribution. We considered increased/aberrant ErbB pathway activation when greater than or equal to two EGFR/HER2 markers were simultaneously upregulated. ErbB pathway dysregulation was identified in ∼20%-30% of samples for most tumor types (medulloblastoma/primitive neuroectodermal tumors 31.1%, high-grade glioma 27.1%, neuroblastoma 22.7%, rhabdomyosarcoma 23.1%, ependymoma 18.8%), 4.2% of diffuse intrinsic pontine gliomas, and no recurrent or refractory low-grade astrocytomas. In medulloblastoma/primitive neuroectodermal tumors and neuroblastoma, this was attributed mainly to high EGFR polysomy/HER2 amplification, whereas EGFR gene amplification was observed in some high-grade glioma samples. EGFR/HER2 overexpression was most prevalent in ependymoma. CONCLUSIONS Overexpression and/or amplification of EGFR/HER2 were identified as potential enrichment biomarkers for clinical trials of ErbB-targeted drugs.
Collapse
Affiliation(s)
- Pascale Varlet
- GHU Psychiatrie et Neurosciences, site Sainte-Anne, service de Neuropathologie, Paris, France
| | - Eric Bouffet
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | - Darren Hargrave
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ruth Ladenstein
- Department of Paediatrics, St. Anna Children's Cancer Research Institute, Medical University, Vienna, Austria
| | - Andy Pearson
- Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK.,Division of Clinical Studies, Institute of Cancer Research, London, UK
| | | | | | | | | | | | - Pilar Garin-Chesa
- Staburo GmbH, Munich, Germany, on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Norbert Schweifer
- Staburo GmbH, Munich, Germany, on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Neil Gibson
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Carina Ittrich
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nicole Krämer
- Staburo GmbH, Munich, Germany, on behalf of Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Britta Stolze
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, INSERM U1015, Université Paris Saclay, Villejuif, France
| |
Collapse
|
14
|
Cheng WL, Feng PH, Lee KY, Chen KY, Sun WL, Van Hiep N, Luo CS, Wu SM. The Role of EREG/EGFR Pathway in Tumor Progression. Int J Mol Sci 2021; 22:ijms222312828. [PMID: 34884633 PMCID: PMC8657471 DOI: 10.3390/ijms222312828] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Aberrant activation of the epidermal growth factor receptor (EGFR/ERBB1) by erythroblastic leukemia viral oncogene homolog (ERBB) ligands contributes to various tumor malignancies, including lung cancer and colorectal cancer (CRC). Epiregulin (EREG) is one of the EGFR ligands and is low expressed in most normal tissues. Elevated EREG in various cancers mainly activates EGFR signaling pathways and promotes cancer progression. Notably, a higher EREG expression level in CRC with wild-type Kirsten rat sarcoma viral oncogene homolog (KRAS) is related to better efficacy of therapeutic treatment. By contrast, the resistance of anti-EGFR therapy in CRC was driven by low EREG expression, aberrant genetic mutation and signal pathway alterations. Additionally, EREG overexpression in non-small cell lung cancer (NSCLC) is anticipated to be a therapeutic target for EGFR-tyrosine kinase inhibitor (EGFR-TKI). However, recent findings indicate that EREG derived from macrophages promotes NSCLC cell resistance to EGFR-TKI treatment. The emerging events of EREG-mediated tumor promotion signals are generated by autocrine and paracrine loops that arise from tumor epithelial cells, fibroblasts, and macrophages in the tumor microenvironment (TME). The TME is a crucial element for the development of various cancer types and drug resistance. The regulation of EREG/EGFR pathways depends on distinct oncogenic driver mutations and cell contexts that allows specific pharmacological targeting alone or combinational treatment for tailored therapy. Novel strategies targeting EREG/EGFR, tumor-associated macrophages, and alternative activation oncoproteins are under development or undergoing clinical trials. In this review, we summarize the clinical outcomes of EREG expression and the interaction of this ligand in the TME. The EREG/EGFR pathway may be a potential target and may be combined with other driver mutation targets to combat specific cancers.
Collapse
Affiliation(s)
- Wan-Li Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan;
- Division of Cardiovascular Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Nguyen Van Hiep
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Synergism of Proneurogenic miRNAs Provides a More Effective Strategy to Target Glioma Stem Cells. Cancers (Basel) 2021; 13:cancers13020289. [PMID: 33466745 PMCID: PMC7831004 DOI: 10.3390/cancers13020289] [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: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary miRNAs function as critical regulators of gene expression and have been defined as contributors of cancer phenotypes by acting as oncogenes or tumor suppressors. Based on these findings, miRNA-based therapies have been explored in the treatment of many different malignancies. The use of single miRNAs has faced some challenges and showed limited success. miRNAs cooperate to regulate distinct biological processes and pathways and, therefore, combination of related miRNAs could amplify the repression of oncogenic factors and the effect on cancer relevant pathways. We established that the combination of tumor suppressor miRNAs miR-124, miR-128, and miR-137 is much more effective than single miRNAs in disrupting proliferation and survival of glioma stem cells and neuroblastoma lines and promoting differentiation and response to radiation. Subsequent genomic analyses showed that other combinations of tumor suppressor miRNAs could be equally effective, and its use could provide new routes to target in special cancer-initiating cell populations. Abstract Tumor suppressor microRNAs (miRNAs) have been explored as agents to target cancer stem cells. Most strategies use a single miRNA mimic and present many disadvantages, such as the amount of reagent required and the diluted effect on target genes. miRNAs work in a cooperative fashion to regulate distinct biological processes and pathways. Therefore, we propose that miRNA combinations could provide more efficient ways to target cancer stem cells. We have previously shown that miR-124, miR-128, and miR-137 function synergistically to regulate neurogenesis. We used a combination of these three miRNAs to treat glioma stem cells and showed that this treatment was much more effective than single miRNAs in disrupting cell proliferation and survival and promoting differentiation and response to radiation. Transcriptomic analyses indicated that transcription regulation, angiogenesis, metabolism, and neuronal differentiation are among the main biological processes affected by transfection of this miRNA combination. In conclusion, we demonstrated the value of using combinations of neurogenic miRNAs to disrupt cancer phenotypes and glioma stem cell growth. The synergistic effect of these three miRNA amplified the repression of oncogenic factors and the effect on cancer relevant pathways. Future therapeutic approaches would benefit from utilizing miRNA combinations, especially when targeting cancer-initiating cell populations.
Collapse
|
16
|
Sarne V, Huter S, Braunmueller S, Rakob L, Jacobi N, Kitzwögerer M, Wiesner C, Obrist P, Seeboeck R. Promoter Methylation of Selected Genes in Non-Small-Cell Lung Cancer Patients and Cell Lines. Int J Mol Sci 2020; 21:E4595. [PMID: 32605217 PMCID: PMC7369760 DOI: 10.3390/ijms21134595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 01/03/2023] Open
Abstract
Specific gene promoter DNA methylation is becoming a powerful epigenetic biomarker in cancer diagnostics. Five genes (CDH1, CDKN2Ap16, RASSF1A, TERT, and WT1) were selected based on their frequently published potential as epigenetic markers. Diagnostic promoter methylation assays were generated based on bisulfite-converted DNA pyrosequencing. The methylation patterns of 144 non-small-cell lung cancer (NSCLC) and 7 healthy control formalin-fixed paraffin-embedded (FFPE) samples were analyzed to evaluate the applicability of the putative diagnostic markers. Statistically significant changes in methylation levels are shown for TERT and WT1. Furthermore, 12 NSCLC and two benign lung cell lines were characterized for promoter methylation. The in vitro tests involved a comparison of promoter methylation in 2D and 3D cultures, as well as therapeutic tests investigating the impact of CDH1/CDKN2Ap16/RASSF1A/TERT/WT1 promoter methylation on sensitivity to tyrosine kinase inhibitor (TKI) and DNA methyl-transferase inhibitor (DNMTI) treatments. We conclude that the selected markers have potential and putative impacts as diagnostic or even predictive marker genes, although a closer examination of the resulting protein expression and pathway regulation is needed.
Collapse
MESH Headings
- Aged
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- DNA Methylation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Prognosis
- Promoter Regions, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Victoria Sarne
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Samuel Huter
- Pathologylab Dr. Obrist & Dr. Brunhuber OG, 6511 Zams, Austria; (S.H.); (P.O.)
| | - Sandrina Braunmueller
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Lisa Rakob
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Nico Jacobi
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Melitta Kitzwögerer
- Clinical Institute of Pathology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, 3100 St. Pölten, Austria;
| | - Christoph Wiesner
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Peter Obrist
- Pathologylab Dr. Obrist & Dr. Brunhuber OG, 6511 Zams, Austria; (S.H.); (P.O.)
| | - Rita Seeboeck
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
- Clinical Institute of Pathology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, 3100 St. Pölten, Austria;
| |
Collapse
|
17
|
Li H, Hu L, Zhang H, Wang D. ErbB1 and ErbB2 overexpression in patients with sinonasal inverted papilloma and inverted papilloma with squamous cell carcinoma in China. Acta Otolaryngol 2019; 139:1104-1111. [PMID: 31556771 DOI: 10.1080/00016489.2019.1650200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Currently, the expression patterns of epidermal growth factor receptor (EGFR) family genes in sinonasal inverted papilloma (SNIP) and inverted papilloma with squamous cell carcinoma (IPwSCC) are not clear.Objective: This study aimed to investigate the expression of EGFR family members and their ligands in SNIP and IPwSCC and to analyze their correlations with SNIP histological grade and Krouse stage.Materials and methods: Data from 25 cases of inverted papilloma patients in China were collected and divided into 16 cases in the SNIP group and 9 in the IPwSCC group. In addition, eight cases of normal nasal mucosa (NNM) were collected and used as the control group. The expression levels of EGFR family members and their ligands in the NNM and SNIP groups and EGFR family members in the IPwSCC group were evaluated using immunohistochemistry and qRT-PCR. In addition, their correlations with the SNIP histological grade and Krouse stage were analyzed. The statistical analysis was performed using the GraphPad Prism 7.0 statistical software.Results: The ErbB1 and ErbB2 mRNA and protein expression levels were significantly higher in the SNIP group than in the NNM group (p < .01). The ErbB1 and ErbB2 protein expression levels were significantly higher in the IPwSCC group than those in the NNM and SNIP groups (p < .01). The ErbB1 and ErbB2 mRNA and protein expression levels in the SNIP group were positively correlated with the SNIP dysplasia grade.Conclusion: Upregulation of ErbB1 and ErbB2 expression may be associated with SNIP pathogenesis and carcinogenesis.
Collapse
Affiliation(s)
- Hongbing Li
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Li Hu
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China
- Department of Research Centre, Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Huankang Zhang
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Dehui Wang
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China
| |
Collapse
|
18
|
Fechter P, Cruz Da Silva E, Mercier MC, Noulet F, Etienne-Seloum N, Guenot D, Lehmann M, Vauchelles R, Martin S, Lelong-Rebel I, Ray AM, Seguin C, Dontenwill M, Choulier L. RNA Aptamers Targeting Integrin α5β1 as Probes for Cyto- and Histofluorescence in Glioblastoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:63-77. [PMID: 31226519 PMCID: PMC6586995 DOI: 10.1016/j.omtn.2019.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 02/07/2023]
Abstract
Nucleic acid aptamers are often referred to as chemical antibodies. Because they possess several advantages, like their smaller size, temperature stability, ease of chemical modification, lack of immunogenicity and toxicity, and lower cost of production, aptamers are promising tools for clinical applications. Aptamers against cell surface protein biomarkers are of particular interest for cancer diagnosis and targeted therapy. In this study, we identified and characterized RNA aptamers targeting cells expressing integrin α5β1. This αβ heterodimeric cell surface receptor is implicated in tumor angiogenesis and solid tumor aggressiveness. In glioblastoma, integrin α5β1 expression is associated with an aggressive phenotype and a decrease in patient survival. We used a complex and original hybrid SELEX (selective evolution of ligands by exponential enrichment) strategy combining protein-SELEX cycles on the recombinant α5β1 protein, surrounded by cell-SELEX cycles using two different cell lines. We identified aptamer H02, able to differentiate, in cyto- and histofluorescence assays, glioblastoma cell lines, and tissues from patient-derived tumor xenografts according to their α5 expression levels. Aptamer H02 is therefore an interesting tool for glioblastoma tumor characterization.
Collapse
Affiliation(s)
- Pierre Fechter
- CNRS, UMR 7242, Biotechnologie et Signalisation Cellulaire, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Elisabete Cruz Da Silva
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Marie-Cécile Mercier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Fanny Noulet
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Nelly Etienne-Seloum
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; Département de Pharmacie, Centre de Lutte Contre le Cancer Paul Strauss, 67000 Strasbourg, France
| | - Dominique Guenot
- EA 3430, Progression Tumorale et Micro-environnement, Approches Translationnelles et Épidémiologie, Université de Strasbourg, 67000 Strasbourg, France
| | - Maxime Lehmann
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Romain Vauchelles
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Sophie Martin
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Isabelle Lelong-Rebel
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Anne-Marie Ray
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Cendrine Seguin
- CNRS, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Monique Dontenwill
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Laurence Choulier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France.
| |
Collapse
|
19
|
Zhu H, Zhao L, Li Z, Wen B, Qiu C, Liu M, Xu Z, Hu S, Li H. Preparation and characterization of humanized nanobodies targeting the dimer interface of epidermal growth factor receptor (EGFR). Protein Expr Purif 2019; 157:57-62. [DOI: 10.1016/j.pep.2019.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/04/2019] [Indexed: 01/06/2023]
|
20
|
Shen H, Yang N, Truskinovsky A, Chen Y, Mussell AL, Nowak NJ, Kobzik L, Frangou C, Zhang J. Targeting TAZ-Driven Human Breast Cancer by Inhibiting a SKP2-p27 Signaling Axis. Mol Cancer Res 2018; 17:250-262. [PMID: 30237296 DOI: 10.1158/1541-7786.mcr-18-0332] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/05/2018] [Accepted: 08/31/2018] [Indexed: 12/21/2022]
Abstract
Deregulated expression of the transcriptional coactivator with PDZ-binding motif (WWTR1/TAZ) is a common feature of basal-like breast cancer (BLBC). Yet, how oncogenic TAZ regulates cell-cycle progression and proliferation in breast cancer remains poorly understood, and whether TAZ is required for tumor maintenance has not been established. Here, using an integrative oncogenomic approach, TAZ-dependent cellular programs essential for tumor growth and progression were identified. Significantly, TAZ-driven tumor cells required sustained TAZ expression, given that its withdrawal impaired both genesis and maintenance of solid tumors. Moreover, temporal inhibition of TAZ diminished the metastatic burden in established macroscopic pulmonary metastases. Mechanistic investigation revealed that TAZ controls distinct gene profiles that determine cancer cell fate through cell-cycle networks, including a specific, causal role for S-phase kinase-associated protein 2 (SKP2) in mediating the neoplastic state. Together, this study elucidates the molecular events that underpin the role of TAZ in BLBC and link to SKP2, a convergent communication node for multiple cancer signaling pathways, as a key downstream effector molecule. IMPLICATIONS: Understanding the molecular role of TAZ and its link to SKP2, a signaling convergent point and key regulator in BLBC, represents an important step toward the identification of novel therapeutic targets for TAZ-dependent breast cancer.
Collapse
Affiliation(s)
- He Shen
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, New York
| | - Nuo Yang
- Department of Anesthesiology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, New York, New York
| | | | - Yanmin Chen
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, New York
| | - Ashley L Mussell
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, New York
| | - Norma J Nowak
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, New York, New York
| | - Lester Kobzik
- Harvard TH Chan School of Public Health, Molecular and Integrative Physiological Sciences, Boston, Massachusetts
| | - Costa Frangou
- Harvard TH Chan School of Public Health, Molecular and Integrative Physiological Sciences, Boston, Massachusetts.
| | - Jianmin Zhang
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, New York.
| |
Collapse
|
21
|
Chandrasekar A, Olde Heuvel F, Wepler M, Rehman R, Palmer A, Catanese A, Linkus B, Ludolph A, Boeckers T, Huber-Lang M, Radermacher P, Roselli F. The Neuroprotective Effect of Ethanol Intoxication in Traumatic Brain Injury Is Associated with the Suppression of ErbB Signaling in Parvalbumin-Positive Interneurons. J Neurotrauma 2018; 35:2718-2735. [PMID: 29774782 DOI: 10.1089/neu.2017.5270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ethanol intoxication (EI) is a frequent comorbidity of traumatic brain injury (TBI), but the impact of EI on TBI pathogenic cascades and prognosis is unclear. Although clinical evidence suggests that EI may have neuroprotective effects, experimental support is, to date, inconclusive. We aimed at elucidating the impact of EI on TBI-associated neurological deficits, signaling pathways, and pathogenic cascades in order to identify new modifiers of TBI pathophysiology. We have shown that ethanol administration (5 g/kg) before trauma enhances behavioral recovery in a weight-drop TBI model. Neuronal survival in the injured somatosensory cortex was also enhanced by EI. We have used phospho-receptor tyrosine kinase (RTK) arrays to screen the impact of ethanol on TBI-induced activation of RTK in somatosensory cortex, identifying ErbB2/ErbB3 among the RTKs activated by TBI and suppressed by ethanol. Phosphorylation of ErbB2/3/4 RTKs were upregulated in vGlut2+ excitatory synapses in the injured cortex, including excitatory synapses located on parvalbumin (PV)-positive interneurons. Administration of selective ErbB inhibitors was able to recapitulate, to a significant extent, the neuroprotective effects of ethanol both in sensorimotor performance and structural integrity. Further, suppression of PV interneurons in somatosensory cortex before TBI, by engineered receptors with orthogonal pharmacology, could mimic the beneficial effects of ErbB inhibitors. Thus, we have shown that EI interferes with TBI-induced pathogenic cascades at multiple levels, with one prominent pathway, involving ErbB-dependent modulation of PV interneurons.
Collapse
Affiliation(s)
| | | | - Martin Wepler
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Rida Rehman
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Annette Palmer
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Alberto Catanese
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Birgit Linkus
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Albert Ludolph
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Tobias Boeckers
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Markus Huber-Lang
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Peter Radermacher
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Francesco Roselli
- 1 Department of Neurology, Ulm University , Ulm, Germany .,4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| |
Collapse
|
22
|
Zhang YB, Wang YP, Liu J. [Research advances in the role of aptamers in the diagnosis and targeted therapy of pediatric cancer]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:421-427. [PMID: 29764582 PMCID: PMC7389069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 03/29/2018] [Indexed: 11/12/2023]
Abstract
Aptamers are single-stranded DNA or RNA which are isolated from synthesized random oligonucleotide library in vitro via systematic evolution of ligands by exponential enrichment (SELEX) and can bind to metal ions, small molecules, carbohydrates, lipids, proteins, and others targets with high affinity and specificity. Aptamers have the advantages of simple preparation, good thermal stability, and low immunogenicity and have great potential in the medical fields such as molecular imaging, biosensing, early diagnosis of diseases, and targeted therapy. Aptamer technology may be useful for early diagnosis and targeted therapy of pediatric cancer, and may avoid the side effects of conventional chemotherapy, such as growth and development disorders and long-term organ dysfunction. This article reviews the latest research advances in the selection and application of aptamers for pediatric cancer.
Collapse
Affiliation(s)
- Yi-Bin Zhang
- Molecular Research Center, School of Life Sciences, Central South University, Changsha 410078, China.
| | | | | |
Collapse
|
23
|
Zhang YB, Wang YP, Liu J. [Research advances in the role of aptamers in the diagnosis and targeted therapy of pediatric cancer]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:421-427. [PMID: 29764582 PMCID: PMC7389069 DOI: 10.7499/j.issn.1008-8830.2018.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 03/29/2018] [Indexed: 06/08/2023]
Abstract
Aptamers are single-stranded DNA or RNA which are isolated from synthesized random oligonucleotide library in vitro via systematic evolution of ligands by exponential enrichment (SELEX) and can bind to metal ions, small molecules, carbohydrates, lipids, proteins, and others targets with high affinity and specificity. Aptamers have the advantages of simple preparation, good thermal stability, and low immunogenicity and have great potential in the medical fields such as molecular imaging, biosensing, early diagnosis of diseases, and targeted therapy. Aptamer technology may be useful for early diagnosis and targeted therapy of pediatric cancer, and may avoid the side effects of conventional chemotherapy, such as growth and development disorders and long-term organ dysfunction. This article reviews the latest research advances in the selection and application of aptamers for pediatric cancer.
Collapse
Affiliation(s)
- Yi-Bin Zhang
- Molecular Research Center, School of Life Sciences, Central South University, Changsha 410078, China.
| | | | | |
Collapse
|
24
|
Wang D, Li JR, Zhang YH, Chen L, Huang T, Cai YD. Identification of Differentially Expressed Genes between Original Breast Cancer and Xenograft Using Machine Learning Algorithms. Genes (Basel) 2018. [PMID: 29534550 PMCID: PMC5867876 DOI: 10.3390/genes9030155] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is one of the most common malignancies in women. Patient-derived tumor xenograft (PDX) model is a cutting-edge approach for drug research on breast cancer. However, PDX still exhibits differences from original human tumors, thereby challenging the molecular understanding of tumorigenesis. In particular, gene expression changes after tissues are transplanted from human to mouse model. In this study, we propose a novel computational method by incorporating several machine learning algorithms, including Monte Carlo feature selection (MCFS), random forest (RF), and rough set-based rule learning, to identify genes with significant expression differences between PDX and original human tumors. First, 831 breast tumors, including 657 PDX and 174 human tumors, were collected. Based on MCFS and RF, 32 genes were then identified to be informative for the prediction of PDX and human tumors and can be used to construct a prediction model. The prediction model exhibits a Matthews coefficient correlation value of 0.777. Seven interpretable interactions within the informative gene were detected based on the rough set-based rule learning. Furthermore, the seven interpretable interactions can be well supported by previous experimental studies. Our study not only presents a method for identifying informative genes with differential expression but also provides insights into the mechanism through which gene expression changes after being transplanted from human tumor into mouse model. This work would be helpful for research and drug development for breast cancer.
Collapse
Affiliation(s)
- Deling Wang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Jia-Rui Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China.
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
25
|
Fu L, Fu H, Qiao J, Pang Y, Xu K, Zhou L, Wu Q, Li Z, Ke X, Xu K, Shi J. High expression of CPNE3 predicts adverse prognosis in acute myeloid leukemia. Cancer Sci 2017; 108:1850-1857. [PMID: 28670859 PMCID: PMC5581509 DOI: 10.1111/cas.13311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 01/17/2023] Open
Abstract
CPNE3, a member of a Ca2+‐dependent phospholipid‐binding protein family, was identified as a ligand of ERBB2 and has a more general role in carcinogenesis. Here, we identified the prognostic significance of CPNE3 expression in acute myeloid leukemia (AML) patients based on two datasets. In the first microarray dataset (n = 272), compared to low CPNE3 expression (CPNE3low), high CPNE3 expression (CPNE3high) was associated with adverse overall survival (OS, P < 0.001) and event‐free survival (EFS, P < 0.001). In the second independent group of AML patients (TCGA dataset, n = 179), CPNE3high was also associated with adverse OS and EFS (OS, P = 0.01; EFS, P = 0.036). Notably, among CPNE3high patients, those received allogenic hematopoietic cell transplantation (HCT) had longer OS and EFS than those with chemotherapy alone (allogeneic HCT, n = 40 vs chemotherapy, n = 46), but treatment modules played an insignificant role in the survival of CPNE3low patients (allogeneic HCT, n = 32 vs chemotherapy, n = 54). These results indicated that CPNE3high is an independent, adverse prognostic factor in AML and might guide treatment decisions towards allogeneic HCT. To understand its inherent mechanisms, we investigated genome‐wide gene/microRNA expression signatures and cell signaling pathways associated with CPNE3 expression. In conclusion, CPNE3high is an adverse prognostic biomarker for AML. Its effect may be attributed to the distinctive genome‐wide gene/microRNA expression and related cell signaling pathways.
Collapse
Affiliation(s)
- Lin Fu
- Department of Hematology and Lymphoma Research Center, Third Hospital, Peking University, Beijing, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Huaping Fu
- Departments of Nuclear Medicine, Chinese PLA General Hospital, Beijing, China
| | - Jianlin Qiao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yifan Pang
- Department of Medicine, William Beaumont Hospital, Royal Oak, MI, USA
| | - Keman Xu
- Northeastern University, Boston, MA, USA
| | - Lei Zhou
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Qingyun Wu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaoyan Ke
- Department of Hematology and Lymphoma Research Center, Third Hospital, Peking University, Beijing, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jinlong Shi
- Departments of Biomedical Engineering, Chinese PLA General Hospital, Beijing, China.,Departments of Medical Big Data, Chinese PLA General Hospital, Beijing, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
26
|
Abstract
Aptamers are nucleic acids referred to as chemical antibodies as they bind to their specific targets with high affinity and selectivity. They are selected via an iterative process known as ‘selective evolution of ligands by exponential enrichment’ (SELEX). Aptamers have been developed against numerous cancer targets and among them, many tumor cell-membrane protein biomarkers. The identification of aptamers targeting cell-surface proteins has mainly been performed by two different strategies: protein- and cell-based SELEX, when the targets used for selection were proteins and cells, respectively. This review aims to update the literature on aptamers targeting tumor cell surface protein biomarkers, highlighting potentials, pitfalls of protein- and cell-based selection processes and applications of such selected molecules. Aptamers as promising agents for diagnosis and therapeutic approaches in oncology are documented, as well as aptamers in clinical development.
Collapse
|