1
|
Abdel-Aziz AAM, El-Azab AS, Brogi S, Ayyad RR, Alkahtani HM, Abuelizz HA, Al-Suwaidan IA, Al-Obaid AM. Synthesis, enzyme inhibition assay, and molecular modeling study of novel pyrazolines linked to 4-methylsulfonylphenyl scaffold: antitumor activity and cell cycle analysis. RSC Adv 2024; 14:22132-22146. [PMID: 39005246 PMCID: PMC11240878 DOI: 10.1039/d4ra03902e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Antitumor activity using 59 cancer cell lines and enzyme inhibitory activity of a newly synthesized pyrazoline-linked 4-methylsulfonylphenyl scaffold (compounds 18a-q) were measured and compared with those of standard drugs. Pyrazolines 18b, 18c, 18f, 18g, 18h, and 18n possessed significant antitumor activity, with a positive cytotoxic effect (PCE) of 22/59, 21/59, 21/59, 48/59, 51/59, and 20/59, respectively. The cancer cell lines HL60, MCF-7, and MDA-MB-231 were used to measure the IC50 values of derivatives 18c, 18g, and 18hvia the MTT assay method, and the results were compared with those of reference drugs. Derivatives 18g and 18h showed potent and broad-spectrum antitumor activities against HL60 (IC50 of 10.43, 8.99 μM, respectively), MCF-7 (IC50 of 11.7 and 12.4 μM, respectively), and MDA-MB-231 (IC50 of 4.07 and 7.18 μM, respectively). Compound 18c exhibited strong antitumor activity against HL60 and MDA-MB-231 cell lines with IC50 values of 8.43 and 12.54 μM, respectively, and moderate antitumor activity against MCF-7 cell lines with an IC50 value of 16.20 μM. Compounds 18c, 18g, and 18h remarkably inhibited VEGFR2 kinase (IC50 = 0.218, 0.168, and 0.135 μM, respectively) compared with the reference drug sorafenib (IC50 = 0.041 μM). Compounds 18g and 18h effectively inhibited HER2 kinase (IC50 = 0.496 and 0.253 μM, respectively) compared with erlotinib (IC50 = 0.085 μM). Compound 18h inhibited EGFR kinase (IC50 = 0.574 μM) with a potency comparable with that of the reference drug erlotinib (IC50 = 0.105 μM). Pyrazolines 18c, 18f, and 18h arrested the S/G2 phase of the cell cycle in HL-60 cells. In addition, derivatives 18c, 18f, and 18h revealed lower Bcl-2 protein expression anti-apoptotic levels and higher Bax, caspase-3, and caspase-9 expression levels. Molecular docking studies of derivative 18h into the binding sites of EGFR, HER2, and VEGFR2 kinases explored the interaction mode of these pyrazoline derivatives and their structural requirements for antitumor activity.
Collapse
Affiliation(s)
- Alaa A-M Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Adel S El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Simone Brogi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Rezk R Ayyad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-AzharUniversity Cairo Egypt
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Hatem A Abuelizz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Ibrahim A Al-Suwaidan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Abdulrahman M Al-Obaid
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| |
Collapse
|
2
|
Zara Rozalen A, Martin JM, Rajendran R, Jain M, Nava VE. Ductal Adenocarcinoma of the Prostate with Novel Genetic Alterations Characterized by Next-Generation Sequencing. Curr Oncol 2024; 31:1556-1561. [PMID: 38534951 PMCID: PMC10968787 DOI: 10.3390/curroncol31030118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/23/2023] [Accepted: 03/17/2024] [Indexed: 05/26/2024] Open
Abstract
Ductal adenocarcinoma of the prostate (DAP) is an uncommon variant of prostate cancer associated with aggressive disease and poor outcome. It presents most frequently as a mixed tumor combined with acinar adenocarcinoma. Although the histopathological features of DAP are well known, its genomic characteristics are still evolving, prompting the suggestion that all DAP would benefit from molecular analysis with the purpose of improving tumor recognition, genetic classification, and, ultimately, personalized therapy. Herein, we report a case of DAP with novel genetic alterations (BCOR P1153S, ERG M219I, KDR A750E, POLE S1896P, and RAD21 T461del).
Collapse
Affiliation(s)
- Alexandra Zara Rozalen
- Department of Pathology, Veterans Affairs Medical Center, Washington, DC 20422, USA; (A.Z.R.); (J.M.M.); (R.R.)
- Department of Pathology, Mount Sinai Morningside and West Hospitals, New York, NY 10019, USA
| | - Jose Manuel Martin
- Department of Pathology, Veterans Affairs Medical Center, Washington, DC 20422, USA; (A.Z.R.); (J.M.M.); (R.R.)
| | - Rithika Rajendran
- Department of Pathology, Veterans Affairs Medical Center, Washington, DC 20422, USA; (A.Z.R.); (J.M.M.); (R.R.)
| | - Maneesh Jain
- Section of Hematology & Oncology, Veterans Affairs Medical Center, Washington, DC 20422, USA;
- Department of Medicine, The George Washington University Hospital, Washington, DC 20037, USA
| | - Victor E. Nava
- Department of Pathology, Veterans Affairs Medical Center, Washington, DC 20422, USA; (A.Z.R.); (J.M.M.); (R.R.)
- Department of Pathology, The George Washington University Hospital, Washington, DC 20037, USA
| |
Collapse
|
3
|
Beig Parikhani A, Dehghan R, Talebkhan Y, Bayat E, Biglari A, Shokrgozar MA, Ahangari Cohan R, Mirabzadeh E, Ajdary S, Behdani M. A novel nanobody-based immunocytokine of a mutant interleukin-2 as a potential cancer therapeutic. AMB Express 2024; 14:19. [PMID: 38337114 PMCID: PMC10857990 DOI: 10.1186/s13568-023-01648-2] [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: 11/16/2023] [Accepted: 11/26/2023] [Indexed: 02/12/2024] Open
Abstract
The immunotherapeutic application of interleukin-2 (IL-2) in cancer treatment is limited by its off-target effects on different cell populations and insufficient activation of anti-tumor effector cells at the site of the tumor upon tolerated doses. Targeting IL-2 to the tumor microenvironment by generating antibody-cytokine fusion proteins (immunocytokine) would be a promising approach to increase efficacy without associated toxicity. In this study, a novel nanobody-based immunocytokine is developed by the fusion of a mutant (m) IL-2 with a decreased affinity toward CD25 to an anti-vascular endothelial growth factor receptor-2 (VEGFR2) specific nanobody, denoted as VGRmIL2-IC. The antigen binding, cell proliferation, IFN-γ-secretion, and cytotoxicity of this new immunocytokine are evaluated and compared to mIL-2 alone. Furthermore, the pharmacokinetic properties are analyzed. Flow cytometry analysis shows that the VGRmIL2-IC molecule can selectively target VEGFR2-positive cells. The results reveal that the immunocytokine is comparable to mIL-2 alone in the stimulation of Primary Peripheral Blood Mononuclear Cells (PBMCs) and cytotoxicity in in vitro conditions. In vivo studies demonstrate improved pharmacokinetic properties of VGRmIL2-IC in comparison to the wild or mutant IL-2 proteins. The results presented here suggest VGRmIL2-IC could be considered a candidate for the treatment of VEGFR2-positive tumors.
Collapse
Affiliation(s)
- Arezoo Beig Parikhani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Rada Dehghan
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Yeganeh Talebkhan
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Bayat
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Biglari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Esmat Mirabzadeh
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Rodríguez-Nava C, Ortuño-Pineda C, Illades-Aguiar B, Flores-Alfaro E, Leyva-Vázquez MA, Parra-Rojas I, Del Moral-Hernández O, Vences-Velázquez A, Cortés-Sarabia K, Alarcón-Romero LDC. Mechanisms of Action and Limitations of Monoclonal Antibodies and Single Chain Fragment Variable (scFv) in the Treatment of Cancer. Biomedicines 2023; 11:1610. [PMID: 37371712 DOI: 10.3390/biomedicines11061610] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Monoclonal antibodies are among the most effective tools for detecting tumor-associated antigens. The U.S. Food and Drug Administration (FDA) has approved more than 36 therapeutic antibodies for developing novel alternative therapies that have significant success rates in fighting cancer. However, some functional limitations have been described, such as their access to solid tumors and low interaction with the immune system. Single-chain variable fragments (scFv) are versatile and easy to produce, and being an attractive tool for use in immunotherapy models. The small size of scFv can be advantageous for treatment due to its short half-life and other characteristics related to the structural and functional aspects of the antibodies. Therefore, the main objective of this review was to describe the current situation regarding the mechanisms of action, applications, and limitations of monoclonal antibodies and scFv in the treatment of cancer.
Collapse
Affiliation(s)
- Cynthia Rodríguez-Nava
- Laboratorio de Investigación en Citopatología e Histoquímica, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
- Laboratorio de Investigación en Inmunobiología y Diagnóstico Molecular, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Carlos Ortuño-Pineda
- Laboratorio de Proteínas y Ácidos Nucleicos, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Investigación en Biomedicina Molecular, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Eugenia Flores-Alfaro
- Laboratorio de Investigación en Epidemiología Clínica y Molecular, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Investigación en Biomedicina Molecular, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | | | - Amalia Vences-Velázquez
- Laboratorio de Investigación en Inmunobiología y Diagnóstico Molecular, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Karen Cortés-Sarabia
- Laboratorio de Investigación en Inmunobiología y Diagnóstico Molecular, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| | - Luz Del Carmen Alarcón-Romero
- Laboratorio de Investigación en Citopatología e Histoquímica, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Mexico
| |
Collapse
|
5
|
Lu RM, Liang KH, Chiang HL, Hsu FF, Lin HT, Chen WY, Ke FY, Kumari M, Chou YC, Tao MH, Yi-Ling Lin, Wu HC. Broadly neutralizing antibodies against Omicron variants of SARS-CoV-2 derived from mRNA-lipid nanoparticle-immunized mice. Heliyon 2023; 9:e15587. [PMID: 37090428 PMCID: PMC10111857 DOI: 10.1016/j.heliyon.2023.e15587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 08/02/2023] Open
Abstract
The COVID-19 pandemic continues to threaten human health worldwide as new variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerge. Currently, the predominant circulating strains around the world are Omicron variants, which can evade many therapeutic antibodies. Thus, the development of new broadly neutralizing antibodies remains an urgent need. In this work, we address this need by using the mRNA-lipid nanoparticle immunization method to generate a set of Omicron-targeting monoclonal antibodies. Five of our novel K-RBD-mAbs show strong binding and neutralizing activities toward all SARS-CoV-2 variants of concern (Alpha, Beta, Gamma, Delta and Omicron). Notably, the epitopes of these five K-RBD-mAbs are overlapping and localized around Y453 and F486 of the spike protein receptor binding domain (RBD). Chimeric derivatives of the five antibodies (K-RBD-chAbs) neutralize Omicron sublineages BA.1 and BA.2 with low IC50 values ranging from 5.7 to 12.9 ng/mL. Additionally, we performed antibody humanization on broadly neutralizing chimeric antibodies to create K-RBD-hAb-60 and -62, which still retain excellent neutralizing activity against Omicron. Our results collectively suggest that these five therapeutic antibodies may effectively combat current and emerging SARS-CoV-2 variants, including Omicron BA.1 and BA.2. Therefore, the antibodies can potentially be used as universal neutralizing antibodies against SARS-CoV-2.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Kang-Hao Liang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Hsiao-Ling Chiang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Fu-Fei Hsu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Hsiu-Ting Lin
- Institute of Cellular and Organismic Biology (ICOB), Academia Sinica, Taipei, Taiwan
| | - Wan-Yu Chen
- Institute of Cellular and Organismic Biology (ICOB), Academia Sinica, Taipei, Taiwan
| | - Feng-Yi Ke
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Monika Kumari
- Institute of Cellular and Organismic Biology (ICOB), Academia Sinica, Taipei, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Mi-Hua Tao
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Sciences (IBMS), Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Sciences (IBMS), Academia Sinica, Taipei, Taiwan
| | - Han-Chung Wu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
- Institute of Cellular and Organismic Biology (ICOB), Academia Sinica, Taipei, Taiwan
| |
Collapse
|
6
|
Hussain W, Yang X, Ullah M, Wang H, Aziz A, Xu F, Asif M, Ullah MW, Wang S. Genetic engineering of bacteriophages: Key concepts, strategies, and applications. Biotechnol Adv 2023; 64:108116. [PMID: 36773707 DOI: 10.1016/j.biotechadv.2023.108116] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023]
Abstract
Bacteriophages are the most abundant biological entity in the world and hold a tremendous amount of unexplored genetic information. Since their discovery, phages have drawn a great deal of attention from researchers despite their small size. The development of advanced strategies to modify their genomes and produce engineered phages with desired traits has opened new avenues for their applications. This review presents advanced strategies for developing engineered phages and their potential antibacterial applications in phage therapy, disruption of biofilm, delivery of antimicrobials, use of endolysin as an antibacterial agent, and altering the phage host range. Similarly, engineered phages find applications in eukaryotes as a shuttle for delivering genes and drugs to the targeted cells, and are used in the development of vaccines and facilitating tissue engineering. The use of phage display-based specific peptides for vaccine development, diagnostic tools, and targeted drug delivery is also discussed in this review. The engineered phage-mediated industrial food processing and biocontrol, advanced wastewater treatment, phage-based nano-medicines, and their use as a bio-recognition element for the detection of bacterial pathogens are also part of this review. The genetic engineering approaches hold great potential to accelerate translational phages and research. Overall, this review provides a deep understanding of the ingenious knowledge of phage engineering to move them beyond their innate ability for potential applications.
Collapse
Affiliation(s)
- Wajid Hussain
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaohan Yang
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mati Ullah
- Department of Biotechnology, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Huan Wang
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ayesha Aziz
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fang Xu
- Huazhong University of Science and Technology Hospital, Wuhan 430074, China
| | - Muhammad Asif
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Muhammad Wajid Ullah
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Shenqi Wang
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
7
|
Cao Y, Sun C, Huo G, Wang H, Wu Y, Wang F, Liu S, Zhai S, Zhang X, Zhao H, Hu M, Gu W, Yang Y, Wang S, Liang C, Lyu J, Lu T, Wang Y, Xie L, Fan C. Novel hKDR mouse model depicts the antiangiogenesis and apoptosis-promoting effects of neutralizing antibodies targeting vascular endothelial growth factor receptor 2. Cancer Sci 2022; 114:115-128. [PMID: 36114822 PMCID: PMC9807522 DOI: 10.1111/cas.15594] [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: 05/02/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 01/07/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2)/KDR plays a critical role in tumor growth, diffusion, and invasion. The amino acid sequence homology of KDR between mouse and human in the VEGF ligand-binding domain was low, thus the WT mice could not be used to evaluate Abs against human KDR, and the lack of a suitable mouse model hindered both basic research and drug developments. Using the CRISPR/Cas9 technique, we successfully inserted different fragments of the human KDR coding sequence into the chromosomal mouse Kdr exon 4 locus to obtain an hKDR humanized mouse that can be used to evaluate the marketed Ab ramucirumab. In addition, the humanized mAb VEGFR-HK19 was developed, and a series of comparative assays with ramucirumab as the benchmark revealed that VEGFR-HK19 has higher affinity and superior antiproliferation activity. Moreover, VEGFR-HK19 selectively inhibited tumor growth in the hKDR mouse model but not in WT mice. The most important binding epitopes of VEGFR2-HK19 are D257, L313, and T315, located in the VEGF binding region. Therefore, the VEGFR2-HK19 Ab inhibits tumor growth by blocking VEGF-induced angiogenesis, inflammation, and promoting apoptosis. To our best knowledge, this novel humanized KDR mouse fills the gaps both in an animal model and the suitable in vivo evaluation method for developing antiangiogenesis therapies in the future, and the newly established humanized Ab is expected to be a drug candidate possibly benefitting tumor patients.
Collapse
Affiliation(s)
- Yuan Cao
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Chunyun Sun
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Guitao Huo
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Huiyu Wang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Yong Wu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Fei Wang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Susu Liu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Shijie Zhai
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Xiao Zhang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Haoyang Zhao
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Meiling Hu
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Wenda Gu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Yanwei Yang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Sanlong Wang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Chunnan Liang
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Jianjun Lyu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Tiangong Lu
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Institute for Biological Product ControlNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina,Beijing Key Laboratory of Monoclonal Antibody Research and DevelopmentSino Biological Inc.BeijingChina,Cell Culture Engineering CenterChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changfa Fan
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| |
Collapse
|
8
|
Design and synthesis of some new 6-bromo-2-(pyridin-3-yl)-4-substituted quinazolines as multi tyrosine kinase inhibitors. Bioorg Chem 2022; 128:106099. [PMID: 35994884 DOI: 10.1016/j.bioorg.2022.106099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Abstract
The present study involves design and synthesis of five series of 6-bromo-2-(pyridin-3-yl)-4-substituted quinazolines 9a-l, 11a-e, 13a-c, 14a-f and 15a-e. Candidates 9a-l and 11a-e were evaluated for their EGFR and HER2 inhibitory activity compared to Lapatinib. Compounds 9b, 9d, 9f, 11b and 11c were further screened for their in vitro cytotoxicity against two human breast cancer cell lines: AU-565 and MDA-MB-231 in addition to normal breast cell line MCF10A. Compound 9d revealed a remarkable cytotoxic efficacy against AU-565 cell line (IC50 = 1.54 µM) relative to Lapatinib (IC50 = 0.48 µM), whereas compounds 9d and 11c showed a superior cytotoxicity towards MDA-MB-231 (IC50 = 2.67 and 1.75 µM, respectively) in comparison to Lapatinib (IC50 = 9.29 µM). Moreover, compounds 13a-c, 13a-c, 14a-f and 15a-e were tested for their VEGFR-2 inhibitory activity compared to Sorafenib. Compounds 13a, 14c and 14e exhibited remarkable inhibition (IC50 = 79.80, 50.22 and 78.02 nM, respectively) relative to Sorafenib (IC50 = 51.87 nM). In vitro cytotoxicity of these compounds against HepG2, HCT-116 and normal cell (WISH) revealed a superior cytotoxicity against HepG2, HCT-116 especially 13a (IC50 = 17.51 and 5.56 µM, respectively) and 14c (IC50 = 10.40 and 3.37 µM, respectively) compared to Sorafenib (IC50 = 19.33 and 6.82 µM, respectively). Compounds 9d, 11c and 14c were subjected to cell cycle analysis and apoptotic assay. Molecular docking and ADME prediction studies were fulfilled to illustrate the interaction of the potent derivatives with the hot spots of the active site of EGFR, HER2 and VEGFR-2 along with prediction of their pharmacokinetic and physicochemical properties.
Collapse
|
9
|
Abuzenadah AM, Al-Sayes F, Mahafujul Alam SS, Hoque M, Karim S, Hussain IMR, Tabrez S. Elucidating Antiangiogenic Potential of Rauwolfia serpentina: VEGFR-2 Targeting-Based Molecular Docking Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6224666. [PMID: 35198035 PMCID: PMC8860507 DOI: 10.1155/2022/6224666] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022]
Abstract
Angiogenesis plays a critical role in tumorigenesis as it provides the necessary blood supply to the newly grown solid tumor. It helps maintain the tumor microenvironment, promotes tumor development, progression, and metastasis. The vascular epithelial growth factor (VEGF), interacting with the tyrosine kinase receptor VEGFR-2 on endothelial cells, exerts its proangiogenic activity. Hence, targeting the VEGFR-2 signaling is considered a promising strategy to inhibit angiogenesis and thus cancer treatment. This study aims to identify the bioactive compounds derived from the medicinal herb Rauwolfia serpentina that effectively binds with VEGFR-2. The bioactive compounds of R. serpentina were first screened for their physicochemical properties using the DataWarrior program (version 5.5.0). Finally, 17 compounds that obeyed Lipinski's rule of five and showed good drug-likeness were selected for molecular docking studies. Molecular docking results showed that the ligands ajmalicidine, 1, 2-dihydrovomilenine, rauwolscine, yohimbine, ajmaline, and papaverine interact strongly with the target VEGFR-2 receptor. Hydrogen bonds and hydrophobic interactions stabilized the interactions of these compounds with VEGFR-2. These compounds showed favourable drug-like properties and possess no significant toxicity. Therefore, the findings of this study indicate that the compounds derived from R. serpentina can be considered for the development of antiangiogenic drug candidates by targeting VEGFR-2.
Collapse
Affiliation(s)
- Adel M. Abuzenadah
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatin Al-Sayes
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mehboob Hoque
- Applied Bio-Chemistry Lab, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibtessam M. R. Hussain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Hwang YC, Lu RM, Su SC, Chiang PY, Ko SH, Ke FY, Liang KH, Hsieh TY, Wu HC. Monoclonal antibodies for COVID-19 therapy and SARS-CoV-2 detection. J Biomed Sci 2022; 29:1. [PMID: 34983527 PMCID: PMC8724751 DOI: 10.1186/s12929-021-00784-w] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is an exceptional public health crisis that demands the timely creation of new therapeutics and viral detection. Owing to their high specificity and reliability, monoclonal antibodies (mAbs) have emerged as powerful tools to treat and detect numerous diseases. Hence, many researchers have begun to urgently develop Ab-based kits for the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ab drugs for use as COVID-19 therapeutic agents. The detailed structure of the SARS-CoV-2 spike protein is known, and since this protein is key for viral infection, its receptor-binding domain (RBD) has become a major target for therapeutic Ab development. Because SARS-CoV-2 is an RNA virus with a high mutation rate, especially under the selective pressure of aggressively deployed prophylactic vaccines and neutralizing Abs, the use of Ab cocktails is expected to be an important strategy for effective COVID-19 treatment. Moreover, SARS-CoV-2 infection may stimulate an overactive immune response, resulting in a cytokine storm that drives severe disease progression. Abs to combat cytokine storms have also been under intense development as treatments for COVID-19. In addition to their use as drugs, Abs are currently being utilized in SARS-CoV-2 detection tests, including antigen and immunoglobulin tests. Such Ab-based detection tests are crucial surveillance tools that can be used to prevent the spread of COVID-19. Herein, we highlight some key points regarding mAb-based detection tests and treatments for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yu-Chyi Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Chieh Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Pao-Yin Chiang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Feng-Yi Ke
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Kang-Hao Liang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan
| | - Tzung-Yang Hsieh
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
11
|
Kiamehr P, Shahidi M, Samii A, Zaker F. Dual Effects of Resveratrol on the Expression and Secretion of Angiogenic Factors. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2022; 11:16-30. [PMID: 36397806 PMCID: PMC9653554 DOI: 10.22088/ijmcm.bums.11.1.16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/01/2022] [Accepted: 07/31/2022] [Indexed: 11/24/2022]
Abstract
Angiogenesis is an essential process in the growth, development, and transition of tumors from dormancy to proliferating state. Resveratrol (RSV), as a natural polyphenolic compound, is claimed to be effective in regulating angiogenesis. This study aimed to evaluate the impact of RSV onthe angiogenesis process in HUVECs (human umbilical vein endothelial cells) alone and co-cultured with Jurkat cells. The effects of RSV on HUVECs and Jurkat cell viability and apoptosis were measured by MTT and Annexin-V/PI methods. HUVECs were co-cultured with pre-treated Jurkat cells and incubated for 24 h, 48 h and 72 h. The angiogenesis process in HUVECs and Jurkat cells alone and in co-culture models was investigated by analyzing the expression of VEGF, VEGFR-2, and Interleukin-8 (IL-8) employing qPCR and ELISA. RSV at low concentration (40 µM) had no significant effects on apoptosis rate of HUVECs, but higher concentrations (80-160 µM) increased apoptosis in co-culture method and HUVECs alone. RSV significantly reduced VEGFR2 and IL-8 gene expression also, IL-8 protein concentration in HUVECs, but the effects of this drug in the HUVECs-Jurkats co-culture were different. Expression of VEGF in Jurkat cells increased following treatment with RSV. RSV had direct anti-angiogenic effects on HUVECs. Unexpectedly its indirect effects were not significant on HUVECs-Jurkats co-culture. Results of our study showed, RSV may be effective in anti-angiogenesis therapy, but in some situations, it may induce angiogenesis. So, appropriate concentrations should achieve to minimize the unpredicted effects of RSV.
Collapse
Affiliation(s)
| | - Minoo Shahidi
- Corresponding Author: Minoo Shahidi Address: Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran. E-mail:
| | | | | |
Collapse
|
12
|
Monoclonal Antibodies against Nucleocapsid Protein of SARS-CoV-2 Variants for Detection of COVID-19. Int J Mol Sci 2021; 22:ijms222212412. [PMID: 34830291 PMCID: PMC8623253 DOI: 10.3390/ijms222212412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/13/2021] [Accepted: 11/14/2021] [Indexed: 02/04/2023] Open
Abstract
Mitigation strategies of the coronavirus disease 2019 (COVID-19) pandemic have been greatly hindered by the continuous emergence of SARS-CoV-2 variants. New sensitive, rapid diagnostic tests for the wide-spectrum detection of viral variants are needed. We generated a panel of 41 monoclonal antibodies against the SARS-CoV-2 nucleocapsid protein (NP) by using mice hybridoma techniques. Of these mAbs, nine exhibited high binding activities and were applied in latex-based lateral flow immunoassays (LFIAs). The LFIAs utilizing NP-mAb-7 and -40 had the best sensitivity and lowest limit of detection: 8 pg for purified NP and 625 TCID50/mL for the authentic virus (hCoV-19/Taiwan/4/2020). The specificity tests showed that the NP-mAb-40/7 LFIA strips did not cross-react with five human coronavirus strains or 20 other common respiratory pathogens. Importantly, we found that 10 NP mutants, including alpha (B.1.1.7), beta (B.1.351), gamma (P.1), and delta (B.1.617.2) variants, could be detected by NP-mAb-40/7 LFIA strips. A clinical study (n = 60) of the NP-mAb-40/7 LFIA strips demonstrated a specificity of 100% and sensitivity of 90% in infected individuals with cycle threshold (Ct) values < 29.5. These anti-NP mAbs have strong potential for use in the clinical detection of SARS-CoV-2 infection, whether the virus is wild-type or a variant of concern.
Collapse
|
13
|
Leis-Filho AF, Lainetti PD, Kobayashi PE, Palmieri C, Amorim RL, Fonseca-Alves CE. Expression and prognostic significance of vascular endothelial growth factor-A (VEGF-A) and its receptor in canine prostate cancer. Prostate 2021; 81:1021-1031. [PMID: 34320239 DOI: 10.1002/pros.24199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/11/2021] [Accepted: 07/07/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Vascular endothelial growth factor-A (VEGF-A) and its receptor, VEGF receptor-2 (VEGFR-2), represent a complex family of angiogenic molecules consisting of different ligands and receptors. Due to the importance of VEGF-A/VEGFR-2 signaling in tumor proliferation and angiogenesis, this study aimed to evaluate the protein and gene expression levels of VEGF-A and VEGFR-2 in canine prostate cancer (PC). METHODS We analyzed VEGF-A and VEGFR-2 expression in 87 PC samples by immunohistochemistry and quantitative-polymerase chain reaction. PC samples were graded according to the Gleason score and the immunohistochemical staining for VEGF-A and VEGFR-2 was quantified using a selected threshold from the ImageJ Software. Microvascular density was assessed by cluster of differentiation 31 staining and counting the number of positive vessels. Additionally, the homology of VEGF-A and VEGFR-2 between humans and dogs was assessed, followed by the construction of a protein structure homology model to compare the tertiary structures of these proteins in both species. RESULTS Negative to weakly positive expression levels of VEGF-A and VEGFR-2 were observed in the epithelial cells of the normal prostate (NP) and prostatic hyperplasia samples. In contrast, the canine proliferative atrophy and PC samples exhibited higher VEGF-A (p < .0001) and VEGFR-2 (p < .0001) compared to NP. Moreover, positive correlations between the expression levels of VEGF-A and VEGFR-2 (Spearman's coefficient (r) = .68, p = .013) and the expression levels of VEGF-A and VEGFR-2 proteins (r = .8, p < .0001) were also observed in the NP samples. Additionally, the patients with PC exhibiting higher VEGFR-2 expression levels experienced a shorter survival period (p = .0372). Furthermore, we found an association between the microvascular density and overall survival. Dogs with a higher number of vessels showed a shorter survival time. We further demonstrated that the VEGF-A and VEGFR-2 exhibited high homology between humans and dogs, and identified their protein structures in both species. CONCLUSIONS In conclusion, VEGFR-2 appears to be an independent prognostic factor in animals with PC. VEGF-A and VEGFR-2 are highly conserved between humans and dogs, which can be investigated further in future cross-species studies to explore their therapeutic applications.
Collapse
Affiliation(s)
- Antonio Fernando Leis-Filho
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil
| | - Patricia deFaria Lainetti
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil
| | - Priscila Emiko Kobayashi
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil
| | - Chiara Palmieri
- School of Veterinary Science, The University of Queensland Gatton Campus, Gatton, Queensland, Australia
| | - Renée Laufer Amorim
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil
- Institute of Health Sciences, Paulista University-UNIP, Bauru, Brazil
| |
Collapse
|
14
|
Isatin-Hydrazones with Multiple Receptor Tyrosine Kinases (RTKs) Inhibitory Activity and In-Silico Binding Mechanism. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11093746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recently, we have reported a series of isatin hydrazone, two of them, namely, 3-((2,6-dichlorobenzylidene)hydrazono)indolin-2-one (1) and 3-((2-chloro-6-fluorobenzylidene)hydrazono)indolin-2-one (2) having potent cytotoxicity, showing cyclin-dependent kinases (CDK2) inhibitory activity and bearing recommended drug likeness properties. Since both compounds (1 and 2) showed inhibitory activity against CDK2, we assumed it would also have multiple receptor tyrosine kinases (RTKs) inhibitory activity. Considering those points, here, above-mentioned two isatin hydrazone 1 and 2 were synthesized using previously reported method for further investigation of their potency on RTKs (EGFR, VEGFR-2 and FLT-3) inhibitory activity. As expected, Compound 1 exhibited excellent inhibitory activity against epidermal growth factor receptor (EGFR, IC50 = 0.269 µM), vascular epidermal growth factor receptor 2 (VEGFR-2, IC50 = 0.232 µM) and FMS-like tyrosine kinase-3 (FLT-3, IC50 = 1.535 µM) tyrosine kinases. On the other hand, Compound 2 also exhibited excellent inhibitory activity against EGFR (IC50 = 0.369 µM), VEGFR-2 (IC50 = 0.266 µM) and FLT-3 (IC50 = 0.546 µM) tyrosine kinases. A molecular docking study with EGFR, VEGFR-2 and FLT-3 kinase suggested that both compounds act as type I ATP competitive inhibitors against EGFR and VEGFR-2, and type II ATP non-competitive inhibitors against FLT-3.
Collapse
|
15
|
Heinen H, Seyler L, Popp V, Hellwig K, Bozec A, Uder M, Ellmann S, Bäuerle T. Morphological, functional, and molecular assessment of breast cancer bone metastases by experimental ultrasound techniques compared with magnetic resonance imaging and histological analysis. Bone 2021; 144:115821. [PMID: 33348127 DOI: 10.1016/j.bone.2020.115821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND The imaging of bone metastases, which is regularly performed by cross-sectional modalities, is clinically vital when characterizing and staging osseous lesions. In this paper, we aimed to establish a novel methodology using experimental ultrasound (US) techniques to assess the morphological, functional, and molecular features of breast cancer bone metastases in an animal model, compared with magnetic resonance imaging (MRI) and histological analysis. MATERIALS AND METHODS Nude rats were implanted intra-arterially with MDA-MB-231 breast cancer cells to induce osteolytic metastasis in their right hind legs. Once tumors had developed, an experimental US technique using automatic 3D scanning and MRI were performed. For assessment of perfusion, functional imaging techniques included contrast-enhanced US (CEUS) and dynamic contrast-enhanced MRI (DCE-MRI). For molecular ultrasound, anti-VEGFR2 conjugated microbubbles were applied and correlated with immunostaining for VEGFR2 expression. RESULTS 3D US enabled the automatic assessment of osteolytic lesions, including the largest tumor diameters along the x-, y- and z-axes as well as the segmented tumor volumes, without significant differences between US and MRI (p > 0.18). The CEUS and DCE-MRI of osseous lesions showed corresponding results for the parameters peak enhancement, wash-in area under the curve (both, r > 0.5) and wash-in perfusion index (r > 0.3) when differentiating between tumor, necrotic tissue and healthy muscle tissue (all, p < 0.01). Finally, molecular US allowed the non-invasive assessment of increased VEGFR2 expression in skeletal lesions compared with surrounding muscle tissue (p = 0.03), while a control antibody could not discriminate between these tissues (p = 0.44)-a factor which was confirmed by histological analysis. CONCLUSION To the best of our knowledge, this is the first report on an imaging protocol for breast cancer bone metastasis using an experimental US scanner. Therefore, we present a novel methodology to characterize these osseous lesions on the morphological, functional, and molecular level in correlation with MRI and histological analysis.
Collapse
Affiliation(s)
- Henrik Heinen
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany; Institute of Radiology, University Hospital, Paracelsus University, Prof.-Ernst-Nathan-Str. 1, 90419 Nuremberg, Germany
| | - Lisa Seyler
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Vanessa Popp
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Konstantin Hellwig
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Aline Bozec
- Medical Clinic 3 - Rheumatology and Immunology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Stephan Ellmann
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany.
| |
Collapse
|
16
|
Song Y, Zhuang G, Li J, Zhang M. BAIAP2L2 facilitates the malignancy of prostate cancer (PCa) via VEGF and apoptosis signaling pathways. Genes Genomics 2021; 43:421-432. [PMID: 33646530 DOI: 10.1007/s13258-021-01061-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/09/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is the second most common type of male cancer in western. Despite key roles of brain-specific angiogenesis inhibitor 1-associated protein like 2 (BAIAP2L2) in several cancers, the function of BAIAP2L2 in PCa is never reported. OBJECTIVE We aimed to investigate the role of BAIAP2L2 in the progression of PCa and decipher the underlying mechanisms. METHODS RNA sequencing data from TCGA database were used to evaluate the expression of BAIAP2L2 in PCa. Survival analysis and Cox regression model analysis were conducted to evaluate the prognostic value of BAIAP2L2. BAIAP2L2-associated pathways were preliminary analyzed by Gene Set Enrichment Analysis (GSEA) method and confirmed by western blot assays. Cell proliferation and transwell assays were performed to determine biological behaviors in BAIAP2L2 knocked-down or overexpressed PCa cell lines including LNCaP and PC-3 cells. RESULTS In our study, BAIAP2L2 was significantly up-regulated in PCa tissues and cell lines and independently associated with the poor prognosis of PCa patients. Knockdown of BAIAP2L2 notably repressed proliferation, migration and invasion of PCa cells. And overexpression of BAIAP2L2 obtained the contrary results. Mechanically, GSEA method and western blot results of key molecules in signaling pathways implicated that the depletion of BAIAP2L2 inactivated the vascular endothelial growth factors (VEGFs) and induced apoptosis signaling pathways in PCa cells. CONCLUSIONS Overall, these findings revealed that BAIAP2L2 may support tumorigenesis and malignant development of prostate cancer cells via VEGF and apoptosis signaling pathways, and it could be considered as a promising biomarker and independent prognostic predictor of prostate cancer.
Collapse
Affiliation(s)
- Yuanzi Song
- Department of Urology, Zibo First Hospital, Emeishan East Road, Zibo, China
| | - Guishan Zhuang
- Department of Urology, Weifang People's Hospital, 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong, China
| | - Jiazhen Li
- Intravenous Medication Center of Binzhou People's Hospital, Binzhou, Shandong, China
| | - Mingqing Zhang
- Department of Urology, Weifang People's Hospital, 151 Guangwen Street, Kuiwen District, Weifang, 261041, Shandong, China.
| |
Collapse
|
17
|
Olejarz W, Kubiak-Tomaszewska G, Chrzanowska A, Lorenc T. Exosomes in Angiogenesis and Anti-angiogenic Therapy in Cancers. Int J Mol Sci 2020; 21:ijms21165840. [PMID: 32823989 PMCID: PMC7461570 DOI: 10.3390/ijms21165840] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/09/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis is the process through which new blood vessels are formed from pre-existing ones. Exosomes are involved in angiogenesis in cancer progression by transporting numerous pro-angiogenic biomolecules like vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), and microRNAs. Exosomes promote angiogenesis by suppressing expression of factor-inhibiting hypoxia-inducible factor 1 (HIF-1). Uptake of tumor-derived exosomes (TEX) by normal endothelial cells activates angiogenic signaling pathways in endothelial cells and stimulates new vessel formation. TEX-driven cross-talk of mesenchymal stem cells (MSCs) with immune cells blocks their anti-tumor activity. Effective inhibition of tumor angiogenesis may arrest tumor progression. Bevacizumab, a VEGF-specific antibody, was the first antiangiogenic agent to enter the clinic. The most important clinical problem associated with cancer therapy using VEGF- or VEFGR-targeting agents is drug resistance. Combined strategies based on angiogenesis inhibitors and immunotherapy effectively enhances therapies in various cancers, but effective treatment requires further research.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.O.); (G.K.-T.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Grażyna Kubiak-Tomaszewska
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.O.); (G.K.-T.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Alicja Chrzanowska
- Chair and Department of Biochemistry, Medical University of Warsaw, ul. Banacha 1, 02-097 Warsaw, Poland;
| | - Tomasz Lorenc
- 1st Department of Clinical Radiology, Medical University of Warsaw, ul. Chałubińskiego 5, 02-004 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-502-1073
| |
Collapse
|
18
|
Fadaly WA, Elshaier YA, Hassanein EH, Abdellatif KR. New 1,2,4-triazole/pyrazole hybrids linked to oxime moiety as nitric oxide donor celecoxib analogs: Synthesis, cyclooxygenase inhibition anti-inflammatory, ulcerogenicity, anti-proliferative activities, apoptosis, molecular modeling and nitric oxide release studies. Bioorg Chem 2020; 98:103752. [DOI: 10.1016/j.bioorg.2020.103752] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022]
|
19
|
Lu RM, Chiu CY, Liu IJ, Chang YL, Liu YJ, Wu HC. Novel human Ab against vascular endothelial growth factor receptor 2 shows therapeutic potential for leukemia and prostate cancer. Cancer Sci 2019; 110:3773-3787. [PMID: 31578782 PMCID: PMC6890446 DOI: 10.1111/cas.14208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) is highly expressed in tumor‐associated endothelial cells, where it modulates tumor‐promoting angiogenesis, and it is also found on the surface of tumor cells. Currently, there are no Ab therapeutics targeting VEGFR2 approved for the treatment of prostate cancer or leukemia. Therefore, development of novel efficacious anti‐VEGFR2 Abs will benefit cancer patients. We used the Institute of Cellular and Organismic Biology human Ab library and affinity maturation to develop a fully human Ab, anti‐VEGFR2‐AF, which shows excellent VEGFR2 binding activity. Anti‐VEGFR2‐AF bound Ig‐like domain 3 of VEGFR2 extracellular region to disrupt the interaction between VEGF‐A and VEGFR2, neutralizing downstream signaling of the receptor. Moreover, anti‐VEGFR2‐AF inhibited capillary structure formation and exerted Ab‐dependent cell‐mediated cytotoxicity and complement‐dependent cytotoxicity in vitro. We found that VEGFR2 is expressed in PC‐3 human prostate cancer cell line and associated with malignancy and metastasis of human prostate cancer. In a PC‐3 xenograft mouse model, treatment with anti‐VEGFR2‐AF repressed tumor growth and angiogenesis as effectively and safely as US FDA‐approved anti‐VEGFR2 therapeutic, ramucirumab. We also report for the first time that addition of anti‐VEGFR2 Ab can enhance the efficacy of docetaxel in the treatment of a prostate cancer mouse model. In HL‐60 human leukemia‐xenografted mice, anti‐VEGFR2‐AF showed better efficacy than ramucirumab with prolonged survival and reduced metastasis of leukemia cells to ovaries and lymph nodes. Our findings suggest that anti‐VEGFR2‐AF has strong potential as a cancer therapy that could directly target VEGFR2‐expressing tumor cells in addition to its anti‐angiogenic action.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chiung-Yi Chiu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - I-Ju Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Ling Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yaw-Jen Liu
- Research and Development Center, United Biopharma Inc., Hsinshu, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|