1
|
Li G, Luo Y, Hu Z, Shi Z, Cao X, Xu R, Mi Y, Yao Y, Mao H, Zhang H, Zhu Y. Recent Advances in Peptide-Functionalized Hydrogels for Bone Tissue Engineering. ACS Biomater Sci Eng 2025; 11:1970-1989. [PMID: 40178194 PMCID: PMC12002065 DOI: 10.1021/acsbiomaterials.4c02198] [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: 11/20/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Efficient therapeutic approaches for bone regeneration are urgently required to address the significant challenges associated with the repair of large-scale or long-segment bone defects. Peptide-functionalized hydrogels (PFHs) have emerged as important bioactive materials in bone tissue engineering because they produce biomimetic microenvironments enriched with multiple biochemical signals. This review summarizes the key fabrication techniques for PFHs and discusses their diverse applications in different fields. Furthermore, we systematically highlighted the biochemical functionalization of PFHs, which includes basic functions such as cell adhesion, cell recruitment, and osteoinduction; improved functions such as angiogenesis, biomineralization, immune regulation, and hormone regulation; and other functions, including antimicrobial and antitumor effects. Finally, critical biosafety considerations associated with PFHs and perspectives on developing intelligent PFHs are addressed. This review aims to inspire further research on PFHs and accelerate their applications in bone tissue engineering.
Collapse
Affiliation(s)
- Guanrong Li
- Department
of Orthopaedic Surgery, The First Affiliated
Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yang Luo
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Zeming Hu
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Zheyuan Shi
- Department
of Orthopaedic Surgery, The First Affiliated
Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xu Cao
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Rong Xu
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yunfeng Mi
- Department
of Orthopaedic Surgery, The First Affiliated
Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Yudong Yao
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Haijiao Mao
- Department
of Orthopaedic Surgery, The First Affiliated
Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Hua Zhang
- Research
Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
- State
Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Yingchun Zhu
- Department
of Orthopaedic Surgery, The First Affiliated
Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| |
Collapse
|
2
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Asim MN, Asif T, Mehmood F, Dengel A. Peptide classification landscape: An in-depth systematic literature review on peptide types, databases, datasets, predictors architectures and performance. Comput Biol Med 2025; 188:109821. [PMID: 39987697 DOI: 10.1016/j.compbiomed.2025.109821] [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: 09/28/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
Peptides are gaining significant attention in diverse fields such as the pharmaceutical market has seen a steady rise in peptide-based therapeutics over the past six decades. Peptides have been utilized in the development of distinct applications including inhibitors of SARS-COV-2 and treatments for conditions like cancer and diabetes. Distinct types of peptides possess unique characteristics, and development of peptide-specific applications require the discrimination of one peptide type from others. To the best of our knowledge, approximately 230 Artificial Intelligence (AI) driven applications have been developed for 22 distinct types of peptides, yet there remains significant room for development of new predictors. A Comprehensive review addresses the critical gap by providing a consolidated platform for the development of AI-driven peptide classification applications. This paper offers several key contributions, including presenting the biological foundations of 22 unique peptide types and categorizes them into four main classes: Regulatory, Therapeutic, Nutritional, and Delivery Peptides. It offers an in-depth overview of 47 databases that have been used to develop peptide classification benchmark datasets. It summarizes details of 288 benchmark datasets that are used in development of diverse types AI-driven peptide classification applications. It provides a detailed summary of 197 sequence representation learning methods and 94 classifiers that have been used to develop 230 distinct AI-driven peptide classification applications. Across 22 distinct types peptide classification tasks related to 288 benchmark datasets, it demonstrates performance values of 230 AI-driven peptide classification applications. It summarizes experimental settings and various evaluation measures that have been employed to assess the performance of AI-driven peptide classification applications. The primary focus of this manuscript is to consolidate scattered information into a single comprehensive platform. This resource will greatly assist researchers who are interested in developing new AI-driven peptide classification applications.
Collapse
Affiliation(s)
- Muhammad Nabeel Asim
- German Research Center for Artificial Intelligence, Kaiserslautern, 67663, Germany; Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany.
| | - Tayyaba Asif
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany
| | - Faiza Mehmood
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; Institute of Data Sciences, University of Engineering and Technology, Lahore, Pakistan
| | - Andreas Dengel
- German Research Center for Artificial Intelligence, Kaiserslautern, 67663, Germany; Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany
| |
Collapse
|
4
|
Ojo OA, Grant S, Nwafor-Ezeh PI, Maduakolam-Aniobi TC, Akinborode TI, Ezenabor EH, Ojo AB. Ferroptosis as the new approach to cancer therapy. Cancer Treat Res Commun 2025; 43:100913. [PMID: 40187205 DOI: 10.1016/j.ctarc.2025.100913] [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: 11/26/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025]
Abstract
Cancer is characterized by unregulated cell proliferation, evasion of apoptosis, and a propensity for metastasis, making it a leading cause of morbidity and mortality globally. Major challenges in cancer treatment include drug resistance and tumor heterogeneity, which hinder the clinical efficacy of existing therapies. To enhance treatment outcomes, it is essential to integrate emerging biological insights and technological advancements with conventional therapeutic strategies. Recent research has identified various forms of cell death, which can be classified as either regulated or unregulated. Regulated cell death involves specific biochemical and signaling pathways, while unregulated cell death occurs passively and uncontrollably. Apoptosis, the most extensively studied form of regulated cell death, is primarily mediated by the activation of caspase proteases. Nevertheless, the resistance of many tumors to apoptotic pathways has shifted focus towards non-apoptotic forms of cell death, such as ferroptosis. Ferroptosis is an iron-dependent form of regulated necrosis characterized by extensive membrane damage resulting from lipid peroxidation. Numerous preclinical studies have demonstrated that inducing ferroptosis can significantly reduce tumor growth across a variety of cancer types. For instance, in a study involving breast cancer models, the use of ferroptosis inducers such as erastin and RSL3 led to a marked decrease in tumor volume and weight. This review aims to explore the potential of ferroptosis as a novel therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Oluwafemi Adeleke Ojo
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria; Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Susan Grant
- Department of Biochemistry, Landmark University, Omu-Aran, Nigeria
| | - Pearl Ifunanya Nwafor-Ezeh
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | | | | | - Emmanuel Henry Ezenabor
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | - Adebola Busola Ojo
- Department of Environmental Management and Toxicology, University of Ilesa, Ilesa, Nigeria
| |
Collapse
|
5
|
Hemavathy N, Ranganathan S, Umashankar V, Jeyakanthan J. Computational Development of Allosteric Peptide Inhibitors Targeting LIM Kinases as a Novel Therapeutic Intervention. Cell Biochem Biophys 2025:10.1007/s12013-025-01718-1. [PMID: 40100341 DOI: 10.1007/s12013-025-01718-1] [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] [Accepted: 03/02/2025] [Indexed: 03/20/2025]
Abstract
LIM Kinases (LIMKs) have emerged as critical therapeutic targets in cancer research due to their central role in regulating cytoskeletal dynamics and cell motility via cofilin phosphorylation. Allosteric inhibitors, which bind outside the ATP-binding pocket, offer distinct advantages over ATP-competitive inhibitors, such as increased specificity, reduced off-target effects, and the ability to overcome resistance. This study investigates a series of novel tetrapeptides mimicking the binding mode of TH470, an allosteric LIMK inhibitor, using in silico docking and molecular dynamics simulations to identify potential lead compounds with high specificity, binding affinity, and favorable pharmacokinetic properties. Structural analyses revealed critical interactions between TH470 and LIMKs, particularly with conserved residues such as Thr405 (gatekeeper residue), Ile408 (hinge region), and Asp469 (XDFG motif), which are essential for stabilizing inhibitor binding. Molecular dynamics simulations confirmed the stability of TH470-LIMK1 and TH470-LIMK2 complexes, with lower RMS deviations and robust interaction patterns enhancing binding affinity. From the set of tetrapeptides mimicking TH470 binding mode, only YFYW, WPHW, and YWFP for LIMK1, and PYWG, FYWV, and WFVW for LIMK2 demonstrated high binding affinities, non-toxic profiles, and promising anti-cancer, anti-angiogenic, and anti-inflammatory properties. Among the studied peptides, LIMK1-YFYW and LIMK2-WFVW exhibited the most substantial binding affinities, supported by high hydrogen bond occupancy with key residues such as Ile416 and Thr405. The findings highlight the therapeutic potential of allosteric peptide inhibitors targeting LIMK-mediated pathways in cancer progression. The study underscores the importance of specific interactions with conserved LIMK residues, providing a foundation for further developing selective inhibitors to modulate actin dynamics and combat cancer-related processes.
Collapse
Affiliation(s)
- Nagarajan Hemavathy
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | | | - Vetrivel Umashankar
- Virology & Biotechnology/Bioinformatics Division, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamil Nadu, India
| | - Jeyaraman Jeyakanthan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
6
|
Gómez Hernández NA, Pérez GL, Arteaga AV, Garay Pérez HE, Arguellez BO, Rico AC, Guardia AL, Fernández Massó JR. A sandwich ELISA for the quantification of the anticancer peptide CIGB-552 in human plasma. Anal Biochem 2025; 698:115725. [PMID: 39608624 DOI: 10.1016/j.ab.2024.115725] [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: 09/12/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024]
Abstract
CIGB-552 is a synthetic anticancer peptide that has been evaluated in vitro and in vivo in lung and colon cancer models. To optimize therapy in the clinic, pharmacokinetic studies are necessary. Previously, a sandwich-type enzyme-linked immunosorbent assay (ELISA) had been developed by our working group for the quantification of CIGB-552 in biological matrices. The objective of this work was to carry out the full validation of the ELISA to support its application in clinical trials. First, we obtained a polyclonal antibody specific for CIGB-552 and with purity greater than 95 %. The lower limit of quantification and the upper limit of quantification were 3125 ng/ml and 200 ng/ml, respectively. The method is exact and precise in the quantification of the peptide with relative error and coefficient of variation values less than 20 %. The ELISA is specific in the presence of CIGB-552 metabolites in the sample, and also presents robustness to certain protocol variations. In summary, the validated ELISA meets the requirements for its application in upcoming clinical trials as part of pharmacokinetic studies.
Collapse
Affiliation(s)
| | - Gilda Lemos Pérez
- Chemistry-Physics Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | - Amalia Vazquez Arteaga
- Chemistry-Physics Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | - Hilda Elisa Garay Pérez
- Peptide Synthesis Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | | | - Ania Cabrales Rico
- Chemistry-Physics Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | - Airela Llamo Guardia
- Monoclonal Antibody Production Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | | |
Collapse
|
7
|
Lucchetti D, Rinaldi R, Artemi G, Salvia R, De Stefano F, Scieuzo C, Falabella P, Sgambato A. Peptide Fractions Extracted from the Hemolymph of Hermetia illucens Inhibit Growth and Motility and Enhance the Effects of Traditional Chemotherapeutics in Human Colorectal Cancer Cells. Int J Mol Sci 2025; 26:1891. [PMID: 40076518 PMCID: PMC11899838 DOI: 10.3390/ijms26051891] [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: 01/15/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer is a leading cause of death worldwide, characterized by uncontrolled cell growth and multiple mutations. Chemotherapy is often associated with harmful side effects, and cancer cells may become resistant through various mechanisms. New approaches, which are able to address both the toxicity and resistance issues of chemotherapy, are of primary importance in cancer research. Antimicrobial peptides (AMPs), naturally occurring molecules in the innate immune system of all living organisms, have a wide spectrum of cytotoxic activities against cancer cells and could be a promising alternative to actual chemotherapeutics. Here, we tested peptide fractions, rich in AMPs, extracted from the hemolymph of the larvae of the insect Hermetia illucens on the HT29 and HCT116 human colorectal cancer cells, observing cell growth inhibition by cell accumulation in the G2/M phase and increased apoptosis. Furthermore, the peptide extract induced a significant cytoskeleton reorganization, resulting in reduced motility. These effects were more evident with the peptide fractions obtained from the Escherichia coli-infected larvae. The peptide fractions also enhanced the effects of traditional chemotherapeutics. Overall, the results obtained suggest the presence of biologically active molecules in the hemolymph of H. illucens larvae, confirming that insect-derived peptides are a promising research area in oncology.
Collapse
Affiliation(s)
- Donatella Lucchetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.L.); (G.A.)
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00136 Rome, Italy
| | - Roberta Rinaldi
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.); (C.S.)
- Centro di Riferimento Oncologico della Basilicata IRCCS (IRCCS-CROB), 85028 Rionero in Vulture, Italy
| | - Giulia Artemi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.L.); (G.A.)
| | - Rosanna Salvia
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.); (C.S.)
- Spinoff XFlies S.R.L., University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Federica De Stefano
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.); (C.S.)
| | - Carmen Scieuzo
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.); (C.S.)
- Spinoff XFlies S.R.L., University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Patrizia Falabella
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.); (C.S.)
- Spinoff XFlies S.R.L., University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.L.); (G.A.)
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00136 Rome, Italy
| |
Collapse
|
8
|
Zaghloul NA, Gouda MK, Elbahloul Y, El Halfawy NM. Azurin a potent anticancer and antimicrobial agent isolated from a novel Pseudomonas aeruginosa strain. Sci Rep 2025; 15:3735. [PMID: 39885219 PMCID: PMC11782508 DOI: 10.1038/s41598-025-86649-w] [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: 08/08/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Azurin, a bacterial blue-copper protein, has garnered significant attention as a potential anticancer drug in recent years. Among twenty Pseudomonas aeruginosa isolates, we identified one isolate that demonstrated potent and remarkable azurin synthesis using the VITEK 2 system and 16S rRNA sequencing. The presence of the azurin gene was confirmed in the genomic DNA using specific oligonucleotide primers, and azurin expression was also detected in the synthesized cDNA, which revealed that the azurin expression is active. Furthermore, crude azurin protein was extracted, precipitated using 70% ammonium sulfate, dialyzed, and subjected to purification using carboxymethyl-Sephadex in affinity chromatography as a cheap method for purification. The partially purified azurin protein was characterized using polyacrylamide gel electrophoresis, energy-dispersive X-ray spectroscopy, Fourier-transform infrared spectroscopy, and nuclear magnetic resonance spectroscopy. Notably, qualitative elemental analysis by EDX showed the presence of copper and sulfur, corresponding to the copper-core and disulfide-bridge, respectively, in the purified azurin fraction. Moreover, FTIR spectroscopy revealed characteristic amide I and II absorption peaks (1500-1700 cm- 1), revealing the possible secondary structure of azurin. The results of NMR revealed the presence of characteristic amino acids such as methionine and cysteine, which confirmed the EDX results for sulfur-containing amino acids. Purified azurin exhibited antimicrobial activity against Staphylococcus aureus, Bacillus subtilis, Escherichia coli, and Klebsiella pneumoniae. Additionally, its anticancer properties were determined using the MTT assay and cell cycle analysis, revealing a preference for inhibiting the MCF7 breast cancer cell line where breast cancer is most common in Egypt. Overall, the research findings suggest that the local isolate, P. aeruginosa strain 105, could be a potential source of azurin protein for incorporation into cancer treatment strategies.
Collapse
Affiliation(s)
- Nourhan A Zaghloul
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mona K Gouda
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Yasser Elbahloul
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nancy M El Halfawy
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
9
|
Shoari A, Ashja Ardalan A, Dimesa AM, Coban MA. Targeting Invasion: The Role of MMP-2 and MMP-9 Inhibition in Colorectal Cancer Therapy. Biomolecules 2024; 15:35. [PMID: 39858430 PMCID: PMC11762759 DOI: 10.3390/biom15010035] [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: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the most prevalent and lethal cancers worldwide, prompting ongoing research into innovative therapeutic strategies. This review aims to systematically evaluate the role of gelatinases, specifically MMP-2 and MMP-9, as therapeutic targets in CRC, providing a critical analysis of their potential to improve patient outcomes. Gelatinases, specifically MMP-2 and MMP-9, play critical roles in the processes of tumor growth, invasion, and metastasis. Their expression and activity are significantly elevated in CRC, correlating with poor prognosis and lower survival rates. This review provides a comprehensive overview of the pathophysiological roles of gelatinases in CRC, highlighting their contribution to tumor microenvironment modulation, angiogenesis, and the metastatic cascade. We also critically evaluate recent advancements in the development of gelatinase inhibitors, including small molecule inhibitors, natural compounds, and novel therapeutic approaches like gene silencing techniques. Challenges such as nonspecificity, adverse side effects, and resistance mechanisms are discussed. We explore the potential of gelatinase inhibition in combination therapies, particularly with conventional chemotherapy and emerging targeted treatments, to enhance therapeutic efficacy and overcome resistance. The novelty of this review lies in its integration of recent findings on diverse inhibition strategies with insights into their clinical relevance, offering a roadmap for future research. By addressing the limitations of current approaches and proposing novel strategies, this review underscores the potential of gelatinase inhibitors in CRC prevention and therapy, inspiring further exploration in this promising area of oncological treatment.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Arghavan Ashja Ardalan
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | | | - Mathew A. Coban
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
10
|
Lee YK, Sehar M, Botcha L, Chuang PK. Acetylated Globotetraose (Ac-Gb4) Suppresses Triple-Negative Breast Cancer Through FAK/AKT Signaling Pathway. Int J Mol Sci 2024; 25:13353. [PMID: 39769120 PMCID: PMC11677054 DOI: 10.3390/ijms252413353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) remains a significant therapeutic challenge due to its unresponsiveness to hormone and HER2-targeted treatments. This study investigated the potential of acetylated globotetraose (Ac-Gb4) as a novel therapeutic approach targeting glycolipid-mediated signaling in breast cancer cells. We synthesized acetylated globotetraose (Gb4) to enhance its membrane permeability while preserving its biological recognition properties. Flow cytometry analysis revealed that Ac-Gb4 treatment significantly decreased SSEA3 and SSEA4 expression in MDA-MB-231 breast cancer cells, which are Globo-H-negative cells. Notably, Ac-Gb4 demonstrated selective cytotoxicity against cancer cells by significantly reducing proliferation and inducing apoptosis in MDA-MB-231 cells while sparing hTERT-HME1 normal breast epithelial cells. Mechanistic studies through Western blot analysis revealed that Ac-Gb4 simultaneously modulated multiple signaling pathways, including FAK cleavage, reduced AKT expression, and increased caspase-3 activation, particularly at the 4 mM concentration. These molecular changes correlated with decreased cancer cell invasion capability in a dose-dependent manner. Our findings demonstrated that Ac-Gb4 effectively targeted breast cancer cells through the modulation of critical signaling pathways involved in cell survival and invasion while maintaining a minimal impact on normal cells. This anti-cancer activity suggests that Ac-Gb4 represents a promising therapeutic candidate for breast cancer treatment, particularly for aggressive subtypes such as TNBC.
Collapse
Affiliation(s)
- Yung-Kuo Lee
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Misbah Sehar
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80284, Taiwan
| | - Lavanya Botcha
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80284, Taiwan
| | - Po-Kai Chuang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80284, Taiwan
| |
Collapse
|
11
|
Zare-Zardini H, Saberian E, Jenča A, Ghanipour-Meybodi R, Jenča A, Petrášová A, Jenčová J. From defense to offense: antimicrobial peptides as promising therapeutics for cancer. Front Oncol 2024; 14:1463088. [PMID: 39445062 PMCID: PMC11496142 DOI: 10.3389/fonc.2024.1463088] [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: 07/11/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Antimicrobial peptides (AMPs), naturally occurring components of innate immunity, are emerging as a promising new class of anticancer agents. This review explores the potential of AMPs as a novel class of anticancer agents. AMPs, naturally occurring peptides with broad-spectrum antimicrobial activity, exhibit several characteristics that make them attractive candidates for cancer therapy, including selectivity for cancer cells, broad-spectrum activity, and immunomodulatory effects. Analysis of a dataset of AMPs with anticancer activity reveals that their effectiveness is influenced by various structural properties, including net charge, length, Boman index, and hydrophobicity. These properties contribute to their ability to target and disrupt cancer cell membranes, interfere with intracellular processes, and modulate the immune response. The review highlights the promising potential of AMPs as a new frontier in cancer treatment, offering hope for more effective and less toxic therapies. AMPs demonstrate promising potential in cancer therapy through multiple mechanisms, including direct cytotoxicity, immune response modulation, and targeting of the tumor microenvironment, as evidenced by extensive preclinical studies in animal models showing tumor regression, metastasis inhibition, and improved survival rates. AMPs show significant potential as cancer therapeutics through their direct cytotoxicity, immune response modulation, and tumor microenvironment targeting, with promising results from preclinical studies and early-phase clinical trials. Future research should focus on optimizing AMP properties, developing novel delivery strategies, and exploring synergistic combination therapies to fully realize their potential as effective cancer treatments, while addressing challenges related to stability, delivery, and potential toxicity.
Collapse
Affiliation(s)
- Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | | | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| |
Collapse
|
12
|
Hassoon AA, Smith SJ, Harrison RG. Cadmium and silver complexes of a pyridine containing ligand: syntheses, structural studies, biological activity and docking studies. RSC Adv 2024; 14:31850-31860. [PMID: 39380643 PMCID: PMC11459448 DOI: 10.1039/d4ra05305b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
The current study aimed to synthesize seven new metal coordination complexes (Q1-Q7) with potential biomedical applications. Novel mononuclear, polynuclear and mixed-ligand coordination compounds of the elements, cadmium(ii) and silver(i) derived from a pyridine containing ligand (2,4,6-tris-(2-pyridyl)-1,3,5-triazine (TPT)) have been synthesized successfully with the general formulae [Cd(TPT)Cl6]·H2O and [Ag x (TPT) y (L)2(ClO4)](ClO4) z (x = 1,2,3, y = 1,2,3, L = PPh3 or phen, z = 1,2). The structural features were fully characterized using various spectroscopic techniques, such as infrared, ultraviolet-visible spectroscopy, 1D and 2D-NMR (1H, 13C, 31P, 1H-1H COSY and 1H-13C HSQCAD), CHN analysis, molar conductance (Λ), thermogravimetric analysis (TGA), and powder X-ray diffraction analysis. The structure of complex Q6 was also confirmed by single-crystal X-ray analysis. The luminescence and electrochemical properties of complexes, in solution, have been studied. X-ray crystallographic determination of the [Ag(TPT)(PPh3)2]ClO4·EtOH (Q6) complex shows that the Ag+ cation is bonded to one tridentate TPT ligand through NNN set of donor atoms and two triphenylphosphine ligands, giving the Ag+ a distorted trigonal bipyramidal geometry. X-ray powder diffraction analysis showed that metal complexes Q3, Q6 and Q7 display crystalline peaks. The complexes were evaluated for their in vitro antibacterial efficacy against various bacterial and fungal species. The in vitro efficacy against the MCF-7 human breast cancer cell line was assessed to determine the anticancer activities. The tri-nuclear silver complex Q3 shows great potential as a therapeutic candidate for treating breast cancer, since it exhibits a half-maximal inhibition concentration (IC50) of 13.45 ± 0.9 μM. Molecular docking simulations were also carried out to evaluate the interaction strength and properties of the metal complexes with selected cancer and bacteria relevant proteins namely cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 6 (CDK6), signal transducer and activator of transcription 3 (STAT3), and beta-lactamases from Escherichia coli and Staphylococcus aureus.
Collapse
Affiliation(s)
- Azza A Hassoon
- Chemistry Department, Faculty of Science, Mansoura University Mansoura 35516 Egypt
| | - Stacey J Smith
- Department of Chemistry & Biochemistry, Brigham Young University USA
| | - Roger G Harrison
- Department of Chemistry & Biochemistry, Brigham Young University USA
| |
Collapse
|
13
|
Jalil AT, Abdulhadi MA, Al-Ameer LR, Taher WM, Abdulameer SJ, Abosaooda M, Fadhil AA. Peptide-Based Therapeutics in Cancer Therapy. Mol Biotechnol 2024; 66:2679-2696. [PMID: 37768503 DOI: 10.1007/s12033-023-00873-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023]
Abstract
A monster called cancer is still one of the most challenging human problems and one of the leading causes of death in the world. Different types of treatment methods are used for cancer therapy; however, there are challenges such as high cost and harmful side effects in using these methods. Recent years have witnessed a surge in the development of therapeutic peptides for a wide range of diseases, notably cancer. Peptides are preferred over antibiotics, radiation therapy, and chemotherapy in the treatment of cancer due to a number of aspects, including flexibility, easy modification, low immunogenicity, and inexpensive cost of production. The use of therapeutic peptides in cancer treatment is a novel and intriguing strategy. These peptides provide excellent prospects for targeted drug delivery because of their high selectivity, specificity, small dimensions, good biocompatibility, and simplicity of modification. Target specificity and minimal toxicity are benefits of therapeutic peptides. Additionally, peptides can be used to design antigens or adjuvants for vaccine development. Here, types of therapeutic peptides for cancer therapy will be discussed, such as peptide-based cancer vaccines and tumor-targeting peptides (TTP) and cell-penetrating peptides (CPP).
Collapse
Affiliation(s)
- Abduladheem Turki Jalil
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hilla, Babylon, 51001, Iraq.
| | - Mohanad Ali Abdulhadi
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Lubna R Al-Ameer
- College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | | | - Sada Jasim Abdulameer
- Biology Department, College of Education for Pure Science, Wasit University, Kut, Wasit, Iraq
| | | | - Ali A Fadhil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
14
|
Zareei S, Khorsand B, Dantism A, Zareei N, Asgharzadeh F, Zahraee SS, Mashreghi Kashan S, Hekmatirad S, Amini S, Ghasemi F, Moradnia M, Vaghf A, Hemmatpour A, Hourfar H, Niknia S, Johari A, Salimi F, Fariborzi N, Shojaei Z, Asiaei E, Shabani H. PeptiHub: a curated repository of precisely annotated cancer-related peptides with advanced utilities for peptide exploration and discovery. Database (Oxford) 2024; 2024:baae092. [PMID: 39308247 PMCID: PMC11417155 DOI: 10.1093/database/baae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 08/07/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024]
Abstract
Peptihub (https://bioinformaticscollege.ir/peptihub/) is a meticulously curated repository of cancer-related peptides (CRPs) that have been documented in scientific literature. A diverse collection of CRPs is included in the PeptiHub, showcasing a spectrum of effects and activities. While some peptides demonstrated significant anticancer efficacy, others exhibited no discernible impact, and some even possessed alternative non-drug functionalities, including drug carrier or carcinogenic attributes. Presently, Peptihub houses 874 CRPs, subjected to evaluation across 10 distinct organism categories, 26 organs, and 438 cell lines. Each entry in the database is accompanied by easily accessible 3D conformations, obtained either experimentally or through predictive methodology. Users are provided with three search frameworks offering basic, advanced, and BLAST sequence search options. Furthermore, precise annotations of peptides enable users to explore CRPs based on their specific activities (anticancer, no effect, insignificant effect, carcinogen, and others) and their effectiveness (rate and IC50) under cancer conditions, specifically within individual organs. This unique property facilitates the construction of robust training and testing datasets. Additionally, PeptiHub offers 1141 features with the convenience of selecting the most pertinent features to address their specific research questions. Features include aaindex1 (in six main subcategories: alpha propensities, beta propensity, composition indices, hydrophobicity, physicochemical properties, and other properties), amino acid composition (Amino acid Composition and Dipeptide Composition), and Grouped Amino Acid Composition (Grouped amino acid composition, Grouped dipeptide composition, and Conjoint triad) categories. These utilities not only speed up machine learning-based peptide design but also facilitate peptide classification. Database URL: https://bioinformaticscollege.ir/peptihub/.
Collapse
Affiliation(s)
- Sara Zareei
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, South Mofateh Ave. , Tehran 15719-14911, Iran
| | - Babak Khorsand
- Department of Neurology, University of California, 200 S. Manchester Ave., Suite 206 Orange, Irvine, CA 92868-4280, USA
- Department of Computer Engineering, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square , Mashhad 9177948974, Iran
| | - Alireza Dantism
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Jalal AlAhmad HWY, Tehran 14115-111, Iran
| | - Neda Zareei
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili Str, Shiraz 7193711351, Iran
| | - Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Mashhad 9177948564, Iran
| | - Shadi Shams Zahraee
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Dr. Shahriari Sq., Tehran 1983969411, Iran
| | - Samane Mashreghi Kashan
- Department of Medicinal Biotechnology, Faculty of Advanced Technology in Medicine, Golestan University of Medical Sciences, Shast Kola Road, Gorgan 4918936316, Iran
| | - Shirin Hekmatirad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, 16 Azar Ave, Tehran 1416753955, Iran
| | - Shila Amini
- Department of Genetics, Faculty of Advanced Science and Technology, Medical Sciences Branch, Islamic Azad University, Shariati St., Tehran 19395/1495, Iran
| | - Fatemeh Ghasemi
- Department of Computer Engineering, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square , Mashhad 9177948974, Iran
| | - Maryam Moradnia
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Lund BOX 117,221 00, Sweden
| | - Atena Vaghf
- Department of Medical Biotechnology, Faculty of Advanced Technologies, Shahrekord University of Medical Science, Kashani BLVD., Shahrekord 8815713471, Iran
| | - Anahid Hemmatpour
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Aalam Sq., Yazd 8915173149, Iran
| | - Hamdam Hourfar
- Bioprocess Engineering Research Group, Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology, Tehran-Karaj HWY, Tehran 14965/161, Iran
| | - Soudabeh Niknia
- Department of Biology, Kavian Institute of Higher Education, Elahiyeh Blv., Mashhad 91863-74915, Iran
| | - Ali Johari
- Department of Biology, Kavian Institute of Higher Education, Elahiyeh Blv., Mashhad 91863-74915, Iran
| | - Fatemeh Salimi
- Department of Clinical Science, Faculty of Veterinary Medicine, Razi University, Taq-e Bostan, Kermanshah 6714414971, Iran
| | - Neda Fariborzi
- Department of Biology and Biotechnology, Faculty of Molecular Biology and Genetics, University of Pavia, S.da Nuova, Pavia 65, 27100, Italy
| | - Zohreh Shojaei
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, South Mofateh Ave. , Tehran 15719-14911, Iran
| | - Elaheh Asiaei
- Systems Biotechnology Research Group, Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology, Tehran-Karaj HWY., Tehran 14965/161, Iran
| | - Hossein Shabani
- Department of Biology, Faculty of Biosciences, Tehran North Branch, Islamic Azad University, Vafadar Blv., Tehran 1651153311, Iran
| |
Collapse
|
15
|
Gholami S, Mafakher L, Fotouhi F, Bambai B, Cohan RA, Mehrbod P, Shokouhi H, Farahmand B. Computational peptide engineering approach for selection of the new C05 antibody-driven peptide with potency to blocking influenza a virus attachment; from in silico to in vivo. J Biomol Struct Dyn 2024; 42:7730-7746. [PMID: 37553776 DOI: 10.1080/07391102.2023.2241554] [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: 07/21/2023] [Indexed: 08/10/2023]
Abstract
Antiviral drugs are currently used to prevent or treat viral infections like influenza A Virus (IAV). Nonetheless, annual genetic mutations of influenza viruses make them resistant to efficient treatment by current medications. Antiviral peptides have recently attracted researchers' attention and can potentially supplant the current medications. This study aimed to design peptides against IAV propagation. For this purpose, P2 and P3 peptides were computationally designed based on the HCDR3 region of the C05 antibody (a monoclonal antibody that neutralizes influenza HA protein and inhibits the virus attachment). The synthesized peptides were tested against the influenza A virus (A/Puerto Rico/8/34 (H1N1)) in vitro, and the most efficient peptide was selected for in vivo experiments. It was shown that the designed peptide shows much more prophylactic and therapeutic effects against the virus. These findings demonstrated that the designed peptide can control the virus infection without any cytotoxicity effect. Antiviral peptide design is acknowledged as a critical tactic to manage viral infections by preventing viral binding to the host cells.Communicated by Ramaswamy H. Sarma.
Collapse
MESH Headings
- Antiviral Agents/pharmacology
- Antiviral Agents/chemistry
- Peptides/chemistry
- Peptides/pharmacology
- Animals
- Humans
- Virus Attachment/drug effects
- Influenza A virus/drug effects
- Influenza A virus/immunology
- Dogs
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/immunology
- Protein Engineering/methods
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Madin Darby Canine Kidney Cells
- Molecular Dynamics Simulation
- Mice
- Computer Simulation
- Amino Acid Sequence
- Molecular Docking Simulation
- Orthomyxoviridae Infections/virology
- Orthomyxoviridae Infections/drug therapy
- Orthomyxoviridae Infections/immunology
- Influenza, Human/virology
- Influenza, Human/drug therapy
- Influenza, Human/immunology
- Protein Binding
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/pharmacology
- Antibodies, Neutralizing/chemistry
Collapse
Affiliation(s)
- Shima Gholami
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Ladan Mafakher
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Fotouhi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Bijan Bambai
- Department of Systems Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Parvaneh Mehrbod
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Hadiseh Shokouhi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Behrokh Farahmand
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
16
|
List J, Gattringer J, Huszarek S, Marinovic S, Neubauer HA, Kudweis P, Putz EM, Hellinger R, Gotthardt D. Boosting the anti-tumor activity of natural killer cells by caripe 8 - A Carapichea ipecacuanha isolated cyclotide. Biomed Pharmacother 2024; 177:117057. [PMID: 38976957 DOI: 10.1016/j.biopha.2024.117057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
Abstract
Cyclotides are head-to-tail cyclized peptides with a unique cystine-knot motif. Their structure provides exceptional resistance against enzymatic, chemical, or thermal degradation compared to other peptides. Peptide-based therapeutics promise high specificity, selectivity and lower immunogenicity, making them safer alternatives to small molecules or large biologicals. Cyclotides were researched due to their anti-cancer properties by inducing apoptosis in tumor cells in the past, but the impact of cyclotides on cytotoxic immune cells was poorly studied. Natural Killer (NK) cells are cytotoxic innate lymphoid cells and play an important role in the defense against infected, stressed and transformed cells. NK cells do not need prior sensitization and act in an antigen independent manner, holding promising potential in the field of immunotherapy. To investigate the effect of immunomodulatory cyclotides on NK cells, we evaluated several peptide-enriched plant extracts on NK cell mediated cytotoxicity. We observed that the extract samples derived from Carapichea ipecacuanha (Brot.) L. Andersson augments the killing potential of mouse NK cells against different tumor targets in vitro. Subsequent isolation of cyclotides from C. ipecacuanha extracts led to the identification of a primary candidate that enhances cytotoxicity of both mouse and human NK cells. The augmented killing is facilitated by the increased degranulation capacity of NK cells. In addition, we noted a direct toxic effect of caripe 8 on tumor cells, suggesting a dual therapeutic potential in cancer treatment. This study offers novel insights how natural peptides can influence NK cell cytotoxicity. These pre-clinical findings hold significant promise for advancing current immunotherapeutic approaches.
Collapse
Affiliation(s)
- Julia List
- University of Veterinary Medicine Vienna, Vienna, Austria
| | - Jasmin Gattringer
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Sonja Marinovic
- Department of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | | | - Petra Kudweis
- University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eva-M Putz
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roland Hellinger
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | | |
Collapse
|
17
|
Dai J, Ashrafizadeh M, Aref AR, Sethi G, Ertas YN. Peptide-functionalized, -assembled and -loaded nanoparticles in cancer therapy. Drug Discov Today 2024; 29:103981. [PMID: 38614161 DOI: 10.1016/j.drudis.2024.103981] [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: 09/17/2023] [Revised: 03/20/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
The combination of peptides and nanoparticles in cancer therapy has shown synergistic results. Nanoparticle functionalization with peptides can increase their targeting ability towards tumor cells. In some cases, the peptides can develop self-assembled nanoparticles, in combination with drugs, for targeted cancer therapy. The peptides can be loaded into nanoparticles and can be delivered by other drugs for synergistic cancer removal. Multifunctional types of peptide-based nanoparticles, including pH- and redox-sensitive classes, have been introduced in cancer therapy. The tumor microenvironment remolds, and the acceleration of immunotherapy and vaccines can be provided by peptide nanoparticles. Moreover, the bioimaging and labeling of cancers can be mediated by peptide nanoparticles. Therefore, peptides can functionalize nanoparticles in targeted cancer therapy.
Collapse
Affiliation(s)
- Jingyuan Dai
- School of Computer Science and Information Systems, Northwest Missouri State University, Maryville, MO, USA
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Turkey; Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
18
|
Marunganathan V, Guru A, Panda SP, Arockiaraj J. Exploring Therapeutic Potential: A Comprehensive Review of Antimicrobial Peptides in Oral Cancer Management. Int J Pept Res Ther 2024; 30:43. [DOI: 10.1007/s10989-024-10621-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2024] [Indexed: 01/03/2025]
|
19
|
Gibisch M, Müller M, Tauer C, Albrecht B, Hahn R, Cserjan-Puschmann M, Striedner G. A production platform for disulfide-bonded peptides in the periplasm of Escherichia coli. Microb Cell Fact 2024; 23:166. [PMID: 38840157 PMCID: PMC11155123 DOI: 10.1186/s12934-024-02446-6] [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: 04/03/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Recombinant peptide production in Escherichia coli provides a sustainable alternative to environmentally harmful and size-limited chemical synthesis. However, in-vivo production of disulfide-bonded peptides at high yields remains challenging, due to degradation by host proteases/peptidases and the necessity of translocation into the periplasmic space for disulfide bond formation. RESULTS In this study, we established an expression system for efficient and soluble production of disulfide-bonded peptides in the periplasm of E. coli. We chose model peptides with varying complexity (size, structure, number of disulfide bonds), namely parathyroid hormone 1-84, somatostatin 1-28, plectasin, and bovine pancreatic trypsin inhibitor (aprotinin). All peptides were expressed without and with the N-terminal, low molecular weight CASPON™ tag (4.1 kDa), with the expression cassette being integrated into the host genome. During BioLector™ cultivations at microliter scale, we found that most of our model peptides can only be sufficiently expressed in combination with the CASPON™ tag, otherwise expression was only weak or undetectable on SDS-PAGE. Undesired degradation by host proteases/peptidases was evident even with the CASPON™ tag. Therefore, we investigated whether degradation happened before or after translocation by expressing the peptides in combination with either a co- or post-translational signal sequence. Our results suggest that degradation predominantly happened after the translocation, as degradation fragments appeared to be identical independent of the signal sequence, and expression was not enhanced with the co-translational signal sequence. Lastly, we expressed all CASPON™-tagged peptides in two industry-relevant host strains during C-limited fed-batch cultivations in bioreactors. We found that the process performance was highly dependent on the peptide-host-combination. The titers that were reached varied between 0.6-2.6 g L-1, and exceeded previously published data in E. coli. Moreover, all peptides were shown by mass spectrometry to be expressed to completion, including full formation of disulfide bonds. CONCLUSION In this work, we demonstrated the potential of the CASPON™ technology as a highly efficient platform for the production of soluble peptides in the periplasm of E. coli. The titers we show here are unprecedented whenever parathyroid hormone, somatostatin, plectasin or bovine pancreatic trypsin inhibitor were produced in E. coli, thus making our proposed upstream platform favorable over previously published approaches and chemical synthesis.
Collapse
Affiliation(s)
- Martin Gibisch
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Matthias Müller
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Christopher Tauer
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Bernd Albrecht
- Boehringer-Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna, Austria
| | - Rainer Hahn
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Monika Cserjan-Puschmann
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria.
| | - Gerald Striedner
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| |
Collapse
|
20
|
Ghafoor MH, Song BL, Zhou L, Qiao ZY, Wang H. Self-Assembly of Peptides as an Alluring Approach toward Cancer Treatment and Imaging. ACS Biomater Sci Eng 2024; 10:2841-2862. [PMID: 38644736 DOI: 10.1021/acsbiomaterials.4c00491] [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] [Indexed: 04/23/2024]
Abstract
Cancer is a severe threat to humans, as it is the second leading cause of death after cardiovascular diseases and still poses the biggest challenge in the world of medicine. Due to its higher mortality rates and resistance, it requires a more focused and productive approach to provide the solution for it. Many therapies promising to deliver favorable results, such as chemotherapy and radiotherapy, have come up with more negatives than positives. Therefore, a new class of medicinal solutions and a more targeted approach is of the essence. This review highlights the alluring properties, configurations, and self-assembly of peptide molecules which benefit the traditional approach toward cancer therapy while sparing the healthy cells in the process. As targeted drug delivery systems, self-assembled peptides offer a wide spectrum of conjugation, biocompatibility, degradability-controlled responsiveness, and biomedical applications, including cancer treatment and cancer imaging.
Collapse
Affiliation(s)
- Muhammad Hamza Ghafoor
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ben-Li Song
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lei Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Zeng-Ying Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
21
|
Otani T, Suzuki M, Takakura H, Hanaoka H. Synthesis and biological evaluation of EGFR binding peptides for near-infrared photoimmunotherapy. Bioorg Med Chem 2024; 105:117717. [PMID: 38614014 DOI: 10.1016/j.bmc.2024.117717] [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: 02/21/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a new cancer treatment that involves photoimmunotherapy drug injection and NIR light exposure. In NIR-PIT, antibodies are commonly used as target-directed molecules carrying IRDye700DX (IR700). However, antibodies have disadvantages, such as high cost, complex development strategies, and poor tumor penetration. In contrast, peptides have lower production costs, can be easy to chemically synthesize and modify, and can also be used for tumor-targeting like antibodies. In this study, we developed a novel PIT drug using a peptide as the target-directed molecule. Epidermal growth factor receptor (EGFR) was selected as the target, and monovalent and bivalent EGFR-binding peptides were synthesized. The bivalent peptide showed sufficient binding to EGFR-positive cells, and a bivalent peptide-IR700 conjugate with a long linker induced morphological changes in EGFR-positive cells. Additionally, the drug significantly reduced cell viability in vitro in an NIR light-dose- and drug-concentration-dependent manner. These results indicate the feasibility of NIR-PIT in treating cancer using peptide-based drugs.
Collapse
Affiliation(s)
- Takuya Otani
- Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Motofumi Suzuki
- Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Hideo Takakura
- Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Hirofumi Hanaoka
- Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan.
| |
Collapse
|
22
|
Zhang W, Zhang K, Shi J, Qiu H, Kan C, Ma Y, Hou N, Han F, Sun X. The impact of the senescent microenvironment on tumorigenesis: Insights for cancer therapy. Aging Cell 2024; 23:e14182. [PMID: 38650467 PMCID: PMC11113271 DOI: 10.1111/acel.14182] [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: 01/17/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
The growing global burden of cancer, especially among people aged 60 years and over, has become a key public health issue. This trend suggests the need for a deeper understanding of the various cancer types in order to develop universally effective treatments. A prospective area of research involves elucidating the interplay between the senescent microenvironment and tumor genesis. Currently, most oncology research focuses on adulthood and tends to ignore the potential role of senescent individuals on tumor progression. Senescent cells produce a senescence-associated secretory phenotype (SASP) that has a dual role in the tumor microenvironment (TME). While SASP components can remodel the TME and thus hinder tumor cell proliferation, they can also promote tumorigenesis and progression via pro-inflammatory and pro-proliferative mechanisms. To address this gap, our review seeks to investigate the influence of senescent microenvironment changes on tumor development and their potential implications for cancer therapies.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Fang Han
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| |
Collapse
|
23
|
Liang J, Du B, Wan M, Sun L, Qin S, Nian F, Tang D. Effects of L-Leu-L-Leu peptide on growth, proliferation, and apoptosis in broiler intestinal epithelial cells. Poult Sci 2024; 103:103582. [PMID: 38457989 PMCID: PMC11067774 DOI: 10.1016/j.psj.2024.103582] [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: 11/12/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/10/2024] Open
Abstract
Small peptides are nutrients and bioactive molecules that have dual regulatory effects on nutrition and physiology. They are of great significance for maintaining the intestinal health and production performance of broilers. We here cultured the primary small intestinal epithelial cells (IEC) of chicken in a medium containing L-Leu (Leu) and L-Leu-L-Leu (Leu-Leu) for 24 h. The untreated cells were considered as the control group. The growth, proliferation, and apoptosis of IEC were examined. By combining RNA-seq and label-free sequencing technology, candidate genes, proteins, and pathways related to the growth, proliferation, and apoptosis of IEC were screened. Immunofluorescence detection revealed that the purity of the isolated primary IEC was >90%. The Leu-Leu group significantly promoted IEC growth and proliferation and significantly inhibited IEC apoptosis, and the effect was better than those of the Leu and control groups. Using transcriptome sequencing, four candidate genes, CCL20, IL8L1, IL8, and IL6, were screened in the Leu group, and one candidate gene, IL8, was screened in the Leu-Leu group. Two candidate genes, IL6 and RGN, were screened in the Leu-Leu group compared with the Leu group. Nonquantitative proteomic marker sequencing results revealed that through the screening of candidate proteins and pathways, found one growth-related candidate protein PGM3 and three proliferation-related candidate proteins RPS17, RPS11, and RPL23, and two apoptosis-related candidate proteins GPX4 and PDPK1 were found in the Leu-Leu group compared with Leu group. In short, Leu-Leu could promote IEC growth and proliferation and inhibit IEC apoptosis. On combining transcriptome and proteome sequencing technologies, multiple immune- and energy-related regulatory signal pathways were found to be related to IEC growth, proliferation, and apoptosis. Three candidate genes of IL8, IL6, and RGN were identified, and six candidate proteins of PGM3, RPS17, RPS11, RPL23, GPX4, and PDPK1 were involved in IEC growth, proliferation, and apoptosis. The results provide valuable data for preliminarily elucidating small peptide-mediated IEC regulation pathways, improving the small peptide nutrition theoretical system, and establishing small peptide nutrition regulation technology.
Collapse
Affiliation(s)
- Jing Liang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Baolong Du
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Minyan Wan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Likun Sun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shizhen Qin
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Fang Nian
- College of Science, Gansu Agricultural University, Lanzhou 730070, China
| | - Defu Tang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
24
|
Guhe V, Singh S. Targeting peptide based therapeutics: Integrated computational and experimental studies of autophagic regulation in host-parasite interaction. ChemMedChem 2024; 19:e202300679. [PMID: 38317307 DOI: 10.1002/cmdc.202300679] [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: 12/03/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/07/2024]
Abstract
Cutaneous leishmaniasis caused by the intracellular parasite Leishmania major, exhibits significant public health challenge worldwide. With limited treatment options available, the identification of novel therapeutic targets is of paramount importance. Present study manifested the crucial role of ATG8 protein as a potential target in combating L. major infection. Using machine learning algorithms, we identified non-conserved motifs within the ATG8 in L. major. Subsequently, a peptide library was generated based on these motifs, and three peptides were selected for further investigation through molecular docking and molecular dynamics simulations. Surface Plasmon Resonance (SPR) experiments confirmed the direct interaction between ATG8 and the identified peptides. Remarkably, these peptides demonstrated the ability to cross the parasite membrane and exert profound effects on L. major. Peptide treatment significantly impacted parasite survival, inducing alterations in the cell cycle and morphology. Furthermore, the peptides were found to modulate autophagosome formation, particularly under starved conditions, indicating their involvement in autophagy regulation within L. major. In vitro studies revealed that the selected peptides effectively decreased the parasite load within the infected host cells. Encouragingly, in vivo experiments corroborated these findings, demonstrating a reduction in parasite burden upon peptide administration. Additionally, the peptides were observed to affect the levels of LC3II, a known autophagy marker within the host cells. Collectively, our findings highlight the efficacy of these novel peptides in targeting L. major ATG8 and disrupting parasite survival, wherein P2 is showing prominent effect on L. major as compared to P1. These results provide valuable insights into the development of innovative therapeutic strategies against leishmaniasis.
Collapse
Affiliation(s)
- Vrushali Guhe
- Systems Medicine Lab, National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India Phone
| | - Shailza Singh
- Systems Medicine Lab, National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India Phone
| |
Collapse
|
25
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 86] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
26
|
Azad H, Akbar MY, Sarfraz J, Haider W, Riaz MN, Ali GM, Ghazanfar S. G-ACP: a machine learning approach to the prediction of therapeutic peptides for gastric cancer. J Biomol Struct Dyn 2024:1-14. [PMID: 38450672 DOI: 10.1080/07391102.2024.2323141] [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: 11/14/2023] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
Conventional Gastrointestinal (GI) cancer treatments are quite expensive and have major hazards. Nowadays, a different strategy places more emphasis on creating tiny biologically active peptides that do not cause severe poisoning. Anticancer peptides (ACPs) are found through experimental screening, which is time-dependent and frequently fraught with difficulties. Gastric ACPs are emerging as a promising GI cancer treatment in the current day. It is crucial to identify novel gastric ACPs to have an improved knowledge of their functioning processes and treatment of gastric cancer. As a result of the post-genomic era's massive production of peptide sequences, rapid and effective ACPs using a computational method are essential. Several adaptive statistical techniques for distinguishing ACPs and non-ACPs have recently been developed. A variety of adapted statistically significant methods have been developed to differentiate between ACPs and non-ACPs. Despite significant progress, there is no specific model for the prediction of gastric ACPs because the specific model will predict a particular type of peptide more accurately and quickly. To overcome this, an initiative is taken for the creation of a reliable framework for the accurate identification of gastric ACPs. The current technique in particular contains four possible features along with one hybrid feature encoding mechanisms which are the target-class motif previously indicated by Amino Acid Composition, Dipeptide Composition, Tripeptide Composition (TPC), Pseudo Amino Acid Composition (PAAC), and their Hybrid. Machine Learning algorithms make high-performance and accurate prediction tools. Moreover, highly variable and ideal deep feature selection is done using an ANOVA-based F score for feature pruning. Experiments on a range of algorithms are carried out to identify the optimal operating strategy due to the diverse nature of learning. Following analysis of the empirical results, Naïve Bayes with TPC and Hybrid feature space outperforms other methods with 0.99 accuracy score on the testing dataset. To find the model generalization an external validation is carried out. In external datasets, the Extra Trees with PAAC features outperforms with the accuracy of 0.94. The comparison study shows that our suggested model will predict gastric ACPs more accurately and will be useful in drug development and gastric cancer. The predictive model can be freely accessed at https://github.com/humeraazad10/G-ACP.git.
Collapse
Affiliation(s)
- Humera Azad
- Department of Biosciences (Bioinformatics) Islamabad, Comsats University Islamabad, Pakistan
| | - Muhammad Yasir Akbar
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Center (NARC), Pakistan
| | | | - Waseem Haider
- Department of Biosciences (Bioinformatics) Islamabad, Comsats University Islamabad, Pakistan
| | - Muhammad Naeem Riaz
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Center (NARC), Pakistan
| | - Ghulam Muhammad Ali
- Department of Biosciences (Bioinformatics) Islamabad, Comsats University Islamabad, Pakistan
| | - Shakira Ghazanfar
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Center (NARC), Pakistan
| |
Collapse
|
27
|
Hayward D, Beekman AM. Strategies for converting turn-motif and cyclic peptides to small molecules for targeting protein-protein interactions. RSC Chem Biol 2024; 5:198-208. [PMID: 38456035 PMCID: PMC10915966 DOI: 10.1039/d3cb00222e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
The development of small molecules that interact with protein-protein interactions is an ongoing challenge. Peptides offer a starting point in the drug discovery process for targeting protein-interactions due to their larger, more flexible structure and the structurally diverse properties that allow for a greater interaction with the protein. The techniques for rapidly identifying potent cyclic peptides and turn-motif peptides are highly effective, but this potential has not yet transferred to approved drug candidates. By applying the properties of the peptide-protein interaction the development of small molecules for drug discovery has the potential to be more efficient. In this review, we discuss the methods that allow for the unique binding properties of peptides to proteins, and the methods deployed to transfer these qualities to potent small molecules.
Collapse
Affiliation(s)
- Deanne Hayward
- School of Pharmacy, University of East Anglia, Norwich Research Park Norwich Norfolk NR47TJ UK
| | - Andrew M Beekman
- School of Pharmacy, University of East Anglia, Norwich Research Park Norwich Norfolk NR47TJ UK
| |
Collapse
|
28
|
Shanthappa PM, Melethadathil N. In silico investigations and molecular insights for designing tRNA-encoded peptides as potential therapeutics for targeting over-expressed receptors in breast cancer. J Biomol Struct Dyn 2024:1-17. [PMID: 38334133 DOI: 10.1080/07391102.2024.2314748] [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: 10/21/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
tRNA- Encoded Peptides (tREPs) have recently been discovered as new functional peptides and hold promise as therapeutics for anti-parasitic applications. In this study, in silico investigations were conducted to design tRNA-encoded peptides with the potential to target over-expressed receptors in breast cancer cells. tRNA genes were translated into corresponding peptides (tREPs) using computational tools. The tREPs, which were predicted as anticancer peptides, were then screened for various ADMET properties. Molecular docking studies were conducted for three cancer target receptors, the Estrogen Receptor (ER), Peroxisome Proliferator-Activated Receptor (PPAR) and the Epidermal Growth Factor Receptor (EGFR). Based on the docking results, specific tREPs were screened and molecular dynamics simulations were performed, and the binding energies were further explored using MMPBSA calculations. The peptide Pep1 (DWIAWRHHNDIVSWLTCGPRFKSWS) and Pep2 (GFIAWWSRHLELAQTRFKSWWS) exhibited a good binding affinity against the Estrogen Receptor (ER) and the Peroxisome Proliferator-Activated Receptor Alpha (PPAR) cancer target. The Pep1-ER and Pep1-PPAR complex maintained an average of two hydrogen bonds throughout the simulation and demonstrated a higher negative binding free energy of -72.27 kcal/mol and -65.16 kcal/mol respectively, as calculated by MMPBSA. Therefore, the tREPs designed as anticancer peptides in this study provide novel approaches for potential anticancer therapeutic modalities.
Collapse
Affiliation(s)
- Pallavi M Shanthappa
- Department of Computer Science, School of Computing, Mysuru, Amrita Vishwa Vidyapeetham, India
| | | |
Collapse
|
29
|
Avgoustakis K, Angelopoulou A. Biomaterial-Based Responsive Nanomedicines for Targeting Solid Tumor Microenvironments. Pharmaceutics 2024; 16:179. [PMID: 38399240 PMCID: PMC10892652 DOI: 10.3390/pharmaceutics16020179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Solid tumors are composed of a highly complex and heterogenic microenvironment, with increasing metabolic status. This environment plays a crucial role in the clinical therapeutic outcome of conventional treatments and innovative antitumor nanomedicines. Scientists have devoted great efforts to conquering the challenges of the tumor microenvironment (TME), in respect of effective drug accumulation and activity at the tumor site. The main focus is to overcome the obstacles of abnormal vasculature, dense stroma, extracellular matrix, hypoxia, and pH gradient acidosis. In this endeavor, nanomedicines that are targeting distinct features of TME have flourished; these aim to increase site specificity and achieve deep tumor penetration. Recently, research efforts have focused on the immune reprograming of TME in order to promote suppression of cancer stem cells and prevention of metastasis. Thereby, several nanomedicine therapeutics which have shown promise in preclinical studies have entered clinical trials or are already in clinical practice. Various novel strategies were employed in preclinical studies and clinical trials. Among them, nanomedicines based on biomaterials show great promise in improving the therapeutic efficacy, reducing side effects, and promoting synergistic activity for TME responsive targeting. In this review, we focused on the targeting mechanisms of nanomedicines in response to the microenvironment of solid tumors. We describe responsive nanomedicines which take advantage of biomaterials' properties to exploit the features of TME or overcome the obstacles posed by TME. The development of such systems has significantly advanced the application of biomaterials in combinational therapies and in immunotherapies for improved anticancer effectiveness.
Collapse
Affiliation(s)
- Konstantinos Avgoustakis
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece;
- Clinical Studies Unit, Biomedical Research Foundation Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece
| | - Athina Angelopoulou
- Department of Chemical Engineering, Polytechnic School, University of Patras, 26504 Patras, Greece
| |
Collapse
|
30
|
Zheng Z, Wei X, Lin Y, Tao S, Li H, Ji Z, Wei H, Jin J, Zhao F, Lang C, Liu J, Chen J. In vivo therapy of osteosarcoma using anion transporters-based supramolecular drugs. J Nanobiotechnology 2024; 22:29. [PMID: 38216937 PMCID: PMC10787436 DOI: 10.1186/s12951-023-02270-x] [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: 09/27/2023] [Accepted: 12/14/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Osteosarcoma represents a serious clinical challenge due to its widespread genomic alterations, tendency for drug resistance and distant metastasis. New treatment methods are urgently needed to address those treatment difficulties in osteosarcoma to improve patient prognoses. In recent years, small-molecule based anion transporter have emerged as innovative and promising therapeutic compound with various biomedical applications. However, due to a lack of efficient delivery methods, using ion transporters as therapeutic drugs in vivo remains a major challenge. RESULT Herein, we developed self-assembled supramolecular drugs based on small-molecule anion transporters, which exhibited potent therapeutic effect towards osteosarcoma both in vitro and in vivo. The anion transporters can disrupt intracellular ion homeostasis, inhibit proliferation, migration, epithelial-mesenchymal transition process, and lead to osteosarcoma cell death. RNA sequencing, western blot and flow cytometry indicated reprogramming of HOS cells and induced cell death through multiple pathways. These pathways included activation of endoplasmic reticulum stress, autophagy, apoptosis and cell cycle arrest, which avoided the development of drug resistance in osteosarcoma cells. Functionalized with osteosarcoma targeting peptide, the assembled supramolecular drug showed excellent targeted anticancer therapy against subcutaneous xenograft tumor and lung metastasis models. Besides good tumor targeting capability and anti-drug resistance, the efficacy of the assembly was also attributed to its ability to regulate the tumor immune microenvironment in vivo. CONCLUSIONS In summary, we have demonstrated for the first time that small-molecule anion transporters are capable of killing osteosarcoma cells through multiple pathways. The assemblies, OTP-BP-L, show excellent targeting and therapeutic effect towards osteosarcoma tumors. Furthermore, the supramolecular drug shows a strong ability to regulate the tumor immune microenvironment in vivo. This work not only demonstrated the biomedical value of small-molecule anion transporters in vivo, but also provided an innovative approach for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Zeyu Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiaoan Wei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yangyang Lin
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hui Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhongyin Ji
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hongxin Wei
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Jiayan Jin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chao Lang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China.
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China.
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
31
|
Stefanik O, Majerova P, Kovac A, Mikus P, Piestansky J. Capillary electrophoresis in the analysis of therapeutic peptides-A review. Electrophoresis 2024; 45:120-164. [PMID: 37705480 DOI: 10.1002/elps.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023]
Abstract
Therapeutic peptides are a growing class of innovative drugs with high efficiency and a low risk of adverse effects. These biomolecules fall within the molecular mass range between that of small molecules and proteins. However, their inherent instability and potential for degradation underscore the importance of reliable and effective analytical methods for pharmaceutical quality control, therapeutic drug monitoring, and compliance testing. Liquid chromatography-mass spectrometry (LC-MS) has long time been the "gold standard" conventional method for peptide analysis, but capillary electrophoresis (CE) is increasingly being recognized as a complementary and, in some cases, superior, highly efficient, green, and cost-effective alternative technique. CE can separate peptides composed of different amino acids owing to differences in their net charge and size, determining their migration behavior in an electric field. This review provides a comprehensive overview of therapeutic peptides that have been used in the clinical environment for the last 25 years. It describes the properties, classification, current trends in development, and clinical use of therapeutic peptides. From the analytical point of view, it discusses the challenges associated with the analysis of therapeutic peptides in pharmaceutical and biological matrices, as well as the evaluation of CE as a whole and the comparison with LC methods. The article also highlights the use of microchip electrophoresis, nonaqueous CE, and nonconventional hydrodynamically closed CE systems and their applications. Overall, the article emphasizes the importance of developing new CE-based analytical methods to ensure the high quality, safety, and efficacy of therapeutic peptides in clinical practice.
Collapse
Affiliation(s)
- Ondrej Stefanik
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Juraj Piestansky
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
| |
Collapse
|
32
|
Alamdari-Palangi V, Jaberi KR, Shahverdi M, Naeimzadeh Y, Tajbakhsh A, Khajeh S, Razban V, Fallahi J. Recent advances and applications of peptide-agent conjugates for targeting tumor cells. J Cancer Res Clin Oncol 2023; 149:15249-15273. [PMID: 37581648 DOI: 10.1007/s00432-023-05144-9] [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: 05/23/2023] [Accepted: 07/08/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Cancer, being a complex disease, presents a major challenge for the scientific and medical communities. Peptide therapeutics have played a significant role in different medical practices, including cancer treatment. METHOD This review provides an overview of the current situation and potential development prospects of anticancer peptides (ACPs), with a particular focus on peptide vaccines and peptide-drug conjugates for cancer treatment. RESULTS ACPs can be used directly as cytotoxic agents (molecularly targeted peptides) or can act as carriers (guiding missile) of chemotherapeutic agents and radionuclides by specifically targeting cancer cells. More than 60 natural and synthetic cationic peptides are approved in the USA and other major markets for the treatment of cancer and other diseases. Compared to traditional cancer treatments, peptides exhibit anticancer activity with high specificity and the ability to rapidly kill target cancer cells. ACP's target and kill cancer cells via different mechanisms, including membrane disruption, pore formation, induction of apoptosis, necrosis, autophagy, and regulation of the immune system. Modified peptides have been developed as carriers for drugs, vaccines, and peptide-drug conjugates, which have been evaluated in various phases of clinical trials for the treatment of different types of solid and leukemia cancer. CONCLUSIONS This review highlights the potential of ACPs as a promising therapeutic option for cancer treatment, particularly through the use of peptide vaccines and peptide-drug conjugates. Despite the limitations of peptides, such as poor metabolic stability and low bioavailability, modified peptides show promise in addressing these challenges. Various mechanism of action of anticancer peptides. Modes of action against cancer cells including: inducing apoptosis by cytochrome c release, direct cell membrane lysis (necrosis), inhibiting angiogenesis, inducing autophagy-mediated cell death and immune cell regulation.
Collapse
Affiliation(s)
- Vahab Alamdari-Palangi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Khojaste Rahimi Jaberi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahshid Shahverdi
- Medical Biotechnology Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| |
Collapse
|
33
|
Nguyen TN, Teimouri H, Kolomeisky AB. Increasing Heterogeneity in Antimicrobial Peptide Combinations Enhances Their Synergistic Activities. J Phys Chem Lett 2023; 14:8405-8411. [PMID: 37708492 DOI: 10.1021/acs.jpclett.3c02216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Antimicrobial peptides (AMPs) are short biopolymers produced by living organisms as an immune system defense against infections. They have been considered as potential alternatives to conventional antibiotics. Experiments suggest that combining several types of different AMPs might enhance their antimicrobial activity more effectively than using single-component AMPs. However, a clear understanding of the underlying microscopic mechanisms is still lacking. We present a theoretical investigation of antibacterial cooperativity mechanisms involving several types of AMPs. It is argued that synergy results from intermolecular interactions when the presence of one type of AMP stimulates the association of another type of AMP to bacteria. It is found that increasing the number of different AMPs in the mixtures increases the number of such interactions, making them more efficient in eliminating infections. Our theoretical framework provides valuable insights into the mechanisms of antimicrobial action.
Collapse
Affiliation(s)
- Thao N Nguyen
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, United States
- Applied Physics Graduate Program, Smalley-Curl Institute, Rice University, Houston, Texas 77005, United States
| | - Hamid Teimouri
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, United States
| | - Anatoly B Kolomeisky
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, United States
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77005, United States
- Department of Physics and Astronomy, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
34
|
Tao H, Shan S, Fu H, Zhu C, Liu B. An Augmented Sample Selection Framework for Prediction of Anticancer Peptides. Molecules 2023; 28:6680. [PMID: 37764455 PMCID: PMC10535447 DOI: 10.3390/molecules28186680] [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: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Anticancer peptides (ACPs) have promising prospects for cancer treatment. Traditional ACP identification experiments have the limitations of low efficiency and high cost. In recent years, data-driven deep learning techniques have shown significant potential for ACP prediction. However, data-driven prediction models rely heavily on extensive training data. Furthermore, the current publicly accessible ACP dataset is limited in size, leading to inadequate model generalization. While data augmentation effectively expands dataset size, existing techniques for augmenting ACP data often generate noisy samples, adversely affecting prediction performance. Therefore, this paper proposes a novel augmented sample selection framework for the prediction of anticancer peptides (ACPs-ASSF). First, the prediction model is trained using raw data. Then, the augmented samples generated using the data augmentation technique are fed into the trained model to compute pseudo-labels and estimate the uncertainty of the model prediction. Finally, samples with low uncertainty, high confidence, and pseudo-labels consistent with the original labels are selected and incorporated into the training set to retrain the model. The evaluation results for the ACP240 and ACP740 datasets show that ACPs-ASSF achieved accuracy improvements of up to 5.41% and 5.68%, respectively, compared to the traditional data augmentation method.
Collapse
Affiliation(s)
- Huawei Tao
- Key Laboratory of Food Information Processing and Control, Ministry of Education, Henan University of Technology, Zhengzhou 450001, China; (H.T.); (S.S.); (H.F.); (C.Z.)
- Henan Engineering Laboratory of Grain IOT Technology, Henan University of Technology, Zhengzhou 450001, China
| | - Shuai Shan
- Key Laboratory of Food Information Processing and Control, Ministry of Education, Henan University of Technology, Zhengzhou 450001, China; (H.T.); (S.S.); (H.F.); (C.Z.)
- Henan Engineering Laboratory of Grain IOT Technology, Henan University of Technology, Zhengzhou 450001, China
| | - Hongliang Fu
- Key Laboratory of Food Information Processing and Control, Ministry of Education, Henan University of Technology, Zhengzhou 450001, China; (H.T.); (S.S.); (H.F.); (C.Z.)
- Henan Engineering Laboratory of Grain IOT Technology, Henan University of Technology, Zhengzhou 450001, China
| | - Chunhua Zhu
- Key Laboratory of Food Information Processing and Control, Ministry of Education, Henan University of Technology, Zhengzhou 450001, China; (H.T.); (S.S.); (H.F.); (C.Z.)
- Henan Engineering Laboratory of Grain IOT Technology, Henan University of Technology, Zhengzhou 450001, China
| | - Boye Liu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China
| |
Collapse
|
35
|
Nwazojie CC, Obayemi JD, Salifu AA, Borbor-Sawyer SM, Uzonwanne VO, Onyekanne CE, Akpan UM, Onwudiwe KC, Oparah JC, Odusanya OS, Soboyejo WO. Targeted drug-loaded PLGA-PCL microspheres for specific and localized treatment of triple negative breast cancer. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:41. [PMID: 37530973 PMCID: PMC10397127 DOI: 10.1007/s10856-023-06738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 07/01/2023] [Indexed: 08/03/2023]
Abstract
The paper presents the results of the experimental and analytical study of targeted drug-loaded polymer-based microspheres made from blend polymer of polylactic-co-glycolic acid and polycaprolactone (PLGA-PCL) for targeted and localized cancer drug delivery. In vitro sustained release with detailed thermodynamically driven drug release kinetics, over a period of three months using encapsulated targeted drugs (prodigiosin-EphA2 or paclitaxel-EphA2) and control drugs [Prodigiosin (PGS), and paclitaxel (PTX)] were studied. Results from in vitro study showed a sustained and localized drug release that is well-characterized by non-Fickian Korsmeyer-Peppas kinetics model over the range of temperatures of 37 °C (body temperature), 41 °C, and 44 °C (hyperthermic temperatures). The in vitro alamar blue, and flow cytometry assays in the presence of the different drug-loaded polymer formulations resulted to cell death and cytotoxicity that was evidence through cell inhibition and late apoptosis on triple negative breast cancer (TNBC) cells (MDA-MB 231). In vivo studies carried out on groups of 4-week-old athymic nude mice that were induced with subcutaneous TNBC, showed that the localized release of the EphA2-conjugated drugs was effective in complete elimination of residual tumor after local surgical resection. Finally, ex vivo histopathological analysis carried out on the euthanized mice revealed no cytotoxicity and absence of breast cancer metastases in the liver, kidney, and lungs 12 weeks after treatment. The implications of the results are then discussed for the development of encapsulated EphA2-conjugated drugs formulation in the specific targeting, localized, and sustain drug release for the elimination of local recurred TNBC tumors after surgical resection.
Collapse
Affiliation(s)
- Chukwudalu C Nwazojie
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - John D Obayemi
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01605, USA
| | - Ali A Salifu
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01605, USA
- Department of Engineering, Boston College, 140 Commonwealth Avenue, Chestnut Hill, USA
| | - Sandra M Borbor-Sawyer
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
- Department of Biology, State University of New York, Buffalo State University, Buffalo, USA
| | - Vanessa O Uzonwanne
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
- Department of Engineering, Boston College, 140 Commonwealth Avenue, Chestnut Hill, USA
| | - Chinyerem E Onyekanne
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Udom M Akpan
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Killian C Onwudiwe
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Josephine C Oparah
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Olushola S Odusanya
- Biotechnology and Genetic Engineering Advanced Laboratory, Sheda Science and Technology Complex (SHESTCO), Abuja, Nigeria
| | - Winston O Soboyejo
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria.
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA.
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01605, USA.
| |
Collapse
|
36
|
Taghipour MJ, Ezzatpanah H, Ghahderijani M. In vitro and in silico studies for the identification of anti-cancer and antibacterial peptides from camel milk protein hydrolysates. PLoS One 2023; 18:e0288260. [PMID: 37437001 DOI: 10.1371/journal.pone.0288260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023] Open
Abstract
Today, breast cancer and infectious diseases are very worrying that led to a widespread effort by researchers to discover natural remedies with no side effects to fight them. In the present study, we isolated camel milk protein fractions, casein and whey proteins, and hydrolyzed them using pepsin, trypsin, and both enzymes. Screening of peptides with anti-breast cancer and antibacterial activity against pathogens was performed. Peptides derived from whey protein fraction with the use of both enzymes showed very good activity against MCF-7 breast cancer with cell viability of 7.13%. The separate use of trypsin and pepsin to digest whey protein fraction yielded peptides with high antibacterial activity against S. aureus (inhibition zone of 4.17 ± 0.30 and 4.23 ± 0.32 cm, respectively) and E. coli (inhibition zone of 4.03 ± 0.15 and 4.03 ± 0.05 cm, respectively). Notably, in order to identify the effective peptides in camel milk, its protein sequences were retrieved and enzymatically digested in silico. Peptides that showed both anticancer and antibacterial properties and the highest stability in intestinal conditions were selected for the next step. Molecular interaction analysis was performed on specific receptors associated with breast cancer and/or antibacterial activity using molecular docking. The results showed that P3 (WNHIKRYF) and P5 (WSVGH) peptides had low binding energy and inhibition constant so that they specifically occupied active sites of protein targets. Our results introduced two peptide-drug candidates and new natural food additive that can be delivered to further animal and clinical trials.
Collapse
Affiliation(s)
- Mohammad Javad Taghipour
- Department of Food Science and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Ezzatpanah
- Department of Food Science and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Ghahderijani
- Department of Agricultural Systems Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
37
|
Muhammad Rehman H, Rehman HM, Naveed M, Khan MT, Shabbir MA, Aslam S, Bashir H. In Silico Investigation of a Chimeric IL24-LK6 Fusion Protein as a Potent Candidate Against Breast Cancer. Bioinform Biol Insights 2023; 17:11779322231182560. [PMID: 37377793 PMCID: PMC10291407 DOI: 10.1177/11779322231182560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted delivery of therapeutic anticancer chimeric molecules enhances the efficacy of drug by improving cellular uptake and circulation time. Engineering the molecules to facilitate the specific interaction between chimeric protein and its receptor is critical to elucidate biological mechanism as well as accuracy in modeling of complexes. A theoretically designed novel protein-protein interfaces can serve as a bottom-up method for comprehensive understanding of interacting protein residues. This study was aimed for in silico analyses of a chimeric fusion protein against breast cancer. The amino acid sequences of the interleukin 24 (IL-24) and LK-6 peptide were used to design the chimeric fusion protein via a rigid linker. The secondary and tertiary structures along with physicochemical properties by ProtParam and solubility were predicted using online software. The validation and quality of the fusion protein was confirmed by Rampage and ERRAT2. The newly designed fusion construct has a total length of 179 amino acids. The top-ranked structure from alpha fold2 showed 18.1 KD molecular weight by ProtParam, quality factor of 94.152 by ERRAT, and a valid structure by a Ramachandran plot with 88.5% residues in the favored region. Finally, the docking and simulation studies were performed using HADDOCK and Desmond module of Schrodinger. The quality, validity, interaction analysis, and stability of the fusion protein depict a functional molecule. The fusion gene IL24-LK6 after cloning and expression in a suitable prokaryotic cell might be a useful candidate for developing a novel anticancer therapy.
Collapse
Affiliation(s)
- Hafiz Muhammad Rehman
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
- University Institute of Medical Lab Technology, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Hafiz Muzzammel Rehman
- School of Biochemistry & Biotechnology, University of the Punjab, Lahore, Pakistan
- Department of Human Genetics and Molecular Biology, University of Health Sciences, Lahore, Pakistan
| | - Muhammad Naveed
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Muhammad Tahir Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Muhammad Aqib Shabbir
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Shakira Aslam
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Hamid Bashir
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
38
|
Dempsey MP, Andersen KE, Wells BM, Taylor MA, Cashman CL, Conrad LB, Kearney CA, Conklin MB, Via ER, Doe EM, Komirisetty R, Dearborn S, Reddy ST, Farias-Eisner R. Stability Characterization of the Novel Anti-Cancer HM-10/10 HDL-Mimetic Peptide. Int J Mol Sci 2023; 24:10054. [PMID: 37373203 PMCID: PMC10298982 DOI: 10.3390/ijms241210054] [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: 04/15/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Epithelial adenocarcinoma of the ovary and colon are associated with the highest rates of cancer-related deaths in women in the U.S. The literature supports the role of HDL-associated apolipoproteins in the treatment of cancer and other pro-inflammatory diseases. Previously, we developed a novel 20-amino acid mimetic peptide, HM-10/10, which potently inhibits tumor development and growth in colon and ovarian cancer. Here, we report the properties of HM-10/10 relative to its stability in vitro. The results demonstrated that HM-10/10 had the highest half-life in human plasma compared to plasma from other species tested. HM-10/10 demonstrated stability in human plasma and simulated gastric environment, increasing its promise as an oral pharmaceutical. However, under conditions modeling the small intestine, HM-10/10 demonstrated significant degradation, likely due to the peptidases encountered therein. Furthermore, HM-10/10 demonstrated no evidence of time-dependent drug-drug interactions, although it demonstrated CYP450 induction slightly above cutoff. As proteolytic degradation is a common limitation of peptide-based therapeutics, we are pursuing strategies to improve the stability properties of HM-10/10 by extending its bioavailability while retaining its low toxicity profile. HM-10/10 holds promise as a new agent to address the international women's health crisis of epithelial carcinomas of the ovary and colon.
Collapse
Affiliation(s)
- Michael P. Dempsey
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
- David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Katelyn E. Andersen
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
- David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Brittney M. Wells
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Mitchell A. Taylor
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Clay L. Cashman
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Lesley B. Conrad
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
- David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA
- Lynch Comprehensive Cancer Research Center, School of Medicine, Creighton University, Omaha, NE 68178, USA
- Office of the President, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Claire A. Kearney
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Mary B. Conklin
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Emily R. Via
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Emily M. Doe
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
| | - Ravikiran Komirisetty
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.K.); (S.T.R.)
| | - Susan Dearborn
- Charles River Laboratories International, Stone Ridge, NY 12484, USA;
| | - Srinivasa T. Reddy
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.K.); (S.T.R.)
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, The University of California at Los Angeles, Los Angeles, CA 90095, USA
- Interdepartmental Program in Molecular Toxicology, School of Public Health, University of California at Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Robin Farias-Eisner
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (M.P.D.); (K.E.A.); (B.M.W.); (M.A.T.); (C.L.C.); (L.B.C.); (C.A.K.); (M.B.C.); (E.R.V.); (E.M.D.)
- David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90095, USA
- Lynch Comprehensive Cancer Research Center, School of Medicine, Creighton University, Omaha, NE 68178, USA
- Office of the President, Western University of Health Sciences, Pomona, CA 91766, USA
- Office of the President, Western University of Health Sciences, Lebanon, OR 97355, USA
| |
Collapse
|
39
|
Nikolaev B, Yakovleva L, Fedorov V, Yudintceva N, Ryzhov V, Marchenko Y, Ischenko A, Zhakhov A, Dobrodumov A, Combs SE, Gao H, Shevtsov M. Magnetic Relaxation Switching Assay Using IFNα-2b-Conjugated Superparamagnetic Nanoparticles for Anti-Interferon Antibody Detection. BIOSENSORS 2023; 13:624. [PMID: 37366989 DOI: 10.3390/bios13060624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/21/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
Type I interferons, particularly IFNα-2b, play essential roles in eliciting adaptive and innate immune responses, being implicated in the pathogenesis of various diseases, including cancer, and autoimmune and infectious diseases. Therefore, the development of a highly sensitive platform for analysis of either IFNα-2b or anti-IFNα-2b antibodies is of high importance to improve the diagnosis of various pathologies associated with the IFNα-2b disbalance. For evaluation of the anti-IFNα-2b antibody level, we have synthesized superparamagnetic iron oxide nanoparticles (SPIONs) coupled with the recombinant human IFNα-2b protein (SPIONs@IFNα-2b). Employing a magnetic relaxation switching assay (MRSw)-based nanosensor, we detected picomolar concentrations (0.36 pg/mL) of anti-INFα-2b antibodies. The high sensitivity of the real-time antibodies' detection was ensured by the specificity of immune responses and the maintenance of resonance conditions for water spins by choosing a high-frequency filling of short radio-frequency pulses of the generator. The formation of a complex of the SPIONs@IFNα-2b nanoparticles with the anti-INFα-2b antibodies led to a cascade process of the formation of nanoparticle clusters, which was further enhanced by exposure to a strong (7.1 T) homogenous magnetic field. Obtained magnetic conjugates exhibited high negative MR contrast-enhancing properties (as shown by NMR studies) that were also preserved when particles were administered in vivo. Thus, we observed a 1.2-fold decrease of the T2 relaxation time in the liver following administration of magnetic conjugates as compared to the control. In conclusion, the developed MRSw assay based on SPIONs@IFNα-2b nanoparticles represents an alternative immunological probe for the estimation of anti-IFNα-2b antibodies that could be further employed in clinical studies.
Collapse
Affiliation(s)
- Boris Nikolaev
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
| | - Ludmila Yakovleva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
| | - Viacheslav Fedorov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
- Department of Inorganic Chemistry and Biophysics, Saint-Petersburg State University of Veterinary Medicine, Chernigovskaya Str. 5, 196084 St. Petersburg, Russia
| | - Natalia Yudintceva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Vyacheslav Ryzhov
- Petersburg Nuclear Physics Institute, National Research Centre "Kurchatov Institute", 188300 Gatchina, Russia
| | - Yaroslav Marchenko
- Petersburg Nuclear Physics Institute, National Research Centre "Kurchatov Institute", 188300 Gatchina, Russia
| | - Alexander Ischenko
- Laboratory of Hybridoma Technologies, Saint-Petersburg Pasteur Institute, Mira Str. 14, 197101 St. Petersburg, Russia
| | - Alexander Zhakhov
- Laboratory of Hybridoma Technologies, Saint-Petersburg Pasteur Institute, Mira Str. 14, 197101 St. Petersburg, Russia
| | - Anatoliy Dobrodumov
- Department of Nuclear Magnetic Resonance, Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), Bolshoi pr. 31, 199004 St. Petersburg, Russia
| | - Stephanie E Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
- Laboratory of Biomedical Cell Technologies, Far Eastern Federal University, 690091 Vladivostok, Russia
| |
Collapse
|
40
|
Erlista GP, Ahmed N, Swasono RT, Raharjo S, Raharjo TJ. Proteome of monocled cobra ( Naja kaouthia) venom and potent anti breast cancer peptide from trypsin hydrolyzate of the venom protein. Saudi Pharm J 2023; 31:1115-1124. [PMID: 37293380 PMCID: PMC10244474 DOI: 10.1016/j.jsps.2023.04.001] [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: 09/24/2022] [Accepted: 04/01/2023] [Indexed: 06/10/2023] Open
Abstract
Anticancer peptide is one of the target in the development of new anticancer drug. Bioactive peptide can be originated from isolated free peptide or produced by hydrolysis of protein. Protein is the main component of Naja kaouthia venom, and due to the toxicity of the venom, it can be assessed as the source of anticancer peptides. This study aims to characterize the venom protein and to identify peptides from the snake venom of N. kaouthia as anticancer. Proteome analysis was employed trypsin hydrolysis of N. kaouthia venom protein completed with HRMS analysis protein database query. Preparative tryptic hydrolysis of the protein followed by reverse-phased fractionation and anti breast cancer activity testing were performed to identify the potent anticancer from the hydrolysate. Proteomic analysis by high-resolution mass spectrometry revealed that there are 20 enzymatic and non-enzymatic proteins in N. kaouthia venom. The 25% methanol peptide fraction had the most active anticancer activity against MCF-7 breast cancer cells and showed promising selectivity (selectivity index = 12.87). Amino acid sequences of eight peptides were identified as potentially providing anticancer compounds. Molecular docking analysis showed that WWSDHR and IWDTIEK peptides gave specific interactions and better binding affinity energy with values of -9.3 kcal/mol and -8.4 kcal/mol, respectively. This study revealed peptides from the snake venom of N. kaouthia became a potent source of new anticancer agents.
Collapse
Affiliation(s)
- Garnis Putri Erlista
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Bulaksumur, Yogyakarta 55281, Indonesia
| | - Naseer Ahmed
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Bulaksumur, Yogyakarta 55281, Indonesia
| | - Respati Tri Swasono
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Bulaksumur, Yogyakarta 55281, Indonesia
| | - Slamet Raharjo
- Department Internal Medicine, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Bulaksumur, Yogyakarta 55281, Indonesia
| | - Tri Joko Raharjo
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Bulaksumur, Yogyakarta 55281, Indonesia
| |
Collapse
|
41
|
Novel antiproliferative inhibitors from salicylamide derivatives with dipeptide moieties using 3D-QSAR, molecular docking, molecular dynamic simulation and ADMET studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
|
42
|
Barman P, Joshi S, Sharma S, Preet S, Sharma S, Saini A. Strategic Approaches to Improvise Peptide Drugs as Next Generation Therapeutics. Int J Pept Res Ther 2023; 29:61. [PMID: 37251528 PMCID: PMC10206374 DOI: 10.1007/s10989-023-10524-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 05/31/2023]
Abstract
In recent years, the occurrence of a wide variety of drug-resistant diseases has led to an increase in interest in alternate therapies. Peptide-based drugs as an alternate therapy hold researchers' attention in various therapeutic fields such as neurology, dermatology, oncology, metabolic diseases, etc. Previously, they had been overlooked by pharmaceutical companies due to certain limitations such as proteolytic degradation, poor membrane permeability, low oral bioavailability, shorter half-life, and poor target specificity. Over the last two decades, these limitations have been countered by introducing various modification strategies such as backbone and side-chain modifications, amino acid substitution, etc. which improve their functionality. This has led to a substantial interest of researchers and pharmaceutical companies, moving the next generation of these therapeutics from fundamental research to the market. Various chemical and computational approaches are aiding the production of more stable and long-lasting peptides guiding the formulation of novel and advanced therapeutic agents. However, there is not a single article that talks about various peptide design approaches i.e., in-silico and in-vitro along with their applications and strategies to improve their efficacy. In this review, we try to bring different aspects of peptide-based therapeutics under one article with a clear focus to cover the missing links in the literature. This review draws emphasis on various in-silico approaches and modification-based peptide design strategies. It also highlights the recent progress made in peptide delivery methods important for their enhanced clinical efficacy. The article would provide a bird's-eye view to researchers aiming to develop peptides with therapeutic applications. Graphical Abstract
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Shubhi Joshi
- Energy Research Centre, Panjab University, Sector 14, Chandigarh, 160014 India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Simran Preet
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Shweta Sharma
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| |
Collapse
|
43
|
Bloom SMK, O’Hare N, Forbes NS. Bacterial delivery of therapeutic proteins to the nuclei of cancer cells. Biotechnol Bioeng 2023; 120:1437-1448. [PMID: 36710503 PMCID: PMC10101893 DOI: 10.1002/bit.28340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Targeting nucleic targets with therapeutic proteins would enhance the treatment of hard-to-treat cancers. However, exogenous proteins are excluded from the nucleus by both the cellular and nuclear membranes. We have recently developed Salmonella that deliver active proteins into the cytoplasm of cancer cells. Here, we hypothesized that bacterially delivered proteins accumulate within nuclei, nuclear localization sequences (NLSs) increase delivery, and bacterially delivered proteins kill cancer cells. To test this hypothesis, we developed intranuclear delivering (IND) Salmonella and quantified the delivery of three model proteins. IND Salmonella delivered both ovalbumin and green fluorescent protein to nuclei of MCF7 cancer cells. The amount of protein in nuclei was linearly dependent on the amount delivered to the cytoplasm. The addition of a NLSs increased both the amount of protein in each nucleus and the number of nuclei that received protein. Delivery of Omomyc, a protein inhibitor of the nuclear transcript factor, Myc, altered cell physiology, and significantly induced cell death. These results show that IND Salmonella deliver functional proteins to the nucleus of cancerous cells. Extending this method to other transcription factors will increase the number of accessible targets for cancer therapy.
Collapse
Affiliation(s)
| | - Nicholas O’Hare
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|
44
|
Liu Y, Chang R, Xing R, Yan X. Bioactive Peptide Nanodrugs Based on Supramolecular Assembly for Boosting Immunogenic Cell Death-Induced Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201708. [PMID: 36720041 DOI: 10.1002/smtd.202201708] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/12/2023] [Indexed: 05/17/2023]
Abstract
Immunogenic cell death (ICD)-induced immunotherapy holds promise for complete elimination and long-term protective immune responses against cancer by combining direct tumor cell killing and antitumor immune response. Some therapeutic approaches (such as hyperthermia, photodynamic therapy, or radiotherapy) and inducers (certain chemotherapy drugs, oncolytic viruses) have been devoted to initiating and/or boosting ICD, leading to the activation of tumor-specific immune responses. Recently, supramolecular assembled bioactive peptide nanodrugs have been employed to improve the efficacy of ICD-induced cancer immunotherapy by increasing tumor targeted accumulation as well as responsive release of ICD inducers, directly inducing high levels of ICD and realizing the simultaneous enhancement of immune response through the immune function of the active peptide itself. Here, the authors review bioactive peptide nanodrugs based on supramolecular assembly, mainly as an intelligent delivery system, a direct ICD inducer and an immune response enhancer, for boosting ICD induced cancer immunotherapy. The functions of diverse bioactive peptides used in the construction of nanodrugs are described. The design of a supramolecular assembly, the mechanism of boosting ICD, and synergetic effects of bioactive peptides combined immunotherapy are critically emphasized.
Collapse
Affiliation(s)
- Yamei Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Rui Chang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ruirui Xing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- Center for Mesoscience, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
45
|
Azari M, Bahreini F, Uversky VN, Rezaei N. Current therapeutic approaches and promising perspectives of using bioengineered peptides in fighting chemoresistance in triple-negative breast cancer. Biochem Pharmacol 2023; 210:115459. [PMID: 36813121 DOI: 10.1016/j.bcp.2023.115459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023]
Abstract
Breast cancer is a collation of malignancies that manifest in the mammary glands at the early stages. Among breast cancer subtypes, triple-negative breast cancer (TNBC) shows the most aggressive behavior, with apparent stemness features. Owing to the lack of response to hormone therapy and specific targeted therapies, chemotherapy remains the first line of the TNBC treatment. However, the acquisition of resistance to chemotherapeutic agents increase therapy failure, and promotes cancer recurrence and distant metastasis. Invasive primary tumors are the birthplace of cancer burden, though metastasis is a key attribute of TNBC-associated morbidity and mortality. Targeting the chemoresistant metastases-initiating cells via specific therapeutic agents with affinity to the upregulated molecular targets is a promising step in the TNBC clinical management. Exploring the capacity of peptides as biocompatible entities with the specificity of action, low immunogenicity, and robust efficacy provides a principle for designing peptide-based drugs capable of increasing the efficacy of current chemotherapy agents for selective targeting of the drug-tolerant TNBC cells. Here, we first focus on the resistance mechanisms that TNBC cells acquire to evade the effect of chemotherapeutic agents. Next, the novel therapeutic approaches employing tumor-targeting peptides to exploit the mechanisms of drug resistance in chemorefractory TNBC are described.
Collapse
Affiliation(s)
- Mandana Azari
- School of Chemical Engineering-Biotechnology, College of Engineering, University of Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Bahreini
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Gareev K, Tagaeva R, Bobkov D, Yudintceva N, Goncharova D, Combs SE, Ten A, Samochernych K, Shevtsov M. Passing of Nanocarriers across the Histohematic Barriers: Current Approaches for Tumor Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1140. [PMID: 37049234 PMCID: PMC10096980 DOI: 10.3390/nano13071140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Over the past several decades, nanocarriers have demonstrated diagnostic and therapeutic (i.e., theranostic) potencies in translational oncology, and some agents have been further translated into clinical trials. However, the practical application of nanoparticle-based medicine in living organisms is limited by physiological barriers (blood-tissue barriers), which significantly hampers the transport of nanoparticles from the blood into the tumor tissue. This review focuses on several approaches that facilitate the translocation of nanoparticles across blood-tissue barriers (BTBs) to efficiently accumulate in the tumor. To overcome the challenge of BTBs, several methods have been proposed, including the functionalization of particle surfaces with cell-penetrating peptides (e.g., TAT, SynB1, penetratin, R8, RGD, angiopep-2), which increases the passing of particles across tissue barriers. Another promising strategy could be based either on the application of various chemical agents (e.g., efflux pump inhibitors, disruptors of tight junctions, etc.) or physical methods (e.g., magnetic field, electroporation, photoacoustic cavitation, etc.), which have been shown to further increase the permeability of barriers.
Collapse
Affiliation(s)
- Kamil Gareev
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia
| | - Ruslana Tagaeva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Danila Bobkov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Daria Goncharova
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
| | - Artem Ten
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| |
Collapse
|
47
|
Li L, Liu C, Qin Y, Gao F, Wang Q, Zhu Y. Identification of cancer protein biomarker based on cell specific peptide and its potential role in predicting tumor metastasis. J Proteomics 2023; 275:104826. [PMID: 36708809 DOI: 10.1016/j.jprot.2023.104826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 11/19/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023]
Abstract
The identification of tumor related membrane protein is important for both cancer diagnosis and targeted therapy. Currently, the number of ideal clinical biomarkers is still limited partially because of lacking efficient methods in biomarker discovery. Targeting peptides are generated by library screening and can recognize their cognate targets with high specificity and affinity. In addition, these functional peptides have been considered to be a valuable molecule for both imaging detection and targeting therapy in oncology. The selected peptides can be used to identify cell-surface protein biomarkers of cancer cells. In our study, the peptide (VECYLIRDNLCIY) derived from the bacteria displaying library worked as a bait to capture its binding partner and aldolase A was identified as the candidate. The results indicated that aldolase A' expression level on the cell membrane was regulated by PI3K and aldolase A located on the membrane could inhibit the aggression of tumors through mediating cell metabolic pathway. Aldolase A could work as the joint for the metabolic and signal pathways related to tumor progression. In our work, we demonstrated a promising technology for selecting and identifying binding partners for cell-specific peptides that enables discovery of new tumor biomarkers, showing great scientific study values and clinical translation potencies. SIGNIFICANCE: MS-based cancer biomarker discovery provides promising target candidates for cancer diagnosis and therapy. However, the inevitable limits make it inconvenient in the process of sample preparation and data analysis. In this way, the small molecular probes show some advantages due to their readily availability and specific binding affinity such as the aptamers screened with SELEX technology and peptides derived from displaying libraries. In the present study, aldolase A was proved to be the membrane binding partner of a specific peptidic ligand towards ZR-75-1 tumor cell. It was discovered that membrane aldolase A was more sensitive and observable than other subcellular fractions in response to cellular metabolic state alteration or glucose availability. In addition, the reduced membrane-localized aldolase A expression indicated a more aggressive tumor phenotype and was accompanied by the upregulation of MMP-2/MMP-9. The non-glycolysis activity endowed it with potential utility as a tumor diagnostic marker and therapeutic target. This work demonstrates the practicability of screened peptide in cancer biomarker discovery, facilitating the development of diagnostic tools and therapeutic approaches to cancer, and markedly improves our understanding of cancer biology.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Cuijuan Liu
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Yingzhou Qin
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Fan Gao
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Qianqian Wang
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Yimin Zhu
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
48
|
Nieland L, Mahjoum S, Grandell E, Breyne K, Breakefield XO. Engineered EVs designed to target diseases of the CNS. J Control Release 2023; 356:493-506. [PMID: 36907561 DOI: 10.1016/j.jconrel.2023.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/28/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
Diseases of the central nervous system (CNS) are challenging to treat, mainly due to the blood-brain barrier (BBB), which restricts drugs in circulation from entering target regions in the brain. To address this issue extracellular vesicles (EVs) have gained increasing scientific interest as carriers able to cross the BBB with multiplex cargos. EVs are secreted by virtually every cell, and their escorted biomolecules are part of an intercellular information gateway between cells within the brain and with other organs. Scientists have undertaken efforts to safeguard the inherent features of EVs as therapeutic delivery vehicles, such as protecting and transferring functional cargo, as well as loading them with therapeutic small molecules, proteins, and oligonucleotides and targeting them to specific cell types for the treatment of CNS diseases. Here, we review current emerging approaches that engineer the EV surface and cargo to improve targeting and functional responses in the brain. We summarize existing applications of engineered EVs as a therapeutic delivery platform for brain diseases, some of which have been evaluated clinically.
Collapse
Affiliation(s)
- Lisa Nieland
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery, Leiden University Medical Center, Leiden 2300 RC, the Netherlands.
| | - Shadi Mahjoum
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Emily Grandell
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA; Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Koen Breyne
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Xandra O Breakefield
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
49
|
Tavakoli S, Firoozpour L, Davoodi J. The synergistic effect of chimeras consisting of N-terminal smac and modified KLA peptides in inducing apoptosis in breast cancer cell lines. Biochem Biophys Res Commun 2023; 655:138-144. [PMID: 36934589 DOI: 10.1016/j.bbrc.2023.03.008] [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: 02/10/2023] [Revised: 02/26/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Drug resistance is one of the most important obstacles in effective cancer therapy triggered through various mechanisms. One of these mechanisms is caused by the upregulation of Inhibitor of Apoptosis Proteins (IAPs). IAPs, inhibit apoptosis through direct and/or indirect caspase inhibition, which themselves are antagonized by an endogenous protein called Second Mitochondrial-derived Activator of Caspases, Smac/Diablo, mediated by the presence of a tetrapeptide IAP binding motif at its N-terminus. Accordingly, Smac-based peptides are under intense investigation as anti-cancer drugs and have reached Phase 2 clinical trials, although, Smac based peptides or mimetics alone have not been effective as anti-cancer agents. On the other hand, KLA peptide has shown major toxicity against cancer cells through the induction of apoptosis. Consequently, we designed an anti-cancer chimera by fusing an octa-peptide from the N-terminus of mature Smac protein to a modified proapoptotic KLA peptide (KLAKLCKKLAKLCK) to be called Smac-KLA. This chimera, therefore, possesses both proapoptotic and anti-IAP activities. In addition, we dimerized this chimera via intermolecular disulfide bonds in order to enhance their cellular permeability. Both the Smac-KLA monomeric and dimeric peptides exhibited cytotoxic activity against both MCF-7 and MDA-MB231 breast cancer cell lines at low micromolar concentrations. Importantly, the dimerization of the chimeras enhanced their potency 2-4- fold due to higher cellular uptake.
Collapse
Affiliation(s)
- Somayeh Tavakoli
- Institute of Biochemistry and Biophysics, University of Tehran, Postal code: 1417614335, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Davoodi
- Institute of Biochemistry and Biophysics, University of Tehran, Postal code: 1417614335, Tehran, Iran.
| |
Collapse
|
50
|
Singh NK, Agarwal M, Radhakrishna M. Understanding the helical stability of charged peptides. Proteins 2023; 91:268-276. [PMID: 36121161 DOI: 10.1002/prot.26427] [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: 07/05/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023]
Abstract
Cationic helical peptides play a crucial role in applications such as anti-microbial and anticancer activity. The activity of these peptides directly correlates with their helicity. In this study, we have performed extensive all-atom molecular dynamics simulations of 25 Lysine-Leucine co-polypeptide sequences of varying charge density ( λ ) and patterns. Our findings showed that, an increase in the charge density on the peptide leads to a gradual decrease in the helicity up to a critical charge density λ c . Beyond λ c , a complete helix to coil transition was observed. The decrease in the helicity is correlated with the increased number of water molecules in first solvation shell, solvent-exposed surface area, and a higher value of the radius of gyration of the peptide.
Collapse
Affiliation(s)
- Nitin Kumar Singh
- Discipline of Chemical Engineering, Indian Institute of Technology (IIT), Gandhinagar, Gujarat, India
| | - Manish Agarwal
- Computer Services Centre, Indian Institute of Technology (IIT), Delhi, India
| | - Mithun Radhakrishna
- Discipline of Chemical Engineering, Indian Institute of Technology (IIT), Gandhinagar, Gujarat, India.,Center for Biomedical Engineering, Indian Institute of Technology (IIT), Gandhinagar, Gujarat, India
| |
Collapse
|