1
|
Qureshi S, Ahmed N, Rehman HM, Amirzada MI, Saleem F, Waheed K, Chaudhry A, Kafait I, Akram M, Bashir H. Investigation of therapeutic potential of the Il24-p20 fusion protein against breast cancer: an in-silico approach. In Silico Pharmacol 2024; 12:84. [PMID: 39301086 PMCID: PMC11408464 DOI: 10.1007/s40203-024-00252-x] [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: 06/20/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
Targeted delivery of therapeutic anticancer chimeric molecules enhances drug efficacy. Numerous studies have focused on developing novel treatments by employing cytokines, particularly interleukins, to inhibit the growth of cancer cells. In the present study, we fused interleukin 24 with the tumor-targeting peptide P20 through a rigid linker to selectively target cancer cells. The secondary structure, tertiary structure, and physicochemical characteristics of the constructed chimeric IL-24-P20 protein were predicted by using bioinformatics tools. In-silico analysis revealed that the fusion construct has a basic nature with 175 amino acids and a molecular weight of 20 kDa. By using the Rampage and ERRAT2 servers, the validity and quality of the fusion protein were evaluated. The results indicated that 93% of the chimeric proteins contained 90.1% of the residues in the favoured region, resulting in a reliable structure. Finally, docking and simulation studies were conducted via ClusPro and Desmond Schrödinger, respectively. Our results indicate that the constructed fusion protein exhibits excellent quality, interaction capabilities, validity, and stability. These findings suggest that the fusion protein is a promising candidate for targeted cancer therapy.
Collapse
Affiliation(s)
- Shahnila Qureshi
- Centre for Applied Molecular Biology, University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
| | - Nadeem Ahmed
- Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
| | - Hafiz Muhammad Rehman
- Centre for Applied Molecular Biology, University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
- University Institute of Medical Laboratory Technology, Faculty of Allied Health Science, University of Lahore, Lahore, 54590 Pakistan
| | | | - Fiza Saleem
- Centre for Applied Molecular Biology, University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
- University Institute of Medical Laboratory Technology, Faculty of Allied Health Science, University of Lahore, Lahore, 54590 Pakistan
| | - Kainat Waheed
- Centre for Applied Molecular Biology, University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
- University Institute of Medical Laboratory Technology, Faculty of Allied Health Science, University of Lahore, Lahore, 54590 Pakistan
| | - Afeefa Chaudhry
- Department of Biology, Lahore Garrison University, Avenue 4, sector phase 6 DHA, Lahore, Pakistan
| | - Iram Kafait
- Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria
| | - Muhammad Akram
- Centre for Applied Molecular Biology, University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
| | - Hamid Bashir
- Centre for Applied Molecular Biology, University of the Punjab, 87-West canal, Bank Road, Lahore, 53700 Pakistan
| |
Collapse
|
2
|
Feng C, Yu A, Wang Z, Wang K, Chen J, Wu Y, Deng T, Chen H, Hou Y, Ma S, Dai X, Huang L. A novel PDPN antagonist peptide CY12-RP2 inhibits melanoma growth via Wnt/β-catenin and modulates the immune cells. J Exp Clin Cancer Res 2024; 43:9. [PMID: 38167452 PMCID: PMC10759609 DOI: 10.1186/s13046-023-02910-y] [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: 09/05/2023] [Accepted: 11/17/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Podoplanin (PDPN) is a highly conserved, mucin-type protein specific to the lymphatic system. Overexpression of PDPN is associated with the progression of various solid tumors, and plays an important roles in the tumor microenvironment by regulating the immune system. However, the role of PDPN-mediated signal activation in the progression of melanoma is still unknown. METHODS PDPN expression was first analyzed in 112 human melanoma tissue microarrays and melanoma cell lines. Functional experiments including proliferation, clone formation, migration, and metastasis were utilized to identify the suppressive effects of PDPN. The Ph.D.TM-12 Phage Display Peptide Library was used to obtain a PDPN antagonist peptide, named CY12-RP2. The immunofluorescence, SPR assay, and flow cytometry were used to identify the binding specificity of CY12-RP2 with PDPN in melanoma cells. Functional and mechanistic assays in vivo and in vitro were performed for discriminating the antitumor and immune activation effects of CY12-RP2. RESULTS PDPN was overexpressed in melanoma tissue and cells, and inhibited melanoma cells proliferation, migration, and metastasis by blocking the EMT and Wnt/β-catenin pathway. PDPN antagonistic peptide, CY12-RP2, could specifically bind with PDPN, suppressing melanoma various functions inducing apoptosis in both melanoma cells and 3D spheroids. CY12-RP2 also enhanced the anti-tumor capacity of PBMC, and inhibited melanoma cells growth both in xenografts and allogeneic mice model. Moreover, CY12-RP2 could inhibit melanoma lung metastasis, and abrogated the immunosuppressive effects of PDPN by increasing the proportion of CD3 + CD4 + T cells, CD3 + CD8 + T cells, CD49b + Granzyme B + NK cells, and CD11b + CD86 + M1-like macrophages and the levels of IL-1β, TNF-α, and IFN-γ. CONCLUSIONS This study has demonstrated the important role of PDPN in the progression of melanoma and formation of immunosuppressive environment, and provided a potential approach of treating melanoma using the novel CY12-RP2 peptide. In melanoma, PDPN is overexpressed in the cancer cells, and promotes melanoma cells growth and metastasis through activating the Wnt/β-catenin pathway. Treatment with the PDPN antagonistic peptide CY12-RP2 could not only inhibit the melanoma growth and metastasis both in vitro and in vivo through Wnt/β-catenin pathway blockade, but also abrogate the immunosuppressive effects of PDPN through modulating immune cells.
Collapse
Affiliation(s)
- Chunyan Feng
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Albert Yu
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Zhongfu Wang
- Department of Interventional Radiology, Shenzhen People's Hospital, 1017 Dongmen North Road, Shenzhen, 518020, NoGuangdong, China
| | - Kun Wang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Jiawei Chen
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Yaojiong Wu
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Ting Deng
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Huaqing Chen
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Yibo Hou
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China
| | - Xiaoyong Dai
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China.
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Laiqiang Huang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China.
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
3
|
Rodrigues EG, Dobroff AS, Arruda DC, Tada DB, Paschoalin T, Polonelli L. A limitless Brazilian scientist: Professor Travassos and his contribution to cancer biology. Braz J Microbiol 2023; 54:2551-2560. [PMID: 37589929 PMCID: PMC10689629 DOI: 10.1007/s42770-023-01085-0] [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: 05/04/2023] [Accepted: 07/24/2023] [Indexed: 08/18/2023] Open
Abstract
Luiz Rodolpho Travassos, a Brazilian scientist recognized in several areas of research, began his studies in the field of oncology in the late 1970s when he took a sabbatical at the Memorial Sloan Kettering Cancer Center, NY, USA. At that time, the discovery and characterization of human melanoma glycoprotein antigens yielded important publications. This experience allowed 16 years later, and Dr. Travassos founded UNONEX, significantly contributing with discoveries in the area of oncology and training of researchers. This review will address all the contributions of team of researchers who, together with Dr. Travassos, collaborated with investigations into molecules and processes that lead to the development of melanoma.
Collapse
Affiliation(s)
- Elaine G Rodrigues
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil.
| | - Andrey S Dobroff
- University of New Mexico Comprehensive Cancer Center (UNMCCC), Albuquerque, USA
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico (UNM) School of Medicine, Albuquerque, USA
| | - Denise C Arruda
- Integrated Group of Biotechnology, University of Mogi das Cruzes, UMC, Mogi das Cruzes, SP, Brazil
| | - Dayane B Tada
- Laboratory of Nanomaterials and Nanotoxicology, Institute of Science and Technology, Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Thaysa Paschoalin
- Department of Biophysics, Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil.
| | - Luciano Polonelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
4
|
Koskela SA, Figueiredo CR. From antimicrobial to anticancer: the pioneering works of Prof. Luiz Rodolpho Travassos on bioactive peptides. Braz J Microbiol 2023; 54:2561-2570. [PMID: 37725261 PMCID: PMC10689714 DOI: 10.1007/s42770-023-01118-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Prof. Luiz Rodolpho Travassos, a distinguished Brazilian scientist, was instrumental in fostering an interdisciplinary research approach that seamlessly combined microbiology and oncology. This work has opened new pathways into the understanding of tumorigenesis and aided in the development of innovative therapeutic tools. One significant area of his work has been the exploration of bioactive peptides, many of which were first identified for their antimicrobial properties. These peptides demonstrate promise as potential cancer therapeutics due to their selectivity, cost-effectiveness, ease of synthesis, low antigenicity, and excellent tissue penetration. Prof. Travassos' pioneering work uncovered on the potential of peptides derived from microbiological sources, such as those obtained using phage display techniques. More importantly, in international cooperation, peptides derived from complementarity-determining regions (CDRs) that showed antimicrobial activity against Candida albicans further showed to be promising tools with cytotoxic properties against cancer cells. Similarly, peptides derived from natural sources, such as the gomesin peptide, not only had shown antimicrobial properties but could treat cutaneous melanoma in experimental models. These therapeutic tools allowed Prof. Travassos and his group to navigate the intricate landscape of factors and pathways that drive cancer development, including persistent proliferative signaling, evasion of tumor suppressor genes, inhibition of programmed cell death, and cellular immortality. This review examines the mechanisms of action of these peptides, aligning them with the universally recognized hallmarks of cancer, and evaluates their potential as drug candidates. It highlights the crucial need for more selective, microbiology-inspired anti-cancer strategies that spare healthy cells, a challenge that current therapies often struggle to address. By offering a comprehensive assessment of Prof. Travassos' innovative contributions and a detailed discussion on the increasing importance of microbiology-derived peptides, this review presents an informed and robust perspective on the possible future direction of cancer therapy.
Collapse
Affiliation(s)
- Saara A Koskela
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Carlos R Figueiredo
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
| |
Collapse
|
5
|
Ross A, Sauce-Guevara MA, Alarcon EI, Mendez-Rojas MA. Peptide Biomaterials for Tissue Regeneration. Front Bioeng Biotechnol 2022; 10:893936. [PMID: 35992354 PMCID: PMC9388858 DOI: 10.3389/fbioe.2022.893936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
Expanding the toolbox of therapeutic materials for soft tissue and organ repair has become a critical component of tissue engineering. While animal- and plant-derived proteins are the foundation for developing biomimetic tissue constructs, using peptides as either constituents or frameworks for the materials has gained increasing momentum in recent years. This mini review discusses recent advances in peptide-based biomaterials' design and application. We also discuss some of the future challenges posed and opportunities opened by peptide-based structures in the field of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Alex Ross
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Mildred A. Sauce-Guevara
- Department of Chemical and Biological Sciences, Universidad de Las Américas Puebla, Puebla, Mexico
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Miguel A. Mendez-Rojas
- Department of Chemical and Biological Sciences, Universidad de Las Américas Puebla, Puebla, Mexico
| |
Collapse
|
6
|
Selection and identification of a specific peptide binding to ovarian cancer cells from a phage-displayed peptide library. Biotechnol Lett 2022; 44:951-960. [PMID: 35771408 DOI: 10.1007/s10529-022-03263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/03/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVES Ovarian cancer is one of the most fatal gynecological malignancies. It is emergently needed to select a novel molecular fragment as a targeting element for the future development of molecular imaging diagnosis and targeting chemotherapy to ovarian cancer. RESULTS After five rounds of biopanning, a total of 44 positive phage clones were selected from final phage displayed peptide library. Nine consensus sequences were found based on the assay of sequencing results, then one clone of each consensus group was characterized and identified further by immunofluorescence assay. The result showed the phage clone R20 presents best targeting capacity. Then we synthesized peptide (OSP2) clone R20 displayed, it was characterized with high specificity and sensitivity binding to human ovarian cancer by a tissue chip assay. The target of OSP2 was predicted and docked as human carbonic anhydrase XII (CA12), an important protein usually deregulated in cancer. CONCLUSIONS Taken together, OSP2 and its target indicate a novel investigation way in future to develop novel agent or drug delivery formulation for molecular imaging diagnosis and targeting chemotherapy of ovarian cancer.
Collapse
|
7
|
Rafiq S, Gulzar N, Sameen A, Huma N, Hayat I, Ijaz R. Functional role of bioactive peptides with special reference to cheeses. INT J DAIRY TECHNOL 2020. [DOI: 10.1111/1471-0307.12732] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Saima Rafiq
- Department of Food Science and Technology Faculty of Agriculture University of Poonch Rawalakot 12350 Azad kashmirPakistan
| | - Nabila Gulzar
- Department of Dairy Technology University of Veterinary and Animal Sciences Lahore Lahore55300Pakistan
| | - Aysha Sameen
- National Institute of Food Science and Technology University of Agriculture Faisalabad38040Pakistan
| | - Nuzhat Huma
- National Institute of Food Science and Technology University of Agriculture Faisalabad38040Pakistan
| | - Imran Hayat
- Department of Food Science and Technology Faculty of Agriculture University of Poonch Rawalakot 12350 Azad kashmirPakistan
| | - Raina Ijaz
- Department of Horticulture Faculty of Agriculture University of Poonch Rawalakot 12350 Azad Kashmir Pakistan
| |
Collapse
|
8
|
Xu H, Cao B, Li Y, Mao C. Phage nanofibers in nanomedicine: Biopanning for early diagnosis, targeted therapy, and proteomics analysis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1623. [PMID: 32147974 DOI: 10.1002/wnan.1623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/02/2020] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Display of a peptide or protein of interest on the filamentous phage (also known as bacteriophage), a biological nanofiber, has opened a new route for disease diagnosis and therapy as well as proteomics. Earlier phage display was widely used in protein-protein or antigen-antibody studies. In recent years, its application in nanomedicine is becoming increasingly popular and encouraging. We aim to review the current status in this research direction. For better understanding, we start with a brief introduction of basic biology and structure of the filamentous phage. We present the principle of phage display and library construction method on the basis of the filamentous phage. We summarize the use of the phage displayed peptide library for selecting peptides with high affinity against cells or tissues. We then review the recent applications of the selected cell or tissue targeting peptides in developing new targeting probes and therapeutics to advance the early diagnosis and targeted therapy of different diseases in nanomedicine. We also discuss the integration of antibody phage display and modern proteomics in discovering new biomarkers or target proteins for disease diagnosis and therapy. Finally, we propose an outlook for further advancing the potential impact of phage display on future nanomedicine. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Hong Xu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
9
|
Rafiq S, Gulzar N, Huma N, Hussain I, Murtaza MS. Evaluation of anti‐proliferative activity of Cheddar cheeses using colon adenocarcinoma (HCT‐116) cell line. INT J DAIRY TECHNOL 2019. [DOI: 10.1111/1471-0307.12665] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Saima Rafiq
- Department of Food Science and Technology University of the Poonch Rawalakot 12350 Azad Kashmir Pakistan
| | - Nabila Gulzar
- National Institute of Food Science and Technology University of Agriculture Faisalabad 38040Pakistan
| | - Nuzhat Huma
- National Institute of Food Science and Technology University of Agriculture Faisalabad 38040Pakistan
| | - Imtiaz Hussain
- Department of Food Science and Technology University of the Poonch Rawalakot 12350 Azad Kashmir Pakistan
| | - Mian Shamas Murtaza
- Department of Food Science and Technology Muhammad Nawaz Shareef University of Agriculture Multan Pakistan
| |
Collapse
|
10
|
Mahzoon S, Detamore MS. Chondroinductive Peptides: Drawing Inspirations from Cell–Matrix Interactions. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:249-257. [DOI: 10.1089/ten.teb.2018.0003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Salma Mahzoon
- School of Aerospace and Mechanical Engineering, University of Oklahoma, Norman, Oklahoma
| | - Michael S. Detamore
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma
| |
Collapse
|
11
|
Wang Y, Gao S, Lv J, Lin Y, Zhou L, Han L. Phage Display Technology and its Applications in Cancer Immunotherapy. Anticancer Agents Med Chem 2019; 19:229-235. [PMID: 30370861 DOI: 10.2174/1871520618666181029140814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 06/01/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
Abstract
Background:Phage display is an effective technology for generation and selection targeting protein for a variety of purpose, which is based on a direct linkage between the displayed protein and the DNA sequence encoding it and utilized in selecting peptides, improving peptides affinity and indicating protein-protein interactions. Phage particles displaying peptide have the potential to apply in the identification of cell-specific targeting molecules, identification of cancer cell surface biomarkers, identification anti-cancer peptide, and the design of peptide-based anticancer therapy.Method/Results:Literature searches, reviews and assessments about Phage were performed in this review from PubMed and Medline databases.Conclusion:The phage display technology is an inexpensive method for expressing exogenous peptides, generating unique peptides that bind any given target and investigating protein-protein interactions. Due to the powerful ability to insert exogenous gene and display exogenous peptides on the surface, phages may represent a powerful peptide delivery system that can be utilized to develop rapid, efficient, safe and inexpensive cancer therapy methods.
Collapse
Affiliation(s)
- Yicun Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Third Hospital of Jilin University, Changchun, China
| | - Jiayin Lv
- Third Hospital of Jilin University, Changchun, China
| | - Yang Lin
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Li Zhou
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Liying Han
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Rafiq S, Huma N, Gulzar N, Murtaza MA, Hussain I. Effect of Cheddar cheese peptide extracts on growth inhibition, cell cycle arrest and apoptosis induction in human lung cancer (H-1299) cell line. INT J DAIRY TECHNOL 2018. [DOI: 10.1111/1471-0307.12533] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Saima Rafiq
- Department of Food Science and Technology; Faculty of Agriculture; The University of Poonch Rawalakot; Azad Kashmir 12350 Pakistan
| | - Nuzhat Huma
- National Institute of Food Science and Technology; University of Agriculture; Faisalabad 38040 Pakistan
| | - Nabila Gulzar
- National Institute of Food Science and Technology; University of Agriculture; Faisalabad 38040 Pakistan
| | - Mian Anjum Murtaza
- Institute of Food Science and Nutrition; University of Sargodha; Sargodha 40100 Pakistan
| | - Imtiaz Hussain
- Department of Food Science and Technology; Faculty of Agriculture; The University of Poonch Rawalakot; Azad Kashmir 12350 Pakistan
| |
Collapse
|
13
|
Lei Z, Chai N, Tian M, Zhang Y, Wang G, Liu J, Tian Z, Yi X, Chen D, Li X, Yu P, Hu H, Xu B, Jian C, Bian Z, Guo H, Wang J, Peng S, Nie Y, Huang N, Hu S, Wu K. Novel peptide GX1 inhibits angiogenesis by specifically binding to transglutaminase-2 in the tumorous endothelial cells of gastric cancer. Cell Death Dis 2018; 9:579. [PMID: 29785022 PMCID: PMC5962530 DOI: 10.1038/s41419-018-0594-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/21/2018] [Accepted: 04/12/2018] [Indexed: 12/28/2022]
Abstract
The clinical application of GX1, an optimal gastric cancer (GC) targeting peptide, is greatly limited because its receptor in the GC vasculature is unknown. In this study, we screened the candidate receptor of GX1, transglutaminase-2(TGM2), by co-immunoprecipitation (co-IP) combined with mass spectrometry. We found that TGM2 was up-regulated in GC vascular endothelial cells and that GX1 receptor expression was suppressed correspondingly after TGM2 downregulation. A highly consistent co-localization of GX1 receptor and TGM2 was detected at both the cellular and tissue levels. High TGM2 expression was evident in GC tissues from patients with poor prognosis. After TGM2 downregulation, the GX1-mediated inhibition of proliferation and migration and the induction of the apoptosis of GC vascular endothelial cells were weakened or even reversed. Finally, we observed that GX1 could inhibit the GTP-binding activity of TGM2 by reducing its intracellular distribution and downregulating its downstream molecular targets (nuclear factor-kappa B, NF-κB; hypoxia-inducible factor 1-α, HIF1α) in GC vascular endothelial cells. Our study confirms that peptide GX1 can inhibit angiogenesis by directly binding to TGM2, subsequently reducing the GTP-binding activity of TGM2 and thereby suppressing its downstream pathway(NF-κB/HIF1α). Our conclusions suggest that GX1/TGM2 may provide a new target for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Zhijie Lei
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Na Chai
- Department of Radiology, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Miaomiao Tian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Ying Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Guodong Wang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Jian Liu
- Department of Radiology, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Zuhong Tian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Xiaofang Yi
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Di Chen
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Xiaowei Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Pengfei Yu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Hao Hu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Bing Xu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Chao Jian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Zhenyuan Bian
- Department of Hepatobiliary Surgery, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Hao Guo
- Department of Neurosurgery, Tangdu Hospital of Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China
| | - Jinpeng Wang
- Department of Orthopedics, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Shiming Peng
- National Institute of Biological Sciences, Beijing, 102206, People's Republic of China
| | - Yongzhan Nie
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Niu Huang
- National Institute of Biological Sciences, Beijing, 102206, People's Republic of China.
| | - Sijun Hu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| | - Kaichun Wu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| |
Collapse
|
14
|
Fernandes TB, Cunha MR, Sakata RP, Candido TM, Baby AR, Tavares MT, Barbosa EG, Almeida WP, Parise-Filho R. Synthesis, Molecular Modeling, and Evaluation of Novel Sulfonylhydrazones as Acetylcholinesterase Inhibitors for Alzheimer's Disease. Arch Pharm (Weinheim) 2017; 350. [PMID: 28940630 DOI: 10.1002/ardp.201700163] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and related to the degeneration of hippocampal cholinergic neurons, which dramatically affects cognitive ability. Acetylcholinesterase (AChE) inhibitors are employed as drugs for AD therapy. Three series of sulfonylhydrazone compounds were designed, and their ability to inhibit AChE was evaluated. Fifteen compounds were synthesized and twelve of them had IC50 values of 0.64-51.09 μM. The preliminary structure-activity relationships indicated that the methylcatechol moiety and arylsulfonyl substituents generated better compounds than both the benzodioxole and alkylsulfonyl chains. Molecular dynamics studies of compound 6d showed that the interaction with the peripheral binding site of AChE was similar to donepezil, which may explain its low IC50 (0.64 μM). Furthermore, the drug-likeness of 6d suggests that the compound may have appropriate oral absorption and brain penetration. Compound 6d also presented antiradical activity and was not cytotoxic to LL24 cells, suggesting that this compound might be considered safe. Our findings indicate that arylsulfonylhydrazones may be a promising scaffold for the design of new drug candidates for the treatment of AD.
Collapse
Affiliation(s)
- Thais B Fernandes
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Micael R Cunha
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Renata P Sakata
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Campinas, Campinas, Brazil
| | - Thalita M Candido
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - André R Baby
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Maurício T Tavares
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Euzébio G Barbosa
- Health Sciences Centre, Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Wanda P Almeida
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of Campinas, Campinas, Brazil
| | - Roberto Parise-Filho
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Arruda DC, de Oliveira TD, Cursino PHF, Maia VSC, Berzaghi R, Travassos LR, Tada DB. Inhibition of melanoma metastasis by dual-peptide PLGA NPS. Biopolymers 2017; 108. [DOI: 10.1002/bip.23029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Denise Costa Arruda
- Integrated Group of Biotechnology, University of Mogi das Cruzes, UMC; Mogi das Cruzes SP Brazil
| | | | | | | | - Rodrigo Berzaghi
- Experimental Oncology Unit (UNONEX), Universidade Federal de São Paulo (UNIFESP); São Paulo SP 04023-062 Brazil
| | - Luiz R. Travassos
- Experimental Oncology Unit (UNONEX), Universidade Federal de São Paulo (UNIFESP); São Paulo SP 04023-062 Brazil
| | - Dayane Batista Tada
- Institute of Science and Technology, Federal University of São Paulo; São José dos Campos SP Brazil
| |
Collapse
|
16
|
David A. Peptide ligand-modified nanomedicines for targeting cells at the tumor microenvironment. Adv Drug Deliv Rev 2017; 119:120-142. [PMID: 28506743 DOI: 10.1016/j.addr.2017.05.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/17/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Since their initial discovery more than 30years ago, tumor-homing peptides have become an increasingly useful tool for targeted delivery of therapeutic and diagnostic agents into tumors. Today, it is well accepted that cells at the tumor microenvironment (TME) contribute in many ways to cancer development and progression. Tumor-homing peptide-decorated nanomedicines can interact specifically with surface receptors expressed on cells in the TME, improve cellular uptake of nanomedicines by target cells, and impair tumor growth and progression. Moreover, peptide ligand-modified nanomedicines can potentially accumulate in the target tissue at higher concentrations than would small conjugates, thus increasing overall target tissue exposure to the therapeutic agent, enhance therapeutic efficacy and reduce side effects. This review describes the most studied peptide ligands aimed at targeting cells in the TME, discusses major obstacles and principles in the design of ligands for drug targeting and provides an overview of homing peptides in ligand-targeted nanomedicines that are currently in development for cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, and the Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
17
|
Liu R, Li X, Xiao W, Lam KS. Tumor-targeting peptides from combinatorial libraries. Adv Drug Deliv Rev 2017; 110-111:13-37. [PMID: 27210583 DOI: 10.1016/j.addr.2016.05.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major and leading causes of death worldwide. Two of the greatest challenges in fighting cancer are early detection and effective treatments with no or minimum side effects. Widespread use of targeted therapies and molecular imaging in clinics requires high affinity, tumor-specific agents as effective targeting vehicles to deliver therapeutics and imaging probes to the primary or metastatic tumor sites. Combinatorial libraries such as phage-display and one-bead one-compound (OBOC) peptide libraries are powerful approaches in discovering tumor-targeting peptides. This review gives an overview of different combinatorial library technologies that have been used for the discovery of tumor-targeting peptides. Examples of tumor-targeting peptides identified from each combinatorial library method will be discussed. Published tumor-targeting peptide ligands and their applications will also be summarized by the combinatorial library methods and their corresponding binding receptors.
Collapse
Affiliation(s)
- Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; Division of Hematology & Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Zhou C, Kang J, Wang X, Wei W, Jiang W. Phage display screening identifies a novel peptide to suppress ovarian cancer cells in vitro and in vivo in mouse models. BMC Cancer 2015; 15:889. [PMID: 26555399 PMCID: PMC4641363 DOI: 10.1186/s12885-015-1891-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/01/2015] [Indexed: 02/08/2023] Open
Abstract
Background Ovarian cancer is a possibly lethal gynecological malignancy and this study utilized phage display technology to screen and identify peptides that specifically bind to ovarian cancer cells and explored the effects of these peptides on ovarian cancer cells in vitro and in vivo. Methods The phage displayed peptide library was used to isolate the peptides binding to and internalizing into the ovarian carcinoma cells. Positive phage clones were characterized with DNA sequencing and bioinformatics analysis and then validated with immunofluorescence. Subsequently, the selected peptides were investigated for their cancer-related functions, including cell adhesion, spreading, motility, and invasion in vitro and in vivo. Results Peptide1 read as SWQIGGNwas the positive peptide and showed preferential binding to the target cells. Peptide 1 also inhibited cell proliferation, migration, invasion and adhesion of ovarian cancer HO8910 cells in vitro. In vivo, Peptide 1 led to a lower tumorigenicity of HO8910 cells, which was characterized by the inhibitory effect on tumor growth and metastasis of ovarian cells. Conclusion These studies demonstrate that the phage display-identified tumor cell-binding peptide was able to control ovarian cancer cell viability, migration, invasion, and adhesion capacity in vitro as well as tumor growth and metastasis in vivo. Future studies will be aimed at evaluating the clinical efficacy of the peptide SWQIGGN in ovarian cancer patients.
Collapse
Affiliation(s)
- Cong Zhou
- Department of Obstetrics and Gynecology, Maternity and Children's Healthcare Hospital of Foshan, Foshan, 528000, Guangdong, China.
| | - Jiali Kang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China.
| | - Xiaoxia Wang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China.
| | - Wei Wei
- Foshan Hospital of TCM, Foushan, 52800, China.
| | - Wenyan Jiang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China.
| |
Collapse
|
19
|
Sah BNP, Vasiljevic T, McKechnie S, Donkor ON. Identification of Anticancer Peptides from Bovine Milk Proteins and Their Potential Roles in Management of Cancer: A Critical Review. Compr Rev Food Sci Food Saf 2015; 14:123-138. [DOI: 10.1111/1541-4337.12126] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/03/2014] [Indexed: 01/06/2023]
Affiliation(s)
- B. N. P. Sah
- College of Health and Biomedicine; Victoria Univ; Werribee Campus; PO Box 14428 Melbourne Victoria 8001 Australia
| | - T. Vasiljevic
- College of Health and Biomedicine; Victoria Univ; Werribee Campus; PO Box 14428 Melbourne Victoria 8001 Australia
| | - S. McKechnie
- College of Engineering and Science; Victoria Univ; Werribee Campus; PO Box 14428 Melbourne Victoria 8001 Australia
| | - O. N. Donkor
- College of Health and Biomedicine; Victoria Univ; Werribee Campus; PO Box 14428 Melbourne Victoria 8001 Australia
| |
Collapse
|
20
|
Wu D, Gao Y, Qi Y, Chen L, Ma Y, Li Y. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett 2014; 351:13-22. [PMID: 24836189 DOI: 10.1016/j.canlet.2014.05.002] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/31/2014] [Accepted: 05/01/2014] [Indexed: 01/01/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Conventional cancer therapies mainly focus on mass cell killing without high specificity and often cause severe side effects and toxicities. Peptides are a novel class of anticancer agents that could specifically target cancer cells with lower toxicity to normal tissues, which will offer new opportunities for cancer prevention and treatment. Anticancer peptides face several therapeutic challenges. In this review, we present the sources and mechanisms of anticancer peptides and further discuss modification strategies to improve the anticancer effects of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanfeng Gao
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanming Qi
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Lixiang Chen
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanfang Ma
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanzhang Li
- College of Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
21
|
D'Onofrio N, Caraglia M, Grimaldi A, Marfella R, Servillo L, Paolisso G, Balestrieri ML. Vascular-homing peptides for targeted drug delivery and molecular imaging: meeting the clinical challenges. Biochim Biophys Acta Rev Cancer 2014; 1846:1-12. [PMID: 24704283 DOI: 10.1016/j.bbcan.2014.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/20/2014] [Accepted: 03/22/2014] [Indexed: 12/12/2022]
Abstract
The vasculature of each organ expresses distinct molecular signatures critically influenced by the pathological status. The heterogeneous profile of the vascular beds has been successfully unveiled by the in vivo phage display, a high-throughput tool for mapping normal, diseased, and tumor vasculature. Specific challenges of this growing field are targeted therapies against cancer and cardiovascular diseases, as well as novel bioimaging diagnostic tools. Tumor vasculature-homing peptides have been extensively evaluated in several preclinical and clinical studies both as targeted-therapy and diagnosis. To date, results from several Phase I and II trials have been reported and many other trials are currently ongoing or recruiting patients. In this review, advances in the identification of novel peptide ligands and their corresponding receptors on tumor endothelium through the in vivo phage display technology are discussed. Emphasis is given to recent findings in the clinical setting of vascular-homing peptides selected by in vivo phage display for the treatment of advanced malignancies and their altered vascular beds.
Collapse
Affiliation(s)
- Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Anna Grimaldi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Raffaele Marfella
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, Piazza Miraglia 2, 80138 Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Giuseppe Paolisso
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, Piazza Miraglia 2, 80138 Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy.
| |
Collapse
|
22
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
23
|
Reis CF, Carneiro AP, Vieira CU, Fujimura PT, Morari EC, Silva SJD, Goulart LR, Ward LS. An antibody-like peptide that recognizes malignancy among thyroid nodules. Cancer Lett 2013; 335:306-13. [PMID: 23462224 DOI: 10.1016/j.canlet.2013.02.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 02/01/2013] [Accepted: 02/18/2013] [Indexed: 12/23/2022]
Abstract
There is an urgent need for biomarkers to identify malignant thyroid nodules from indeterminate follicular lesions. We have used a subtractive proteomic strategy to identify novel biomarkers by selecting ligands to goiter tissue from a 12-mer random peptide phage-displayed library using the BRASIL method (Biopanning and Rapid Analysis of Selective Interactive Ligands). After three rounds of selection, two highly reactive clones to the papillary thyroid tumor cell line NPA were further evaluated, and their specific binding to tumor proteins was confirmed using phage-ELISA. The antibody-like peptide CaT12 was tumor-specific, which was further tested by immunohistochemistry against TMAs (tissue microarrays) comprised of 775 human benign and malignant tissues, including 232 thyroid nodular lesions: 15 normal thyroid tissues, 53 nodular goiters (NG), 54 follicular adenomas (FA); 69 papillary thyroid carcinomas (PTC); and 41 follicular carcinomas (FC). CaT12 was able to identify PTC among thyroid nodular lesions with 91.2% sensitivity and 85.1% specificity, despite its non-specificity for thyroid tissues. Additionally, the CaT12 peptide helped characterize follicular lesions distinguishing the follicular variant of PTC (FVPTC) from FA with 91.9% accuracy; FVPTC from NG with 83.1% accuracy; FVPTC from the classic PTC with 57.7% accuracy; and FVPTC from FC with 88.7% accuracy. In conclusion, our strategy to select differentially expressed ligands to thyroid tissue was highly effective and resulted in a useful antibody-like biomarker that recognizes malignancy among thyroid nodules and may help distinguish follicular patterned lesions.
Collapse
Affiliation(s)
- Carolina Fernandes Reis
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences (FCM), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Azevedo RA, Ferreira AK, Auada AVV, Pasqualoto KFM, Marques-Porto R, Maria DA, Lebrun I. Antitumor Effect of Cationic INKKI Peptide from Bovine <i>β</i>-Casein on Melanoma B16F10. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jct.2012.34034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Matsuo AL, Juliano MA, Figueiredo CR, Batista WL, Tanaka AS, Travassos LR. A New Phage-Display Tumor-Homing Peptide Fused to Antiangiogenic Peptide Generates a Novel Bioactive Molecule with Antimelanoma Activity. Mol Cancer Res 2011; 9:1471-8. [DOI: 10.1158/1541-7786.mcr-10-0501] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Peptides targeting inflamed synovial vasculature attenuate autoimmune arthritis. Proc Natl Acad Sci U S A 2011; 108:12857-62. [PMID: 21768392 DOI: 10.1073/pnas.1103569108] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autoimmune diseases, such as rheumatoid arthritis, frequently target one major tissue/organ despite the systemic nature of the immune response. This is particularly perplexing in the case of ubiquitously distributed antigens invoked in arthritis induction. We reasoned that selective targeting of the synovial joints in autoimmune arthritis might be due in part to the unique attributes of the joint vasculature. We examined this proposition using the adjuvant-induced arthritis model of human rheumatoid arthritis, and profiled the synovial vasculature using ex vivo and in vivo screening of a defined phage peptide-display library. We identified phage that preferentially homed to the inflamed joints. The corresponding synthetic peptides showed binding to the joint-derived endothelial cells, as well as specificity in inhibiting binding of the respective phage to the synovial vasculature. Intriguingly, the treatment of arthritic rats with one such peptide resulted in efficient inhibition of the progression of arthritis. The suppression of arthritis was attributable in part to the peptide-induced reduction of T-cell trafficking into the joints and the inhibition of angiogenesis. This peptide differed in sequence, in receptor binding specificity, and in angiogenesis/inflammation-related cell signaling from the previously characterized arginine-glycine-aspartic acid-containing peptide. Thus, our study reveals joint-homing peptides that can be further exploited for the selective delivery of antiarthritic agents into the inflamed joints to enhance their efficacy while reducing systemic toxicity, and also for examining intricacies of the pathogenesis of arthritis. This approach can be customized for application to other organ-specific autoimmune diseases as well.
Collapse
|