1
|
Hai-Na Z, Jun-Jie J, Guang-Meng X. Peptides derived from growth factors: Exploring their diverse impact from antimicrobial properties to neuroprotection. Biomed Pharmacother 2024; 176:116830. [PMID: 38824833 DOI: 10.1016/j.biopha.2024.116830] [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: 01/29/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Growth factor-derived peptides are bioactive molecules that play a crucial role in various physiological processes within the human body. Over the years, extensive research has revealed their diverse applications, ranging from antimicrobial properties to their potential in neuroprotection and treating various diseases. These peptides exhibit innate immune responses and have been found to possess potent antimicrobial properties against a wide range of pathogens. Growth factor-derived peptides have demonstrated the ability to promote neuronal survival, prevent cell death, and stimulate neural regeneration. As a result, they hold immense promise in the treatment of various neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, as well as in the management of traumatic brain injuries. Moreover, growth factor-derived peptides have shown potential for supporting tissue repair and wound healing processes. By enhancing cell proliferation and migration, these peptides contribute to the regeneration of damaged tissues and promote a more efficient healing response. The applications of growth factor-derived peptides extend beyond their therapeutic potential in health; they also have a role in various disease conditions. For example, researchers have explored their influence on cancer cells, where some peptides have demonstrated anti-cancer properties, inhibiting tumor growth and promoting apoptosis in cancer cells. Additionally, their immunomodulatory properties have been investigated for potential applications in autoimmune disorders. Despite the immense promise shown by growth factor-derived peptides, some challenges need to be addressed. Nevertheless, ongoing research and advancements in biotechnology offer promising avenues to overcome these obstacles. The review summarizes the foundational biology of growth factors and the intricate signaling pathways in various physiological processes as well as diseases such as cancer, neurodegenerative disorders, cardiovascular ailments, and metabolic syndromes.
Collapse
Affiliation(s)
- Zhang Hai-Na
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130000, PR China
| | - Jiang Jun-Jie
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130000, PR China
| | - Xu Guang-Meng
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun 130000, PR China.
| |
Collapse
|
2
|
Lee YC, Yu JC, Ni K, Lin YC, Chen CT. Improved prediction of anti-angiogenic peptides based on machine learning models and comprehensive features from peptide sequences. Sci Rep 2024; 14:14387. [PMID: 38909149 PMCID: PMC11193773 DOI: 10.1038/s41598-024-65062-9] [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: 02/24/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Angiogenesis is a key process for the proliferation and metastatic spread of cancer cells. Anti-angiogenic peptides (AAPs), with the capability of inhibiting angiogenesis, are promising candidates in cancer treatment. We propose AAPL, a sequence-based predictor to identify AAPs with machine learning models of improved prediction accuracy. Each peptide sequence was transformed to a vector of 4335 numeric values according to 58 different feature types, followed by a heuristic algorithm for feature selection. Next, the hyperparameters of six machine learning models were optimized with respect to the feature subset. We considered two datasets, one with entire peptide sequences and the other with 15 amino acids from peptide N-termini. AAPL achieved Matthew's correlation coefficients of 0.671 and 0.756 for independent tests based on the two datasets, respectively, outperforming existing predictors by a range of 5.3% to 24.6%. Further analyses show that AAPL yields higher prediction accuracy for peptides with more hydrophobic residues, and fewer hydrophilic and charged residues. The source code of AAPL is available at https://github.com/yunzheng2002/Anti-angiogenic .
Collapse
Affiliation(s)
- Yun-Chen Lee
- Department of Computer Science and Information Engineering, Asia University, Taichung, 41354, Taiwan
| | - Jen-Chieh Yu
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
| | - Kuan Ni
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Yu-Chuan Lin
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
| | - Ching-Tai Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan.
- Center for Precision Health Research, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
3
|
Furukawa N, Yang W, Chao AR, Patil A, Mirando AC, Pandey NB, Popel AS. Chemokine-derived oncolytic peptide induces immunogenic cancer cell death and significantly suppresses tumor growth. Cell Death Discov 2024; 10:161. [PMID: 38565596 PMCID: PMC10987543 DOI: 10.1038/s41420-024-01932-5] [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: 09/19/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Chemokinostatin-1 (CKS1) is a 24-mer peptide originally discovered as an anti-angiogenic peptide derived from the CXCL1 chemokine. Here, we demonstrate that CKS1 acts not only as an anti-angiogenic peptide but also as an oncolytic peptide due to its structural and physical properties. CKS1 induced both necrotic and apoptotic cell death specifically in cancer cells while showing minimal toxicity in non-cancerous cells. Mechanistically, CKS1 disrupted the cell membrane of cancer cells quickly after treatment and activated the apoptotic pathway at later time points. Furthermore, immunogenic molecules were released from CKS1-treated cells, indicating that CKS1 induces immunogenic cell death. CKS1 effectively suppressed tumor growth in vivo. Collectively, these data demonstrate that CKS1 functions as an oncolytic peptide and has a therapeutic potential to treat cancer.
Collapse
Affiliation(s)
- Natsuki Furukawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Wendy Yang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex R Chao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akash Patil
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Furukawa N, Yang W, Chao A, Patil A, Mirando A, Pandey N, Popel A. Chemokine-derived oncolytic peptide induces immunogenic cancer cell death and significantly suppresses tumor growth. RESEARCH SQUARE 2023:rs.3.rs-3335225. [PMID: 37886580 PMCID: PMC10602061 DOI: 10.21203/rs.3.rs-3335225/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Chemokinostatin-1 (CKS1) is a 24-mer peptide originally discovered as an anti-angiogenic peptide derived from the CXCL1 chemokine. Here, we demonstrate that CKS1 acts not only as an anti-angiogenic peptide but also as an oncolytic peptide due to its structural and physical properties. CKS1 induced both necrotic and apoptotic cell death specifically in cancer cells while showing minimal toxicity in non-cancerous cells. Mechanistically, CKS1 disrupted the cell membrane of cancer cells quickly after treatment and activated the apoptotic pathway at later time points. Furthermore, immunogenic molecules were released from CKS1 treated cells, indicating that CKS1 induces immunogenic cell death. CKS1 effectively suppressed tumor growth in vivo. Collectively, these data demonstrate that CKS1 is a unique peptide that functions both as an anti-angiogenic peptide and as an oncolytic peptide and has a therapeutic potential to treat cancer.
Collapse
Affiliation(s)
| | - Wendy Yang
- Johns Hopkins University School of Medicine
| | - Alex Chao
- Johns Hopkins University School of Medicine
| | | | | | | | | |
Collapse
|
5
|
Kang MJ, Roh KH, Lee JS, Lee JH, Park S, Lim DW. Vascular Endothelial Growth Factor Receptor 1 Targeting Fusion Polypeptides with Stimuli-Responsiveness for Anti-angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37384534 DOI: 10.1021/acsami.3c03989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Genetically engineered fusion polypeptides have been investigated to introduce unique bio-functionality and improve some therapeutic activity for anti-angiogenesis. We report herein that stimuli-responsive, vascular endothelial growth factor receptor 1 (VEGFR1) targeting fusion polypeptides composed of a VEGFR1 (fms-like tyrosine kinase-1 (Flt1)) antagonist, an anti-Flt1 peptide, and a thermally responsive elastin-based polypeptide (EBP) were rationally designed at the genetic level, biosynthesized, and purified by inverse transition cycling to develop potential anti-angiogenic fusion polypeptides to treat neovascular diseases. A series of hydrophilic EBPs with different block lengths were fused with an anti-Flt1 peptide, forming anti-Flt1-EBPs, and the effect of EBP block length on their physicochemical properties was examined. While the anti-Flt1 peptide decreased phase-transition temperatures of anti-Flt1-EBPs, compared with EBP blocks, anti-Flt1-EBPs were soluble under physiological conditions. The anti-Flt1-EBPs dose dependently inhibited the binding of VEGFR1 against vascular endothelial growth factor (VEGF) as well as tube-like network formation of human umbilical vein endothelial cells under VEGF-triggered angiogenesis in vitro because of the specific binding between anti-Flt1-EBPs and VEGFR1. Furthermore, the anti-Flt1-EBPs suppressed laser-induced choroidal neovascularization in a wet age-related macular degeneration mouse model in vivo. Our results indicate that anti-Flt1-EBPs as VEGFR1-targeting fusion polypeptides have great potential for efficacious anti-angiogenesis to treat retinal-, corneal-, and choroidal neovascularization.
Collapse
Affiliation(s)
- Min Jeong Kang
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Kug-Hwan Roh
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Jae Sang Lee
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jae Hee Lee
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - SaeGwang Park
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Dong Woo Lim
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
6
|
Khalily MP, Soydan M. Peptide-based diagnostic and therapeutic agents: Where we are and where we are heading? Chem Biol Drug Des 2023; 101:772-793. [PMID: 36366980 DOI: 10.1111/cbdd.14180] [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: 07/26/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Peptides are increasingly present in all branches of medicine as innovative drugs, imaging agents, theragnostic, and constituent moieties of other sophisticated drugs such as peptide-drug conjugates. Due to new developments in chemical synthesis strategies, computational biology, recombinant technology, and chemical biology, peptide drug development has made a great progress in the last decade. Numerous natural peptides and peptide mimics have been obtained and studied, covering multiple therapeutic areas. Even though peptides have been investigated across the wide therapeutic spectrum, oncology, metabolism, and endocrinology are the most frequent medical indications of them. This review summarizes the current use of and the emerging new opportunities of peptides for diagnosis and treatment of various diseases.
Collapse
Affiliation(s)
- Melek P Khalily
- Department of Basic Science and Health, Cannabis Research Institute, Yozgat Bozok University, Yozgat, Turkey
| | - Medine Soydan
- Department of Chemistry, Faculty of Arts and Science, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
7
|
Characterisation of a novel crustin isoform from mud crab, Scylla serrata (Forsskål, 1775) and its functional analysis in silico. In Silico Pharmacol 2022; 11:2. [PMID: 36582926 PMCID: PMC9795441 DOI: 10.1007/s40203-022-00138-w] [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: 10/17/2022] [Accepted: 12/18/2022] [Indexed: 12/29/2022] Open
Abstract
A 336-base pair (bp) sized mRNA sequence encoding 111 amino acid size crustin isoform (MC-crustin) was obtained from the gill sample of the green mud crab, Scylla serrata. MC-crustin possessed an N-terminal signal peptide region comprising of 21 amino acid residues, followed by a 90 amino acid mature peptide region having a molecular weight of 10.164 kDa, charge + 4.25 and theoretical pI of 8.27. Sequence alignment and phylogenetic tree analyses revealed the peptide to be a Type I crustin, with four conserved cysteine residues forming the cysteine rich region, followed by WAP domain. MC-crustin was cationic with cysteine/proline rich structure and was predicted with antimicrobial, anti-inflammatory, anti-angiogenic and anti-hypertensive property making it a potential molecule for possible therapeutic applications.
Collapse
|
8
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
9
|
Karami Fath M, Babakhaniyan K, Anjomrooz M, Jalalifar M, Alizadeh SD, Pourghasem Z, Abbasi Oshagh P, Azargoonjahromi A, Almasi F, Manzoor HZ, Khalesi B, Pourzardosht N, Khalili S, Payandeh Z. Recent Advances in Glioma Cancer Treatment: Conventional and Epigenetic Realms. Vaccines (Basel) 2022; 10:1448. [PMID: 36146527 PMCID: PMC9501259 DOI: 10.3390/vaccines10091448] [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: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most typical and aggressive form of primary brain tumor in adults, with a poor prognosis. Successful glioma treatment is hampered by ineffective medication distribution across the blood-brain barrier (BBB) and the emergence of drug resistance. Although a few FDA-approved multimodal treatments are available for glioblastoma, most patients still have poor prognoses. Targeting epigenetic variables, immunotherapy, gene therapy, and different vaccine- and peptide-based treatments are some innovative approaches to improve anti-glioma treatment efficacy. Following the identification of lymphatics in the central nervous system, immunotherapy offers a potential method with the potency to permeate the blood-brain barrier. This review will discuss the rationale, tactics, benefits, and drawbacks of current glioma therapy options in clinical and preclinical investigations.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Kimiya Babakhaniyan
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran 1996713883, Iran
| | - Mehran Anjomrooz
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1411713135, Iran
| | | | | | - Zeinab Pourghasem
- Department of Microbiology, Islamic Azad University of Lahijan, Gilan 4416939515, Iran
| | - Parisa Abbasi Oshagh
- Department of Biology, Faculty of Basic Sciences, Malayer University, Malayer 6571995863, Iran
| | - Ali Azargoonjahromi
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz 7417773539, Iran
| | - Faezeh Almasi
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 1411734115, Iran
| | - Hafza Zahira Manzoor
- Experimental and Translational Medicine, University of Insubria, Via jean Henry Dunant 3, 21100 Varese, Italy
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
| | - Navid Pourzardosht
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht 4193713111, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran
| | - Zahra Payandeh
- Department of Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, SE-17177 Stockholm, Sweden
| |
Collapse
|
10
|
Bose D, Roy L, Chatterjee S. Peptide therapeutics in the management of metastatic cancers. RSC Adv 2022; 12:21353-21373. [PMID: 35975072 PMCID: PMC9345020 DOI: 10.1039/d2ra02062a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/26/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer remains a leading health concern threatening lives of millions of patients worldwide. Peptide-based drugs provide a valuable alternative to chemotherapeutics as they are highly specific, cheap, less toxic and easier to synthesize compared to other drugs. In this review, we have discussed various modes in which peptides are being used to curb cancer. Our review highlights specially the various anti-metastatic peptide-based agents developed by targeting a plethora of cellular factors. Herein we have given a special focus on integrins as targets for peptide drugs, as these molecules play key roles in metastatic progression. The review also discusses use of peptides as anti-cancer vaccines and their efficiency as drug-delivery tools. We hope this work will give the reader a clear idea of the mechanisms of peptide-based anti-cancer therapeutics and encourage the development of superior drugs in the future.
Collapse
Affiliation(s)
- Debopriya Bose
- Department of Biophysics Bose Institute Unified Academic Campus EN 80, Sector V, Bidhan Nagar Kolkata 700091 WB India
| | - Laboni Roy
- Department of Biophysics Bose Institute Unified Academic Campus EN 80, Sector V, Bidhan Nagar Kolkata 700091 WB India
| | - Subhrangsu Chatterjee
- Department of Biophysics Bose Institute Unified Academic Campus EN 80, Sector V, Bidhan Nagar Kolkata 700091 WB India
| |
Collapse
|
11
|
Ionescu C, Oprea B, Ciobanu G, Georgescu M, Bică R, Mateescu GO, Huseynova F, Barragan-Montero V. The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview. Medicina (B Aires) 2022; 58:medicina58070903. [PMID: 35888622 PMCID: PMC9316440 DOI: 10.3390/medicina58070903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is the process of developing new blood vessels from pre-existing ones. This review summarizes the main features of physiological and pathological angiogenesis and those of angiogenesis activation and inhibition. In healthy adults, angiogenesis is absent apart from its involvement in female reproductive functions and tissue regeneration. Angiogenesis is a complex process regulated by the action of specific activators and inhibitors. In certain diseases, modulating the angiogenic balance can be a therapeutic route, either by inhibiting angiogenesis (for example in the case of tumor angiogenesis), or by trying to activate the process of new blood vessels formation, which is the goal in case of cardiac or peripheral ischemia.
Collapse
Affiliation(s)
- Cătălina Ionescu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania;
- Correspondence: (C.I.); (B.O.)
| | - Bogdan Oprea
- Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania;
- Correspondence: (C.I.); (B.O.)
| | - Georgeta Ciobanu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania;
| | - Milena Georgescu
- Clinic for Plastic Surgery and Burns, County Emergency Hospital Craiova, 200642 Craiova, Romania;
| | - Ramona Bică
- General Hospital—“Victor Babes”, 281 Mihai Bravu St., Sector III, 030303 Bucharest, Romania;
| | - Garofiţa-Olivia Mateescu
- Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania;
| | - Fidan Huseynova
- LBN, University of Montpellier, 34193 Montpellier, France; (F.H.); (V.B.-M.)
- Institute of Molecular Biology and Biotechnologies, Azerbaïjan National Academy of Sciences (ANAS), AZ1073 Baku, Azerbaijan
- Department of Histology, Cytology and Embryology, Azerbaijan Medical University, AZ1078 Baku, Azerbaijan
| | | |
Collapse
|
12
|
Iwanov I, Rossi A, Montesi M, Doytchinova I, Sargsyan A, Momekov G, Panseri S, Naydenova E. Peptide-based targeted cancer therapeutics: design, synthesis and biological evaluation. Eur J Pharm Sci 2022; 176:106249. [PMID: 35779821 DOI: 10.1016/j.ejps.2022.106249] [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: 04/12/2022] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022]
Abstract
Cancer is the leading cause for human mortality together with cardiovascular diseases. Abl (Abelson) tyrosine kinases play a fundamental role in transducing various signals that control proliferation, survival, migration and invasion in several cancers such as Chronic Myeloid Leukemia (CML), breast cancer and brain cancer. For these reasons Abl tyrosine kinases are considered important biological targets in drug discovery. In this study a series of lysine-based oligopeptides with expected Abl inhibitory activity were designed resembling the binding of FDA-approved drugs (i.e. of Imatinib and Nilotinib), synthesized, purified by High Performance Liquid Chromatography (HPLC), analyzed by mass spectrometry (MS) and biologically tested in vitro in CML (AR-230 and K-562), breast cancers (MDA-MB 231 and MDA-MB 468) and glioblastoma cell lines (U87 and U118). The solid-phase peptide synthesis (SPPS) by Fmoc (9-fluorenylmethoxycarbonyl) chemistry was used to synthesize target compounds. AutoDock Vina was applied for simulation binding to Abl. The biological activities were measured evaluating cytotoxic effect, induction of apoptosis and inhibition of cancer cells migration. The new peptides exhibited different concentration-dependent antiproliferative effect against the tumor cell lines after 72 h treatment. The most promising results were obtained with the U87 glioblastoma cell line where a significant reduction of the migration ability was detected with one compound (H-Lys1-Lys2-Lys3-NH2).
Collapse
Affiliation(s)
- Iwan Iwanov
- University of Chemical Technology and Metallurgy, 8 Blvd. Kliment Ohridski, 1756, Sofia, Bulgaria
| | - Arianna Rossi
- Institute of Science and Technology for Ceramics, National Research Council of Italy, via Granarolo 64, Faenza (RA), Italy; University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Piazza Pugliatti 1, Messina (ME), Italy
| | - Monica Montesi
- Institute of Science and Technology for Ceramics, National Research Council of Italy, via Granarolo 64, Faenza (RA), Italy
| | | | - Armen Sargsyan
- Scientific and Production Center "Armbiotechnology" NAS RA, 14 Gyurjyan str., Yerevan, 0056, Armenia
| | - Georgi Momekov
- Medical University of Sofia, 2 Dunav st., Sofia, 1000, Bulgaria
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics, National Research Council of Italy, via Granarolo 64, Faenza (RA), Italy.
| | - Emilia Naydenova
- University of Chemical Technology and Metallurgy, 8 Blvd. Kliment Ohridski, 1756, Sofia, Bulgaria.
| |
Collapse
|
13
|
Akbarian M, Bertassoni LE, Tayebi L. Biological aspects in controlling angiogenesis: current progress. Cell Mol Life Sci 2022; 79:349. [PMID: 35672585 PMCID: PMC10171722 DOI: 10.1007/s00018-022-04348-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 12/25/2022]
Abstract
All living beings continue their life by receiving energy and by excreting waste products. In animals, the arteries are the pathways of these transfers to the cells. Angiogenesis, the formation of the arteries by the development of pre-existed parental blood vessels, is a phenomenon that occurs naturally during puberty due to certain physiological processes such as menstruation, wound healing, or the adaptation of athletes' bodies during exercise. Nonetheless, the same life-giving process also occurs frequently in some patients and, conversely, occurs slowly in some physiological problems, such as cancer and diabetes, so inhibiting angiogenesis has been considered to be one of the important strategies to fight these diseases. Accordingly, in tissue engineering and regenerative medicine, the highly controlled process of angiogenesis is very important in tissue repairing. Excessive angiogenesis can promote tumor progression and lack of enough angiogensis can hinder tissue repair. Thereby, both excessive and deficient angiogenesis can be problematic, this review article introduces and describes the types of factors involved in controlling angiogenesis. Considering all of the existing strategies, we will try to lay out the latest knowledge that deals with stimulating/inhibiting the angiogenesis. At the end of the article, owing to the early-reviewed mechanical aspects that overshadow angiogenesis, the strategies of angiogenesis in tissue engineering will be discussed.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA.
| |
Collapse
|
14
|
Chamani R, Taleqani MH, Imanpour A, Khatami M. New insights into short peptides derived from the collagen NC1 α1, α2, and α3 (IV) domains: An experimental and MD simulations study. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140769. [PMID: 35151893 DOI: 10.1016/j.bbapap.2022.140769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 10/19/2022]
Abstract
Proteolytically cleavage of the collagen NC1 α1 to α3 (IV) domains leads to antiangiogenic proteins called Arresten, Canstatin, and Tumstatin, respectively. The research identified that the two overlapping peptides derived from Tumstatin are more effective than other fragments and amino acids L78, V82, and D84 are essential for their activity. In the present study, the efficacy of a nine amino acid peptide derived from Tumstatin (Tum), containing amino acids L78, V82, and D84 was compared to the corresponding sequence in Arresten (Ars) and Canstatin (Can) in vitro and in vivo. Moreover, CD spectroscopy, MD, and docking simulations were performed to evaluate the structure and the interaction of peptides to integrin αvβ3, respectively. Results demonstrated that peptides inhibit viability, migration, and tube formation in vitro, as well as the growth of tumor in vivo and Canstatin-derived peptide was more potent than others. CD measurement and DSSP calculation revealed that Can had more coil conformation. According to MD simulations, Can had more fluctuation, less intramolecular interactions, and less structural compactness compared to Tum and Ars. It can be assumed that amino acid variations lead to a more flexible and loose structure compared to the other peptides. The Canstatin-derived peptide interacts with the integrin αvβ3 extremely close to RGD binding site by the most negative binding energy and more interactions. In conclusion, we for the first time identified an active peptide derived from Canstatin and showed that the sequence affected structure and thereby interaction of peptide to its receptor.
Collapse
Affiliation(s)
- Reyhane Chamani
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran.
| | | | - Aylar Imanpour
- Department of Fisheries, Faculty of Natural Resources, Urmia University, Urmia, Iran
| | - Mehri Khatami
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| |
Collapse
|
15
|
Maia VSC, Berzaghi R, Arruda DC, Machado FC, Loureiro LL, Melo PMS, Morais AS, Budu A, Travassos LR. PLP2-derived peptide Rb4 triggers PARP-1-mediated necrotic death in murine melanoma cells. Sci Rep 2022; 12:2890. [PMID: 35190586 PMCID: PMC8861012 DOI: 10.1038/s41598-022-06429-8] [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: 07/09/2018] [Accepted: 01/12/2022] [Indexed: 12/28/2022] Open
Abstract
Malignant melanoma is the main cause of death in patients with skin cancer. Overexpression of Proteolipid protein 2 (PLP2) increased tumor metastasis and the knockdown of PLP2 inhibited the growth and metastasis of melanoma cells. In the present work, we studied the antitumor activity of peptide Rb4 derived from protein PLP2. In vitro, Rb4 induced F-actin polymerization, prevented F-actin depolymerization and increased the ER-derived cytosolic calcium. Such effects were associated with necrosis of murine melanoma B16F10-Nex2 cells and with inhibition of the viability of human cancer cell lines. Loss of plasma membrane integrity, dilation of mitochondria, cytoplasm vacuolation and absence of chromatin condensation characterized tumor cell necrosis. Cleavage of PARP-1 and inhibition of RIP1 expression were also observed. In vivo, peptide Rb4 reduced the lung metastasis of tumor cells and delayed the subcutaneous melanoma growth in a syngeneic model. Rb4 induced the expression of two DAMPs molecules, HMGB1 and calreticulin, in B16F10-Nex2. Our results suggest that peptide Rb4 acts directly on tumor cells inducing the expression of DAMPs, which trigger the immunoprotective effect in vivo against melanoma cells. We suggest that peptide Rb4 is a promising compound to be developed as an anticancer drug.
Collapse
Affiliation(s)
| | - Rodrigo Berzaghi
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Denise C Arruda
- Integrated Group of Biotechnology, University of Mogi das Cruzes, UMC, Mogi das Cruzes, SP, Brazil
| | - Fabrício C Machado
- Recepta Biopharma, São Paulo, Brazil.,Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Leticia L Loureiro
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Pollyana M S Melo
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | | | - Alexandre Budu
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Luiz R Travassos
- Recepta Biopharma, São Paulo, Brazil.,Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Robles JP, Zamora M, Siqueiros-Marquez L, Adan-Castro E, Ramirez-Hernandez G, Nuñez FF, Lopez-Casillas F, Millar RP, Bertsch T, Martínez de la Escalera G, Triebel J, Clapp C. The HGR motif is the antiangiogenic determinant of vasoinhibin: implications for a therapeutic orally active oligopeptide. Angiogenesis 2022; 25:57-70. [PMID: 34097181 PMCID: PMC8813873 DOI: 10.1007/s10456-021-09800-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
The hormone prolactin acquires antiangiogenic and antivasopermeability properties after undergoing proteolytic cleavage to vasoinhibin, an endogenous prolactin fragment of 123 or more amino acids that inhibits the action of multiple proangiogenic factors. Preclinical and clinical evidence supports the therapeutic potential of vasoinhibin against angiogenesis-related diseases including diabetic retinopathy, peripartum cardiomyopathy, rheumatoid arthritis, and cancer. However, the use of vasoinhibin in the clinic has been limited by difficulties in its production. Here, we removed this barrier to using vasoinhibin as a therapeutic agent by showing that a short linear motif of just three residues (His46-Gly47-Arg48) (HGR) is the functional determinant of vasoinhibin. The HGR motif is conserved throughout evolution, its mutation led to vasoinhibin loss of function, and oligopeptides containing this sequence inhibited angiogenesis and vasopermeability with the same potency as whole vasoinhibin. Furthermore, the oral administration of an optimized cyclic retro-inverse vasoinhibin heptapeptide containing HGR inhibited melanoma tumor growth and vascularization in mice and exhibited equal or higher antiangiogenic potency than other antiangiogenic molecules currently used as anti-cancer drugs in the clinic. Finally, by unveiling the mechanism that obscures the HGR motif in prolactin, we anticipate the development of vasoinhibin-specific antibodies to solve the on-going challenge of measuring endogenous vasoinhibin levels for diagnostic and interventional purposes, the design of vasoinhibin antagonists for managing insufficient angiogenesis, and the identification of putative therapeutic proteins containing HGR.
Collapse
Affiliation(s)
- Juan Pablo Robles
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Magdalena Zamora
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | | | - Elva Adan-Castro
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | | | - Francisco Freinet Nuñez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Fernando Lopez-Casillas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México City, México
| | - Robert P Millar
- Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital & Paracelsus Medical University, Nuremberg, Germany
| | | | - Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital & Paracelsus Medical University, Nuremberg, Germany
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México.
| |
Collapse
|
17
|
Lin C, Wang L, Shi L. AAPred-CNN: accurate predictor based on deep convolution neural network for identification of anti-angiogenic peptides. Methods 2022; 204:442-448. [PMID: 35031486 DOI: 10.1016/j.ymeth.2022.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/28/2021] [Accepted: 01/09/2022] [Indexed: 12/13/2022] Open
Abstract
Recently, deep learning techniques have been developed for various bioactive peptide prediction tasks. However, there are only conventional machine learning-based methods for the prediction of anti-angiogenic peptides (AAP), which play an important role in cancer treatment. The main reason why no deep learning method has been involved in this field is that there are too few experimentally validated AAPs to support the training of deep models but researchers have believed that deep learning seriously depends on the amounts of labeled data. In this paper, as a tentative work, we try to predict AAP by constructing different classical deep learning models and propose the first deep convolution neural network-based predictor (AAPred-CNN) for AAP. Contrary to intuition, the experimental results show that deep learning models can achieve superior or comparable performance to the state-of-the-art model, although they are given a few labeled sequences to train. We also decipher the influence of hyper-parameters and training samples on the performance of deep learning models to help understand how the model work. Furthermore, we also visualize the learned embeddings by dimension reduction to increase the model interpretability and reveal the residue propensity of AAP through the statistics of convolutional features for different residues. In summary, this work demonstrates the powerful representation ability of AAPred-CNNfor AAP prediction, further improving the prediction accuracy of AAP.
Collapse
Affiliation(s)
- Changhang Lin
- School of Big Data and Artificial Intelligence, Fujian Polytechnic Normal University, Fuzhou, China
| | - Lei Wang
- Beidahuang Industry Group General Hospital, Harbin, China.
| | - Lei Shi
- Department of Spine Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
18
|
Thakur R, Suri CR, Kaur IP, Rishi P. Review. Crit Rev Ther Drug Carrier Syst 2022; 40:49-100. [DOI: 10.1615/critrevtherdrugcarriersyst.2022040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Shoari A, Khodabakhsh F, Ahangari Cohan R, Salimian M, Karami E. Anti-angiogenic peptides application in cancer therapy; a review. Res Pharm Sci 2021; 16:559-574. [PMID: 34760005 PMCID: PMC8562409 DOI: 10.4103/1735-5362.327503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/16/2021] [Accepted: 09/19/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer is a disease advanced via surplus angiogenesis. The development of new anti-angiogenic therapeutic agents with more efficacy and fewer side effects is still quite necessary. Conventional therapies saving the life of many cancer patients but due to drug resistance and lack of specificity utilizing these methods is faced with limits. Recently, new therapeutic agents have been developed and used to treat cancers such as scaffold proteins, monoclonal antibodies, tyrosine kinase inhibitors, and peptides. In antiangiogenic drug development, anti-angiogenic peptides design is a significant aim. Peptides have developed as substantial therapeutics that are being carefully investigated in angiogenesis-dependent diseases because of their high penetrating rate into the cancer cells, high specificity, and low toxicity. In this review, we focus on anti-angiogenic peptides in the field of cancer therapy that are designed, screened, or derived from nanobodies, mimotopes, phage displays, and natural resources.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, I.R. Iran
| | - Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, I.R. Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, I.R. Iran
| | - Morteza Salimian
- Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Elmira Karami
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, I.R. Iran
| |
Collapse
|
20
|
Hazrati F, Saidijam M, Ahmadyousefi Y, Nouri F, Ghadimipour H, Moradi M, Haddadi R, Soleimani M. A novel chimeric protein with enhanced cytotoxic effects on breast cancer in vitro and in vivo. Proteins 2021; 90:936-946. [PMID: 34812523 DOI: 10.1002/prot.26285] [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: 07/25/2021] [Revised: 10/31/2021] [Accepted: 11/15/2021] [Indexed: 11/09/2022]
Abstract
In our previous study, we reported the design and recombinant production of the p28-apoptin as a novel chimeric protein for breast cancer (BC) treatment. This study aimed to evaluate the inhibitory activity of the chimeric protein against BC cells in vitro and in vivo. We developed a novel multifunctional protein, consisting of p28, as a tumor-homing killer peptide fused to apoptin as a tumor-selective killer. The chimeric protein showed significantly higher toxicity in BC cell lines dose-dependently than in non-cancerous control cell lines. IC50 values were 1.41, 1.38, 6.13, and 264.49 μM for 4T1, MDA-MB-468, Vero, and HEK293 cells, respectively. The protein showed significantly enhanced uptake in 4T1 cancer cells compared with non-cancerous Vero cells. We also showed that the p28-apoptin chimeric protein binds significantly higher to human breast cancer tumor sections than the normal human breast tissue section. Also, significant apoptosis induction and tumor growth inhibition were observed in established tumor-bearing mice accompanied by a decreased frequency of metastases. Our results support that the chimeric protein has inhibitory activity in vitro and in vivo, making it a promising choice in targeted cancer therapy.
Collapse
Affiliation(s)
- Fereshte Hazrati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Genetics and Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Genetics and Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Nouri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamidreza Ghadimipour
- Department of Pathology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadreza Moradi
- Department of Genetics and Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology Toxicology, School of Pharmacy Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Meysam Soleimani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
21
|
Malviya R, Verma S, Sundram S. Advancement and Strategies for the Development of Peptide-Drug Conjugates: Pharmacokinetic Modulation, Role and Clinical Evidence Against Cancer Management. Curr Cancer Drug Targets 2021; 22:286-311. [PMID: 34792003 DOI: 10.2174/1568009621666211118111506] [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: 05/11/2021] [Revised: 07/29/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022]
Abstract
Currently, many new treatment strategies are being used for the management of cancer. Among them, chemotherapy based on peptides has been of great interest due to the unique features of peptides. This review discusses the role of peptide and peptides analogues in the treatment of cancer, with special emphasis on their pharmacokinetic modulation and research progress. Low molecular weight, targeted drug delivery, enhanced permeability, etc., of the peptide-linked drug conjugates, lead to an increase in the effectiveness of cancer therapy. Various peptides have recently been developed as drugs and vaccines with an altered pharmacokinetic parameter which has subsequently been assessed in different phases of the clinical study. Peptides have made a great impact in the area of cancer therapy and diagnosis. Targeted chemotherapy and drug delivery techniques using peptides are emerging as excellent tools in minimizing problems with conventional chemotherapy. It can be concluded that new advances in using peptides to treat different types of cancer have been shown by different clinical studies indicating that peptides could be used as an ideal therapeutic method in treating cancer due to the novel advantages of peptides. The development of identifying and synthesizing novel peptides could provide a promising choice to patients with cancer.
Collapse
Affiliation(s)
- Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida. India
| | - Swati Verma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida. India
| | - Sonali Sundram
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida. India
| |
Collapse
|
22
|
Ghasemali S, Farajnia S, Barzegar A, Rahmati-Yamchi M, Negahdari B, Rahbarnia L, Yousefi-Nodeh H. Rational Design of Anti-Angiogenic Peptides to Inhibit VEGF/VEGFR2 Interactions for Cancer Therapeutics. Anticancer Agents Med Chem 2021; 22:ACAMC-EPUB-118914. [PMID: 34792006 DOI: 10.2174/1871520621666211118104051] [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: 02/19/2021] [Revised: 07/19/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Angiogenesis is a critical physiological process that plays a key role in tumor progression, metastatic dissemination, and invasion. In the last two decades, the vascular endothelial growth factor (VEGF) signaling pathway has been the area of extensive researches. VEGF executes its special effects by binding to vascular endothelial growth factor receptors (VEGFRs), particularly VEGFR-2. OBJECTIVE The inhibition of VEGF/VEGFR2 interaction is known as an effective cancer therapy strategy. The current study pointed to design and model an anti-VEGF peptide based on VEGFR2 binding regions. METHOD The large-scale peptide mutation screening was used to achieve a potent peptide with high binding affinity to VEGF for possible application in inhibition of VEGF/VEGFR2 interaction. The AntiCP and Peptide Ranker servers were used to generate the possible peptides library with anticancer activities and prediction of peptides bioactivity. Then, the interaction of VEGF and all library peptides were analyzed using Hex 8.0.0 and ClusPro tools. A number of six peptides with favorable docking scores were achieved. All of the best docking scores of peptides in complexes with VEGF were evaluated to confirm their stability, using molecular dynamics simulation (MD) with the help of the GROMACS software package. RESULTS As a result, two antiangiogenic peptides with 13 residues of PepA (NGIDFNRDFFLGL) and PepC (NGIDFNRDKFLFL) were achieved and introduced to inhibit VEGF/VEGFR2 interactions. CONCLUSIONS In summary, this study provided new insights into peptide-based therapeutics development for targeting VEGF signaling pathway in tumor cells. PepA and PepC are recommended as potentially promising anticancer agents for further experimental evaluations.
Collapse
Affiliation(s)
- Samaneh Ghasemali
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Safar Farajnia
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Abolfazl Barzegar
- Research Center of Bioscience and Biotechnology, University of Tabriz, Tabriz. Iran
| | - Mohammad Rahmati-Yamchi
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran. Iran
| | - Leila Rahbarnia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Hamidreza Yousefi-Nodeh
- Research Center for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
23
|
Zhang Y, He J. Tumor vasculature-targeting nanomedicines. Acta Biomater 2021; 134:1-12. [PMID: 34271167 DOI: 10.1016/j.actbio.2021.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/15/2022]
Abstract
Uncontrolled tumor growth and subsequent distant metastasis are highly dependent on an adequate nutrient supply from tumor blood vessels, which have relatively different pathophysiological characteristics from those of normal vasculature. Obviously, strategies targeting tumor vasculature, such as anti-angiogenic drugs and vascular disrupting agents, are attractive methods for cancer therapy. However, the off-target effects and high dose administration of these drug regimens critically restrict their clinical applications. In recent years, nanomedicines focused on tumor vasculature have been shown to be superior to traditional therapeutic methods and do not induce side effects. This review will first highlight the recent development of tumor vasculature-targeting nanomedicines from the following four aspects: 1) angiogenesis-inhibiting nanomedicines (AINs); 2) vasculature-disrupting nanomedicines (VDNs); 3) vasculature infarction nanomedicines (VINs); and 4) vasculature-regulating nanomedicines (VRNs). Furthermore, the design principles, limitations, and future directions are also discussed. STATEMENT OF SIGNIFICANCE: Based on the essential roles of tumor blood vessels, the therapeutic strategies targeting tumor vasculature have exhibited good clinical therapeutic outcomes. However, poor patient adherence to free drug administration limits their clinical usage. Nanomedicines have great potential to overcome the abovementioned obstacle. This review summarizes the tumor-vasculature targeting nanomedicines from four aspects: 1) angiogenesis-inhibiting nanomedicines (AINs); 2) vasculature-disrupting nanomedicines (VDNs); 3) vasculature infarction nanomedicines (VINs); and 4) vasculature regulating nanomedicines (VRNs). In addition, this review provides perspectives on this research field.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, PR China
| | - Jingni He
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, PR China.
| |
Collapse
|
24
|
Design and In Silico Evaluation of a Novel Cyclic Disulfide-Rich anti-VEGF Peptide as a Potential Antiangiogenic Drug. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10250-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
25
|
A Multifunctional Nanoplatform Made of Gold Nanoparticles and Peptides Mimicking the Vascular Endothelial Growth Factor. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11146333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this work, nanobiohybrids of plasmonic gold nanoparticles (AuNP, anti-angiogenic) and a peptide mimicking the vascular endothelial growth factor (VEGF, pro-angiogenic) were assembled and scrutinized in terms of physicochemical characterization, including optical properties, surface charge, surface chemical structure and morphology of the bioengineered metal nanoparticles, for their potential application as multifunctional theranostic (i.e., therapy + sensing) nanoplatform (AuNP/VEGF). Specifically, a peptide sequence encompassing the VEGF cellular receptor domain 73–101 (VEGF73–101) and its single point cysteine mutated were immobilized onto AuNP by physi- and chemi-sorption, respectively. The new hybrid systems were characterized by means of a multitechnique approach, including dynamic light scattering (DLS) analyses, zeta potential (ZP), spectroscopic (UV-Vis, FT-IR, XPS), spectrometric (TOF-SIMS) and microscopic (AFM, SEM) techniques. Proof-of-work cellular experiments in human umbilical vein endothelial cells (HUVEC) upon the treatment with AuNP/VEGF samples, demonstrated no toxicity up to 24 h (MTT assay) as well an effective internalization (laser confocal microscopy, LSM).
Collapse
|
26
|
Ellingson BM, Patel K, Wang C, Raymond C, Brenner A, de Groot JF, Butowski NA, Zach L, Campian JL, Schlossman J, Rizvi S, Cohen YC, Lowenton-Spier N, Minei TR, Shmueli SF, Wen PY, Cloughesy TF. Validation of diffusion MRI as a biomarker for efficacy using randomized phase III trial of bevacizumab with or without VB-111 in recurrent glioblastoma. Neurooncol Adv 2021; 3:vdab082. [PMID: 34377989 PMCID: PMC8350152 DOI: 10.1093/noajnl/vdab082] [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] [Indexed: 11/14/2022] Open
Abstract
Background Evidence from single and multicenter phase II trials have suggested diffusion MRI is a predictive imaging biomarker for survival benefit in recurrent glioblastoma (rGBM) treated with anti-VEGF therapy. The current study confirms these findings in a large, randomized phase III clinical trial. Methods Patients with rGBM were enrolled in a phase III randomized (1:1), controlled trial (NCT02511405) to compare the efficacy and safety of bevacizumab (BV) versus BV in combination with ofranergene obadenovec (BV+VB-111), an anti-cancer viral therapy. In 170 patients with diffusion MRI available, pretreatment enhancing tumor volume and ADC histogram analysis were used to phenotype patients as having high (>1.24 µm2/ms) or low (<1.24 µm2/ms) ADCL, the mean value of the lower peak of the ADC histogram, within the contrast enhancing tumor. Results Baseline tumor volume (P = .3460) and ADCL (P = .2143) did not differ between treatment arms. Univariate analysis showed patients with high ADCL had a significant survival advantage in all patients (P = .0006), as well as BV (P = .0159) and BV+VB-111 individually (P = .0262). Multivariable Cox regression accounting for treatment arm, age, baseline tumor volume, and ADCL identified continuous measures of tumor volume (P < .0001; HR = 1.0212) and ADCL phenotypes (P = .0012; HR = 0.5574) as independent predictors of OS. Conclusion Baseline diffusion MRI and tumor volume are independent imaging biomarkers of OS in rGBM treated with BV or BV+VB-111.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA Neuro Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Kunal Patel
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Chencai Wang
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Andrew Brenner
- University of Texas Health San Antonio Cancer Center, San Antonio, Texas, USA
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Leor Zach
- Oncology Institute, Chaim Sheba Medical Center, Tel HaShomer, Israel
| | - Jian L Campian
- Division of Medical Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jacob Schlossman
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Shan Rizvi
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | | | | | | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Timothy F Cloughesy
- UCLA Neuro Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
27
|
Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers (Basel) 2021; 13:cancers13092053. [PMID: 33922795 PMCID: PMC8122975 DOI: 10.3390/cancers13092053] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Metastasis, the process by which cancer cells escape primary tumor site and colonize distant organs, is responsible for most cancer-related deaths. The tumor microenvironment (TME), comprises different cell types, including immune cells and cancer-associated fibroblasts, as well as structural elements, such as collagen and hyaluronan that constitute the extracellular matrix (ECM). Intratumoral interactions between the cellular and structural components of the TME regulate the aggressiveness, and dissemination of malignant cells and promote immune evasion. At the secondary site, the TME also facilitates escape from dormancy to enhance metastatic tumor outgrowth. Moreover, the ECM applies mechanical forces on tumors that contribute to hypoxia and cancer cell invasiveness whereas also hinders drug delivery and efficacy in both primary and metastatic sites. In this review, we summarize the latest developments regarding the role of the TME in cancer progression and discuss ongoing efforts to remodel the TME to stop metastasis in its tracks. Abstract The tumor microenvironment (TME) regulates essential tumor survival and promotion functions. Interactions between the cellular and structural components of the TME allow cancer cells to become invasive and disseminate from the primary site to distant locations, through a complex and multistep metastatic cascade. Tumor-associated M2-type macrophages have growth-promoting and immunosuppressive functions; mesenchymal cells mass produce exosomes that increase the migratory ability of cancer cells; cancer associated fibroblasts (CAFs) reorganize the surrounding matrix creating migration-guiding tracks for cancer cells. In addition, the tumor extracellular matrix (ECM) exerts determinant roles in disease progression and cancer cell migration and regulates therapeutic responses. The hypoxic conditions generated at the primary tumor force cancer cells to genetically and/or epigenetically adapt in order to survive and metastasize. In the circulation, cancer cells encounter platelets, immune cells, and cytokines in the blood microenvironment that facilitate their survival and transit. This review discusses the roles of different cellular and structural tumor components in regulating the metastatic process, targeting approaches using small molecule inhibitors, nanoparticles, manipulated exosomes, and miRNAs to inhibit tumor invasion as well as current and future strategies to remodel the TME and enhance treatment efficacy to block the detrimental process of metastasis.
Collapse
Affiliation(s)
- Christiana M. Neophytou
- European University Research Center, Nicosia 2404, Cyprus;
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.P.); (T.S.)
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.P.); (T.S.)
| | - Panagiotis Papageorgis
- European University Research Center, Nicosia 2404, Cyprus;
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus
- Correspondence: ; Tel.: +357-22-713158
| |
Collapse
|
28
|
Abstract
Osteoporosis and cancer are becoming a major public health problem. Some studies have shown that osteoporosis drugs may have anti-cancer effects. To better understand the relationship between drugs for osteoporosis and antineoplastic agents, and to better demonstrate recent developments for patents concerning drugs for osteoporosis, we conducted an analysis of US patents. The results indicated that there was a good correlation between agents for osteoporosis and antineoplastic agents, which indicated that numerous anti-osteoporosis agents displayed antineoplastic activities. Our study was the first one to provide new evidence, through comprehensive analysis, for a correlation between anti-osteoporosis agents and anticancer agents. The present study may open new avenues for developing anticancer drugs and expanding the application role of anti-osteoporosis agents.
Collapse
|
29
|
Furukawa N, Popel AS. Peptides that immunoactivate the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2021; 1875:188486. [PMID: 33276025 PMCID: PMC8369429 DOI: 10.1016/j.bbcan.2020.188486] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/04/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy has achieved positive clinical outcomes and is revolutionizing cancer treatment. However, cancer immunotherapy has thus far failed to improve outcomes for most "cold tumors", which are characterized by low infiltration of immune cells and immunosuppressive tumor microenvironment. Enhancing the responsiveness of cold tumors to cancer immunotherapy by stimulating the components of the tumor microenvironment is a strategy pursued in the last decade. Currently, most of the agents used to modify the tumor microenvironment are small molecules or antibodies. Small molecules exhibit low affinity and specificity towards the target and antibodies have shortcomings such as poor tissue penetration and high production cost. Peptides may overcome these drawbacks and therefore are promising materials for immunomodulating agents. Here we systematically summarize the currently developed immunoactivating peptides and discuss the potential of peptide therapeutics in cancer immunology.
Collapse
Affiliation(s)
- Natsuki Furukawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
30
|
Sarkar B, Siddiqui Z, Kim KK, Nguyen PK, Reyes X, McGill TJ, Kumar VA. Implantable anti-angiogenic scaffolds for treatment of neovascular ocular pathologies. Drug Deliv Transl Res 2020; 10:1191-1202. [PMID: 32232681 PMCID: PMC7483832 DOI: 10.1007/s13346-020-00753-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The retinal physiology can accrue oxidative damage and inflammatory insults due to age and metabolic irregularities. Two notable diseases that involve retinal and choroidal neovascularization are proliferative diabetic retinopathy and wet age-related macular degeneration. Currently, these diseases are mainly treated with anti-VEGF drugs (VEGF = vascular endothelial growth factor), generally on a monthly dosage scheme. We discuss recent developments for the treatment of these diseases, including bioactive tissue-engineered materials, which may reduce frequency of dosage and propose a path forward for improving patient outcomes. Graphical abstract Development of materials for long-term intravitreal delivery for management of posterior segment diseases.
Collapse
Affiliation(s)
- Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Peter K Nguyen
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Xavier Reyes
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Trevor J McGill
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA.
| |
Collapse
|
31
|
Mehrotra N, Kharbanda S, Singh H. Peptide-based combination nanoformulations for cancer therapy. Nanomedicine (Lond) 2020; 15:2201-2217. [PMID: 32914691 DOI: 10.2217/nnm-2020-0220] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Research in cancer therapy is moving towards the use of biomolecules in combination with conventional approaches for improved disease outcome. Among the biomolecules explored, peptides are strong contenders due to their small size, high specificity, low systemic toxicity and wide inter/intracellular targets. The use of nanoformulations for such combination approaches can lead to further improvement in efficacy by reducing off-target cytotoxicity, increasing circulation time, tumor penetration and accumulation. This review focuses on nanodelivery systems for peptide-based combinations with chemo, immuno, radiation and hormone therapy. It gives an overview of the latest therapeutic research being conducted using combination nanoformulations with anticancer peptides, cell penetrating/tumor targeting peptides, peptide nanocarriers, peptidomimetics, peptide-based hormones and peptide vaccines. The challenges hindering clinical translation are also discussed.
Collapse
Affiliation(s)
- Neha Mehrotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Surender Kharbanda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| |
Collapse
|
32
|
Decorin expression is associated with predictive diffusion MR phenotypes of anti-VEGF efficacy in glioblastoma. Sci Rep 2020; 10:14819. [PMID: 32908231 PMCID: PMC7481206 DOI: 10.1038/s41598-020-71799-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Previous data suggest that apparent diffusion coefficient (ADC) imaging phenotypes predict survival response to anti-VEGF monotherapy in glioblastoma. However, the mechanism by which imaging may predict clinical response is unknown. We hypothesize that decorin (DCN), a proteoglycan implicated in the modulation of the extracellular microenvironment and sequestration of pro-angiogenic signaling, may connect ADC phenotypes to survival benefit to anti-VEGF therapy. Patients undergoing resection for glioblastoma as well as patients included in The Cancer Genome Atlas (TCGA) and IVY Glioblastoma Atlas Project (IVY GAP) databases had pre-operative imaging analyzed to calculate pre-operative ADCL values, the average ADC in the lower distribution using a double Gaussian mixed model. ADCL values were correlated to available RNA expression from these databases as well as from RNA sequencing from patient derived mouse orthotopic xenograft samples. Targeted biopsies were selected based on ADC values and prospectively collected during resection. Surgical specimens were used to evaluate for DCN RNA and protein expression by ADC value. The IVY Glioblastoma Atlas Project Database was used to evaluate DCN localization and relationship with VEGF pathway via in situ hybridization maps and RNA sequencing data. In a cohort of 35 patients with pre-operative ADC imaging and surgical specimens, DCN RNA expression levels were significantly larger in high ADCL tumors (41.6 vs. 1.5; P = 0.0081). In a cohort of 17 patients with prospectively targeted biopsies there was a positive linear correlation between ADCL levels and DCN protein expression between tumors (Pearson R2 = 0.3977; P = 0.0066) and when evaluating different targets within the same tumor (Pearson R2 = 0.3068; P = 0.0139). In situ hybridization data localized DCN expression to areas of microvascular proliferation and immunohistochemical studies localized DCN protein expression to the tunica adventitia of blood vessels within the tumor. DCN expression positively correlated with VEGFR1 & 2 expression and localized to similar areas of tumor. Increased ADCL on diffusion MR imaging is associated with high DCN expression as well as increased survival with anti-VEGF therapy in glioblastoma. DCN may play an important role linking the imaging features on diffusion MR and anti-VEGF treatment efficacy. DCN may serve as a target for further investigation and modulation of anti-angiogenic therapy in GBM.
Collapse
|
33
|
Vautrin-Glabik A, Devy J, Bour C, Baud S, Choulier L, Hoarau A, Dupont-Deshorgue A, Sellier C, Brassart B, Oudart JB, Ramont L, Monboisse JC, Brassart-Pasco S. Angiogenesis Inhibition by a Short 13 Amino Acid Peptide Sequence of Tetrastatin, the α4(IV) NC1 Domain of Collagen IV. Front Cell Dev Biol 2020; 8:775. [PMID: 32850867 PMCID: PMC7431705 DOI: 10.3389/fcell.2020.00775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/23/2020] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis is defined as the formation of new capillaries by sprouting from the pre-existing microvasculature. It occurs in physiological and pathological processes particularly in tumor growth and metastasis. α1, α2, α3, and α6 NC1 domains from type IV collagen were reported to inhibit tumor angiogenesis. We previously demonstrated that the α4 NC1 domain from type IV collagen, named Tetrastatin, inhibited tumor growth in a mouse melanoma model. The inhibitory activity was located in a 13 amino acid sequence named QS-13. In the present paper, we demonstrate that QS-13 decreases VEGF-induced-angiogenesis in vivo using the Matrigel plug model. Fluorescence molecular tomography allows the measurement of a 65% decrease in Matrigel plug angiogenesis following QS-13 administration. The results are confirmed by CD31 microvessel density analysis on Matrigel plug slices. QS-13 peptide decreases Human Umbilical Vein Endothelial Cells (HUVEC) migration and pseudotube formation in vitro. Relevant QS-13 conformations were obtained from molecular dynamics simulations and docking. A putative interaction of QS-13 with α5β1 integrin was investigated. The interaction was confirmed by affinity chromatography, solid phase assay, and surface plasmon resonance. QS-13 binding site on α5β1 integrin is located in close vicinity to the RGD binding site, as demonstrated by competition assays. Collectively, our results suggest that QS-13 exhibits a mighty anti-angiogenic activity that could be used in cancer treatment and other pathologies with excessive angiogenesis such as hemangioma, psoriasis or diabetes.
Collapse
Affiliation(s)
- Alexia Vautrin-Glabik
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Jérôme Devy
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Camille Bour
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Stéphanie Baud
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, URCA, Reims, France
| | - Laurence Choulier
- CNRS UMR 7021, Laboratoire de Bioimagerie et Pathologies, Université de Strasbourg, Illkirch, France
| | - Anthony Hoarau
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Aurélie Dupont-Deshorgue
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Christèle Sellier
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Bertrand Brassart
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Jean-Baptiste Oudart
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Laurent Ramont
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Jean Claude Monboisse
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France.,CHU Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Sylvie Brassart-Pasco
- Laboratoire de Biochimie, Université de Reims Champagne-Ardenne (URCA), Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| |
Collapse
|
34
|
Chakraborty P, Dash SP, Sarangi PP. The role of adhesion protein Fibulin7 in development and diseases. Mol Med 2020; 26:47. [PMID: 32429873 PMCID: PMC7238533 DOI: 10.1186/s10020-020-00169-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/22/2020] [Indexed: 01/03/2023] Open
Abstract
Fibulins are a family of secreted glycoproteins, which play an important role in regulating multiple cellular functions such as adhesion, growth, motility, and survival. Fibulin7 (Fbln7) is expressed in developing odontoblasts, in the giant trophoblast layer of the placenta, in the choroid of the eyes as well as in the cartilage. Since its discovery, reports from various research groups have improved our understanding about the roles and effects of Fbln7 and Fbln7 derived fragments and peptides under physiological and pathological conditions such as tooth development, angiogenesis, immunoregulation, cancer pathogenesis and very recently as a possible biomarker for glaucoma. This review will highlight the latest developments in our understanding of the functions, the proposed mechanism of actions, and Fbln7's possible implications in future research and as therapeutics for different diseases.
Collapse
Affiliation(s)
- Papiya Chakraborty
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Shiba Prasad Dash
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Pranita P Sarangi
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
35
|
Abstract
Peptides are one of the most important functional motifs for constructing smart drug delivery systems (DDSs). Functional peptides can be conjugated with drugs or carriers via covalent bonds, or assembled into DDSs via supramolecular forces, which enables the DDSs to acquire desired functions such as targeting and/or environmental responsiveness. In this mini review, we first introduce the different types of functional peptides that are commonly used for constructing DDSs, and we highlight representative strategies for designing smart DDSs by using functional peptides in the past few years. We also state the challenges of peptide-based DDSs and come up with prospects.
Collapse
Affiliation(s)
- Zheng Lian
- People's Public Security University of China, Beijing 100038, China
| | | |
Collapse
|
36
|
Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins (Basel) 2020; 12:toxins12040255. [PMID: 32326531 PMCID: PMC7232197 DOI: 10.3390/toxins12040255] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/25/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
Abstract For decades, natural products in general and snake venoms (SV) in particular have been a rich source of bioactive compounds for drug discovery, and they remain a promising substrate for therapeutic development. Currently, a handful of SV-based drugs for diagnosis and treatment of various cardiovascular disorders and blood abnormalities are on the market. Likewise, far more SV compounds and their mimetics are under investigation today for diverse therapeutic applications, including antibiotic-resistant bacteria and cancer. In this review, we analyze the state of the art regarding SV-derived compounds with therapeutic potential, focusing on the development of antimicrobial and anticancer drugs. Specifically, information about SV peptides experimentally validated or predicted to act as antimicrobial and anticancer peptides (AMPs and ACPs, respectively) has been collected and analyzed. Their principal activities both in vitro and in vivo, structures, mechanisms of action, and attempts at sequence optimization are discussed in order to highlight their potential as drug leads. Key Contribution This review describes the state of the art in snake venom-derived peptides and their therapeutic applications. This work reinforces the potential of snake venom components as therapeutic agents, particularly in the quest for new antimicrobial and anticancer drugs.
Collapse
|
37
|
Sohbati H, Alipour M, Hosseinkhani S, Balalaie S, Hamdan F. Design, Synthesis and Biological Evaluation of Triptorelin Analogs Containing Tetrazole Moiety. ChemistrySelect 2020. [DOI: 10.1002/slct.201903722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Hamidreza Sohbati
- Peptide Chemistry Research CenterK. N. Toosi University of Technology P. O. Box 15875–4416 Tehran Iran
| | - Mohsen Alipour
- Department of Advanced Medical Sciences & Technologies, School of MedicineJahrom, University of Medical Sciences, Jahrom Iran
| | - Saman Hosseinkhani
- Department of Nano Biotechnology, Faculty of Biological SciencesTarbiat Modares University, Tehran Iran
| | - Saeed Balalaie
- Peptide Chemistry Research CenterK. N. Toosi University of Technology P. O. Box 15875–4416 Tehran Iran
- e Medical Biology Research CenterKermanshah University of Medical Sciences, Kermanshah Iran
| | - Fatima Hamdan
- Peptide Chemistry Research CenterK. N. Toosi University of Technology P. O. Box 15875–4416 Tehran Iran
| |
Collapse
|
38
|
Ghadam M, Sardari S, Shokrgozar MA, Mahdavi MS. Design of Anti-Angiogenic Peptidomimetics and Evaluation their Biological Activity by In Vitro Assays. Avicenna J Med Biotechnol 2020; 12:91-98. [PMID: 32431793 PMCID: PMC7229457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND One of the important therapeutic approaches in cancer field is development of compounds which can block the initial tumor growth and the progression of tumor metastasis with no side effects. Thus, the recent study was carried out to design anti-VEGFR2-peptidomimetics as the most significant factor of angiogenesis process- and evaluate their biological activity by in vitro assays. METHODS We designed anti-VEGFR2 peptidomimetics with anti-angiogenic activity, including compound P (lactam derivative) and compound T (indole derivative) by using in silico methods. Then, the inhibitory activity on angiogenesis was evaluated by using angiogenesis specific assays such as Human Umbilical Vein Endothelial Cell (HUVEC) proliferation, tube formation in Matrigel, MTT and Real-Time PCR. IC50 values of the compounds were also determined by cytotoxicity plot in MTT assay. RESULTS Compounds P and T inhibited HUVEC cell proliferation and viability in a dose-dependent manner. The IC50 for compound T and compound P in HUVEC cell line were 113 and 115 μg/ml, respectively. Tube formation assay revealed that both compounds can inhibit angiogenesis effectively. The results of Real-Time PCR also showed these compounds are able to inhibit the expression of CD31 gene in HUVEC cell line. CONCLUSION Our study suggested that compounds P and T may act as therapeutic molecules, or lead compounds for development of angiogenesis inhibitors in VEGF-related diseases.
Collapse
Affiliation(s)
- Mona Ghadam
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Soroush Sardari
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran,Corresponding authors: Soroush Sardari, Ph.D., Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Ali Shokrgozar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran,Mohammad Ali Shokrgozar, Ph.D., National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran, Tel: +98 9122632484, E-mail: ;
| | - Mahdiyeh Sadat Mahdavi
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
39
|
Khodabakhsh F, Muyldermans S, Behdani M, Kazemi-Lomedasht F. Liposomal delivery of vascular endothelial growth factor/receptors and their inhibitors. J Drug Target 2019; 28:379-385. [DOI: 10.1080/1061186x.2019.1693578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mahdi Behdani
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
40
|
Gholibeikian M, Bamoniri A, HoushdarTehrani MH, Fatemeh Mirjalili BB, Bijanzadeh HR. Structure-activity relationship studies of Longicalcynin A analogues, as anticancer cyclopeptides. Chem Biol Interact 2019; 315:108902. [PMID: 31747558 DOI: 10.1016/j.cbi.2019.108902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/29/2019] [Accepted: 11/13/2019] [Indexed: 01/18/2023]
Abstract
Cancer has emerged as the main cause of the highest rate of mortality in the world. Drugs used in cancer, although, show some beneficial effects on cancerous organs, demonstrate side effects on other normal tissues. On the other hand, anticancer peptides, being effective on target tissues, should be safe and less harmful on healthy organs, since peptides have several advantages, i.e., high activity, specificity, affinity, being less immunogenic and not accumulate in the body. In the present work, analogues of Longicalcynin A, a naturally occurring anticancer cyclopeptide, were synthesized and evaluated their cytotoxicity in order to gain information from structure-activity relationships of the such cyclopeptides which may lead to find novel and safer anticancer peptide compound(s) to be used in clinic. Peptides were prepared by the solid-phase peptide synthesis method using trityl-resin. Peptide cyclization was performed in liquid phase. To study anticancer activity of the peptide analogues of Longicalycinin A, several methods including MTT, flow cytometry analysis and Lysosomal membrane integrity assay were employed using two cell lines HepG2 and HT-29. Fibroblast cells were used to control the safety of the synthesized cyclopeptides on normal cells. Two cyclopeptides 11 and 17 with the sequences of cyclo-(Thr-Val-Pro-Phe-Ala) and cyclo-(Phe-Ser-Pro-Phe-Ala), respectively were cytotoxic against the colon as well as hepatic cancer cells with safety profile against fibroblast cells, probably with the mechanism of apoptosis as lysosomal membrane integrity damaged. These cyclopeptides showed to be more favorable compounds better than Longicalycinin A and good candidates to develop cyclopeptides as anticancer agents.
Collapse
Affiliation(s)
| | - Abdolhamid Bamoniri
- Department of Organic Chemistry, Faculty of Chemistry, University of Kashan, Kashan, I.R., Iran.
| | - Mohammad Hassan HoushdarTehrani
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, I.R., Iran.
| | | | | |
Collapse
|
41
|
Rajabi M, Adeyeye M, Mousa SA. Peptide-Conjugated Nanoparticles as Targeted Anti-angiogenesis Therapeutic and Diagnostic in Cancer. Curr Med Chem 2019; 26:5664-5683. [DOI: 10.2174/0929867326666190620100800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 12/25/2022]
Abstract
:Targeting angiogenesis in the microenvironment of a tumor can enable suppression of tumor angiogenesis and delivery of anticancer drugs into the tumor. Anti-angiogenesis targeted delivery systems utilizing passive targeting such as Enhanced Permeability and Retention (EPR) and specific receptor-mediated targeting (active targeting) should result in tumor-specific targeting. One targeted anti-angiogenesis approach uses peptides conjugated to nanoparticles, which can be loaded with anticancer agents. Anti-angiogenesis agents can suppress tumor angiogenesis and thereby affect tumor growth progression (tumor growth arrest), which may be further reduced with the targetdelivered anticancer agent. This review provides an update of tumor vascular targeting for therapeutic and diagnostic applications, with conventional or long-circulating nanoparticles decorated with peptides that target neovascularization (anti-angiogenesis) in the tumor microenvironment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Mary Adeyeye
- Department of Chemistry, University of Albany, State University of New York, Albany, NY 12222, United States
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| |
Collapse
|
42
|
Conte C, Moret F, Esposito D, Dal Poggetto G, Avitabile C, Ungaro F, Romanelli A, Laurienzo P, Reddi E, Quaglia F. Biodegradable nanoparticles exposing a short anti-FLT1 peptide as antiangiogenic platform to complement docetaxel anticancer activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:876-886. [DOI: 10.1016/j.msec.2019.04.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/27/2019] [Accepted: 04/18/2019] [Indexed: 12/20/2022]
|
43
|
Laengsri V, Nantasenamat C, Schaduangrat N, Nuchnoi P, Prachayasittikul V, Shoombuatong W. TargetAntiAngio: A Sequence-Based Tool for the Prediction and Analysis of Anti-Angiogenic Peptides. Int J Mol Sci 2019; 20:E2950. [PMID: 31212918 PMCID: PMC6628072 DOI: 10.3390/ijms20122950] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 11/21/2022] Open
Abstract
Cancer remains one of the major causes of death worldwide. Angiogenesis is crucial for the pathogenesis of various human diseases, especially solid tumors. The discovery of anti-angiogenic peptides is a promising therapeutic route for cancer treatment. Thus, reliably identifying anti-angiogenic peptides is extremely important for understanding their biophysical and biochemical properties that serve as the basis for the discovery of new anti-cancer drugs. This study aims to develop an efficient and interpretable computational model called TargetAntiAngio for predicting and characterizing anti-angiogenic peptides. TargetAntiAngio was developed using the random forest classifier in conjunction with various classes of peptide features. It was observed via an independent validation test that TargetAntiAngio can identify anti-angiogenic peptides with an average accuracy of 77.50% on an objective benchmark dataset. Comparisons demonstrated that TargetAntiAngio is superior to other existing methods. In addition, results revealed the following important characteristics of anti-angiogenic peptides: (i) disulfide bond forming Cys residues play an important role for inhibiting blood vessel proliferation; (ii) Cys located at the C-terminal domain can decrease endothelial formatting activity and suppress tumor growth; and (iii) Cyclic disulfide-rich peptides contribute to the inhibition of angiogenesis and cell migration, selectivity and stability. Finally, for the convenience of experimental scientists, the TargetAntiAngio web server was established and made freely available online.
Collapse
Affiliation(s)
- Vishuda Laengsri
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Pornlada Nuchnoi
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
44
|
|
45
|
A cyclic peptide reproducing the α1 helix of VEGF-B binds to VEGFR-1 and VEGFR-2 and inhibits angiogenesis and tumor growth. Biochem J 2019; 476:645-663. [DOI: 10.1042/bcj20180823] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 11/17/2022]
Abstract
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are pivotal regulators of angiogenesis. The VEGF–VEGFR system is therefore an important target of anti-angiogenesis therapy. Based on the X-ray structure of VEGF-B/VEGFR-1 D2, we designed a cyclic peptide (known as VGB1) reproducing the α1 helix and its adjacent region to interfere with signaling through VEGFR-1. Unexpectedly, VGB1 bound VEGFR-2 in addition to VEGFR-1, leading to inhibition of VEGF-stimulated proliferation of human umbilical vein endothelial cells and 4T1 murine mammary carcinoma cells, which express VGEFR-1 and VEGFR-2, and U87 glioblastoma cells that mostly express VEGFR-2. VGB1 inhibited different aspects of angiogenesis, including proliferation, migration and tube formation of endothelial cells stimulated by VEGF-A through suppression of extracellular signal-regulated kinase 1/2 and AKT (Protein Kinase B) phosphorylation. In a murine 4T1 mammary carcinoma model, VGB1 caused regression of tumors without causing weight loss in association with impaired cell proliferation (decreased Ki67 expression) and angiogenesis (decreased CD31 and CD34 expression), and apoptosis induction (increased TUNEL staining and p53 expression, and decreased Bcl-2 expression). According to far-UV circular dichroism (CD) and molecular dynamic simulation data, VGB1 can adopt a helical structure. These results, for the first time, demonstrate that α1 helix region of VEGF-B recognizes both VEGFR-1 and VEGFR-2.
Collapse
|
46
|
Blanco JL, Porto-Pazos AB, Pazos A, Fernandez-Lozano C. Prediction of high anti-angiogenic activity peptides in silico using a generalized linear model and feature selection. Sci Rep 2018; 8:15688. [PMID: 30356060 PMCID: PMC6200741 DOI: 10.1038/s41598-018-33911-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 10/06/2018] [Indexed: 12/22/2022] Open
Abstract
Screening and in silico modeling are critical activities for the reduction of experimental costs. They also speed up research notably and strengthen the theoretical framework, thus allowing researchers to numerically quantify the importance of a particular subset of information. For example, in fields such as cancer and other highly prevalent diseases, having a reliable prediction method is crucial. The objective of this paper is to classify peptide sequences according to their anti-angiogenic activity to understand the underlying principles via machine learning. First, the peptide sequences were converted into three types of numerical molecular descriptors based on the amino acid composition. We performed different experiments with the descriptors and merged them to obtain baseline results for the performance of the models, particularly of each molecular descriptor subset. A feature selection process was applied to reduce the dimensionality of the problem and remove noisy features – which are highly present in biological problems. After a robust machine learning experimental design under equal conditions (nested resampling, cross-validation, hyperparameter tuning and different runs), we statistically and significantly outperformed the best previously published anti-angiogenic model with a generalized linear model via coordinate descent (glmnet), achieving a mean AUC value greater than 0.96 and with an accuracy of 0.86 with 200 molecular descriptors, mixed from the three groups. A final analysis with the top-40 discriminative anti-angiogenic activity peptides is presented along with a discussion of the feature selection process and the individual importance of each molecular descriptors According to our findings, anti-angiogenic activity peptides are strongly associated with amino acid sequences SP, LSL, PF, DIT, PC, GH, RQ, QD, TC, SC, AS, CLD, ST, MF, GRE, IQ, CQ and HG.
Collapse
Affiliation(s)
- Jose Liñares Blanco
- Department of Computer Science, Faculty of Computer Science, University of A Coruña, A Coruña, 15071, Spain
| | - Ana B Porto-Pazos
- Department of Computer Science, Faculty of Computer Science, University of A Coruña, A Coruña, 15071, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Alejandro Pazos
- Department of Computer Science, Faculty of Computer Science, University of A Coruña, A Coruña, 15071, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Carlos Fernandez-Lozano
- Department of Computer Science, Faculty of Computer Science, University of A Coruña, A Coruña, 15071, Spain. .,Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain.
| |
Collapse
|
47
|
Using a Classifier Fusion Strategy to Identify Anti-angiogenic Peptides. Sci Rep 2018; 8:14062. [PMID: 30218091 PMCID: PMC6138733 DOI: 10.1038/s41598-018-32443-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022] Open
Abstract
Anti-angiogenic peptides perform distinct physiological functions and potential therapies for angiogenesis-related diseases. Accurate identification of anti-angiogenic peptides may provide significant clues to understand the essential angiogenic homeostasis within tissues and develop antineoplastic therapies. In this study, an ensemble predictor is proposed for anti-angiogenic peptide prediction by fusing an individual classifier with the best sensitivity and another individual one with the best specificity. We investigate predictive capabilities of various feature spaces with respect to the corresponding optimal individual classifiers and ensemble classifiers. The accuracy and Matthew’s Correlation Coefficient (MCC) of the ensemble classifier trained by Bi-profile Bayes (BpB) features are 0.822 and 0.649, respectively, which represents the highest prediction results among the investigated prediction models. Discriminative features are obtained from BpB using the Relief algorithm followed by the Incremental Feature Selection (IFS) method. The sensitivity, specificity, accuracy, and MCC of the ensemble classifier trained by the discriminative features reach up to 0.776, 0.888, 0.832, and 0.668, respectively. Experimental results indicate that the proposed method is far superior to the previous study for anti-angiogenic peptide prediction.
Collapse
|
48
|
Nguyen PK, Sarkar B, Siddiqui Z, McGowan M, Iglesias-Montoro P, Rachapudi S, Kim S, Gao W, Lee EJ, Kumar VA. Self-Assembly of an Antiangiogenic Nanofibrous Peptide Hydrogel. ACS APPLIED BIO MATERIALS 2018; 1:865-870. [DOI: 10.1021/acsabm.8b00283] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Vivek A. Kumar
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, New Jersey 07103, United States
| |
Collapse
|
49
|
Bazzazi H, Zhang Y, Jafarnejad M, Popel AS. Computational modeling of synergistic interaction between αVβ3 integrin and VEGFR2 in endothelial cells: Implications for the mechanism of action of angiogenesis-modulating integrin-binding peptides. J Theor Biol 2018; 455:212-221. [PMID: 30036530 DOI: 10.1016/j.jtbi.2018.06.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/13/2018] [Accepted: 06/25/2018] [Indexed: 01/06/2023]
Abstract
Cooperation between VEGFR2 and integrin αVβ3 is critical for neovascularization in wound healing, cardiovascular ischemic diseases, ocular diseases, and tumor angiogenesis. In the present study, we developed a rule-based computational model to investigate the potential mechanism by which the Src-induced integrin association with VEGFR2 enhances VEGFR2 activation. Simulations demonstrated that the main function of integrin is to reduce the degradation of VEGFR2 and hence stabilize the activation signal. In addition, receptor synthesis rate and recruitment from internal compartment were found to be sensitive determinants of the activation state of VEGFR2. The model was then applied to simulate the effect of integrin-binding peptides such as tumstatin and cilengitide on VEGFR2 signaling. Further, computational modeling proposed potential molecular mechanisms for the angiogenesis-modulating activity of other integrin-binding peptides. The model highlights the complexity of the crosstalk between αVβ3 integrin and VEGFR2 and the necessity of utilizing models to elucidate potential mechanisms in angiogenesis-modulating peptide therapy.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States.
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
50
|
Gaustad JV, Simonsen TG, Andersen LMK, Rofstad EK. Properdistatin inhibits angiogenesis and improves vascular function in human melanoma xenografts with low thrombospondin-1 expression. Oncotarget 2018; 7:76806-76815. [PMID: 27756886 PMCID: PMC5363551 DOI: 10.18632/oncotarget.12695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022] Open
Abstract
In this study, the effect of properdistatin, a novel peptide derived from the thrombospondin 1 (TSP-1) domain of properdin, was investigated in three melanoma xenograft models with different TSP-1 expression. The tumors were grown in dorsal window chambers and were treated with 80 mg/kg/day properdistatin or vehicle. Morphological parameters of the tumor vasculature were assessed from high resolution transillumination images. Blood supply time (i.e., the time required for arterial blood to flow from a supplying artery to downstream microvessels) and plasma velocities were assessed from first-pass imaging movies recorded after a bolus of fluorescence-labeled dextran had been administered intravenously. Gene and protein expression of TSP-1 were assessed with quantitative PCR and immunohistochemistry, respectively. Properdistatin treatment inhibited angiogenesis in low TSP-1 expressing tumors but did not alter the vasculature in high TSP-1 expressing tumors. In low TSP-1 expressing tumors, properdistatin selectively removed small-diameter capillaries, but did not change the morphology of tumor arterioles or tumor venules. Properdistatin also reduced blood supply times and increased plasma velocities, implying that the treatment reduced the geometric resistance to blood flow and improved vascular function.
Collapse
Affiliation(s)
- Jon-Vidar Gaustad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Trude G Simonsen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Lise Mari K Andersen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|