1
|
Yang L, Haeckel A, Beindorff N, Poetzsch SML, Mi H, Ni F, Hojjat H, Brenner W, de Moraes PAD, Guo J, Savic LJ, Schellenberger E. Long circulating XTEN864-HGV-Apoptin fusion protein for selective cancer therapy. Int J Biol Macromol 2025; 306:141679. [PMID: 40037452 DOI: 10.1016/j.ijbiomac.2025.141679] [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: 10/25/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
The virus protein CAV-Apoptin and its homologue HGV-Apoptin selectively kill cancer cells but are not suitable for systemic treatment. The aim was to develop Apoptin-based fusion proteins for intravenous application in cancer therapy, which also contain the hydrophilic polypeptide XTEN, a cleavage site for MMP-2/9, and a TAT peptide for cell penetration. Expression of XTEN864-HGV-Apoptin in E. coli and purification using XTEN as a tag yielded 100 mg protein/L tissue culture. The expression of XTEN864-CAV-Apoptin did not generate a sufficient yield. Cytotoxic effects were assessed using MTT and Annexin A5 assays, whereas cellular uptake was visualized using Cy3.5-XTEN864-HGV-Apoptin. Blood half-life and biodistribution were evaluated with 99mTc-XTEN864-HGV-Apoptin using SPECT-CT and gamma counting. The fusion protein significantly reduced cancer cell growth and induced apoptosis with minimal effects on non-cancerous cells. It accumulates in the nucleus and associates with F-actin. In mice, the protein showed a blood half-life of 0.68 h (fast phase) and 17 h (slow phase), with a tumor/muscle ratio of 9.36 ± 6.22 (SD). In a 4T1 mouse tumor model, it effectively inhibited tumor growth. The cancer-specific cytotoxicity and prolonged circulation of XTEN864-HGV-Apoptin suggest its potential for systemically applicable, biodegradable, and E. coli-producible antitumor drugs.
Collapse
Affiliation(s)
- Liu Yang
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Akvile Haeckel
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicola Beindorff
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Simon Marc Levin Poetzsch
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Honglan Mi
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Fei Ni
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Hamidreza Hojjat
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Winfried Brenner
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany; Charité - Universitätsmedizin Berlin, Department of Nuclear Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Pedro Augusto Dantas de Moraes
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Jing Guo
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Lynn Jeanette Savic
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eyk Schellenberger
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
2
|
Tabandeh F, Moradian Tehrani R, Sharifi M, Toopchi E. Investigating therapeutic efficacy of dacarbazine and temozolomide, alone and in combination with BRAF (V600E) siRNA in A375 human melanoma cell line. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:772-783. [PMID: 40343301 PMCID: PMC12057749 DOI: 10.22038/ijbms.2025.84187.18208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/31/2024] [Indexed: 05/11/2025]
Abstract
Objectives Melanoma is one of the most aggressive and deadly skin cancers. Despite advances, effective melanoma treatment is challenging, often requiring a shift from individual therapies to combination approaches. This study explores whether combining dacarbazine (DTIC) and temozolomide (TMZ) with the siRNA approach holds promise for melanoma treatment. Materials and Methods To determine the IC50 values of DTIC and TMZ, the A375 cell line was treated with different drug concentrations for 24-72 hr. The best exposure time of BRAF siRNA transfection was performed. Subsequently, cell viability (using the MTT assay), apoptosis (by flow cytometry), and gene expression levels of B-Raf proto-oncogene, serine/threonine kinase (BRAF), caspase 3 (CASP3), and phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) genes (by quantitative real-time PCR) were assessed in the treated groups (i.e., control, negative controls, DTIC alone, TMZalone, DTIC+ TMZ, BRAF(V600E)siRNA alone, siRNA+ DTIC, siRNA+ TMZ, and siRNA+ DTIC+ TMZ groups). Results Cell viability significantly decreased in the chemotherapy-only and siRNA+drug groups, although no difference was observed between them. The apoptosis percentage in all treated groups indicated a significant difference compared to the control group. The expression of the BRAF gene notably decreased in the BRAF (V600E) siRNA +drug groups compared to the chemotherapy groups. Despite overexpression of CASP3 in the chemotherapy-treated groups, the most effective enhancement was noted in the siRNA+DTIC+TMZ group (P<0.0001). The mean expression of the PIK3R3 gene in siRNA+chemotherapy groups revealed a notable reduction. Conclusion These findings suggest that the siRNA-transfected treatment groups have the potential to provide therapeutic effects comparable to those of chemotherapy.
Collapse
Affiliation(s)
- Fatemeh Tabandeh
- Medical Biotechnology. Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rana Moradian Tehrani
- Department of Genetics and Molecular Biology. School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology. School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elmira Toopchi
- Department of Genetics and Molecular Biology. School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
3
|
Moreno-Vargas LM, Prada-Gracia D. Exploring the Chemical Features and Biomedical Relevance of Cell-Penetrating Peptides. Int J Mol Sci 2024; 26:59. [PMID: 39795918 PMCID: PMC11720145 DOI: 10.3390/ijms26010059] [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: 10/23/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/13/2025] Open
Abstract
Cell-penetrating peptides (CPPs) are a diverse group of peptides, typically composed of 4 to 40 amino acids, known for their unique ability to transport a wide range of substances-such as small molecules, plasmid DNA, small interfering RNA, proteins, viruses, and nanoparticles-across cellular membranes while preserving the integrity of the cargo. CPPs exhibit passive and non-selective behavior, often requiring functionalization or chemical modification to enhance their specificity and efficacy. The precise mechanisms governing the cellular uptake of CPPs remain ambiguous; however, electrostatic interactions between positively charged amino acids and negatively charged glycosaminoglycans on the membrane, particularly heparan sulfate proteoglycans, are considered the initial crucial step for CPP uptake. Clinical trials have highlighted the potential of CPPs in diagnosing and treating various diseases, including cancer, central nervous system disorders, eye disorders, and diabetes. This review provides a comprehensive overview of CPP classifications, potential applications, transduction mechanisms, and the most relevant algorithms to improve the accuracy and reliability of predictions in CPP development.
Collapse
|
4
|
Adhikary S, Roy S, Budhathoki S, Chowdhury S, Stillwell A, Basnakian AG, Tackett A, Avaritt N, Milad M, Alam MA. Thiazole-fused androstenone and ethisterone derivatives: potent β- and γ-actin cytoskeleton inhibitors to treat melanoma tumors. RSC Med Chem 2024; 16:d4md00719k. [PMID: 39703801 PMCID: PMC11653411 DOI: 10.1039/d4md00719k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
Melanoma, the most fatal form of skin cancer, often becomes resistant to the current therapeutic approaches in most patients. To explore new treatment options, fused thiazole derivatives were synthesized, and several of these compounds demonstrated potent anti-melanoma activity both in vitro and in vivo. These compounds exhibited significant cytotoxicity against melanoma cell lines at low concentrations. The lead molecules induced apoptosis and caused G2/M phase cell cycle arrest to a lesser extent. These compounds also displayed remarkable antimetastatic activities in several cell-based and molecular assays, significantly inhibiting key processes of metastasis, such as cell migration and adhesion. mRNA sequencing revealed significant downregulation of β-actin (ACTB) and γ-actin (ACTG1) at the transcriptional level, and a similar effect was observed at the protein level by western immunoblotting and proteomics assays. Actin-rich membrane protrusions formation is crucial for facilitating metastasis by promoting cell migration. Fluorescence microscopy demonstrated that compounds E28 and E47 inhibited the formation of these membrane protrusions and impaired actin cytoskeleton dynamics. Docking studies suggested the lead compounds may suppress tumor proliferation and metastasis by targeting the mechanistic target of Rapamycin complex 2 (mTORC2). All these findings unanimously indicated the translational perspective of ethisterone and androstenone fused thiazole derivatives as potent antimetastatic and antimelanoma agents. In a preclinical mouse melanoma model, compounds E2 and E47 significantly reduced tumor growth and greatly improved overall mice survival, while showing a favorable safety profile based on a comprehensive blood plasma metabolite profile. These lead molecules also displayed promising physicochemical properties, making them strong candidates for further drug development studies.
Collapse
Affiliation(s)
- Sanjay Adhikary
- Department of Chemistry and Physics, College of Sciences and Mathematics, Arkansas State University Jonesboro Arkansas 72467 USA
| | - Subrata Roy
- Department of Chemistry and Physics, College of Sciences and Mathematics, Arkansas State University Jonesboro Arkansas 72467 USA
- Enviromental Sciences Program, College of Sciences and Mathematics, Arkansas State University Jonesboro AR 72467 USA
| | - Shailesh Budhathoki
- Molecular Biosciences Program, College of Sciences and Mathematics, Arkansas State University Jonesboro AR 72467 USA
| | - Siam Chowdhury
- Department of Chemistry and Physics, College of Sciences and Mathematics, Arkansas State University Jonesboro Arkansas 72467 USA
- Computer Science, The College of Engineering and Computer Science, Arkansas State University Jonesboro AR 72468 USA
| | - Abbey Stillwell
- Department of Chemistry and Physics, College of Sciences and Mathematics, Arkansas State University Jonesboro Arkansas 72467 USA
| | - Alexei G Basnakian
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences 4301 W. Markham St Little Rock AR 72205 USA
- Central Arkansas Veterans Healthcare System W. 7th St Little Rock AR 72205 USA
| | - Alan Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Nathan Avaritt
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Mohamed Milad
- The Department of Mathematics and Statistics, Arkansas State University Jonesboro AR 72467 USA
| | - Mohammad Abrar Alam
- Department of Chemistry and Physics, College of Sciences and Mathematics, Arkansas State University Jonesboro Arkansas 72467 USA
- Enviromental Sciences Program, College of Sciences and Mathematics, Arkansas State University Jonesboro AR 72467 USA
- Molecular Biosciences Program, College of Sciences and Mathematics, Arkansas State University Jonesboro AR 72467 USA
- Arkansas Biosciences Institute, Arkansas State University Jonesboro AR 72467 USA
| |
Collapse
|
5
|
Yathindranath V, Safa N, Tomczyk MM, Dolinsky V, Miller DW. Lipid Nanoparticle-Based Inhibitors for SARS-CoV-2 Host Cell Infection. Int J Nanomedicine 2024; 19:3087-3108. [PMID: 38562613 PMCID: PMC10984206 DOI: 10.2147/ijn.s448005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Purpose The global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the lingering threat to public health has fueled the search for effective therapeutics to treat SARS-CoV-2. This study aimed to develop lipid nanoparticle (LNP) inhibitors of SARS-CoV-2 entry to reduce viral infection in the nose and upper airway. Methods Two types of LNP formulations were prepared following a microfluidic mixing method. The LNP-Trap consisted of DOPC, DSPC, cholesterol, and DSPE-PEG-COOH modified with various spike protein binding ligands, including ACE2 peptide, recombinant human ACE2 (rhACE2) or monoclonal antibody to spike protein (mAb). The LNP-Trim consisted of ionizing cationic DLin-MC3-DMA, DSPC, cholesterol, and DMG-PEG lipids encapsulating siACE2 or siTMPRSS2. Both formulations were assayed for biocompatibility and cell uptake in airway epithelial cells (Calu-3). Functional assessment of activity was performed using SARS-CoV-2 spike protein binding assays (LNP-Trap), host receptor knockdown (LNP-Trim), and SARS-CoV-2 pseudovirus neutralization assay (LNP-Trap and LNP-Trim). Localization and tissue distribution of fluorescently labeled LNP formulations were assessed in mice following intranasal administration. Results Both LNP formulations were biocompatible based on cell impedance and MTT cytotoxicity studies in Calu-3 cells at concentrations as high as 1 mg/mL. LNP-Trap formulations were able to bind spike protein and inhibit pseudovirus infection by 90% in Calu-3 cells. LNP-Trim formulations reduced ACE2 and TMPRSS2 at the mRNA (70% reduction) and protein level (50% reduction). The suppression of host targets in Calu-3 cells treated with LNP-Trim resulted in over 90% inhibition of pseudovirus infection. In vivo studies demonstrated substantial retention of LNP-Trap and LNP-Trim in the nasal cavity following nasal administration with minimal systemic exposure. Conclusion Both LNP-Trap and LNP-Trim formulations were able to safely and effectively inhibit SARS-CoV-2 pseudoviral infection in airway epithelial cells. These studies provide proof-of-principle for a localized treatment approach for SARS-CoV-2 in the upper airway.
Collapse
Affiliation(s)
- Vinith Yathindranath
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Centre, Health Science Centre, Winnipeg, MB, Canada
| | - Nura Safa
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Centre, Health Science Centre, Winnipeg, MB, Canada
| | - Mateusz Marek Tomczyk
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- Children’s Hospital Research Institute Manitoba, Health Science Centre, Winnipeg, MB, Canada
| | - Vernon Dolinsky
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- Children’s Hospital Research Institute Manitoba, Health Science Centre, Winnipeg, MB, Canada
| | - Donald W Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Centre, Health Science Centre, Winnipeg, MB, Canada
| |
Collapse
|
6
|
Mohapatra D, Kumar DN, Shreya S, Pandey V, Dubey PK, Agrawal AK, Sahu AN. Quality by design-based development and optimization of fourth-generation ternary solid dispersion of standardized Piper longum extract for melanoma therapy. Drug Deliv Transl Res 2023; 13:3094-3131. [PMID: 37294426 DOI: 10.1007/s13346-023-01375-y] [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] [Accepted: 05/25/2023] [Indexed: 06/10/2023]
Abstract
The study aimed to enhance the solubility, dissolution, and oral bioavailability of standardized Piper longum fruits ethanolic extract (PLFEE) via fourth-generation ternary solid dispersion (SD) for melanoma therapy. With the use of solvent evaporation method, the standardized PLFEE was formulated into SD, optimized using Box-Wilson's central composite design (CCD), and evaluated for pharmaceutical performance and in vivo anticancer activity against melanoma (B16F10)-bearing C57BL/6 mice. The optimized SD showed good accelerated stability, high yield, drug content, and content uniformity for bioactive marker piperine (PIP). The X-ray diffraction (XRD), differential scanning calorimetry (DSC), polarized light microscopy (PLM), and selected area electron diffraction (SAED) analysis revealed its amorphous nature. The attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR) and high-performance thin layer chromatography (HPTLC) revealed the compatibility of excipients with the PLFEE. The contact angle measurement and in vitro dissolution study revealed excellent wetting of SD and improved dissolution profile as compared to the plain PLFEE. The in vivo oral bioavailability of SD reflected a significant (p < 0.05) improvement in bioavailability (Frel = 188.765%) as compared to plain extract. The in vivo tumor regression study revealed the improved therapeutic activity of SD as compared to plain PLFEE. Further, the SD also improved the anticancer activity of dacarbazine (DTIC) as an adjuvant therapy. The overall result revealed the potential of developed SD for melanoma therapy either alone or as an adjuvant therapy with DTIC.
Collapse
Affiliation(s)
- Debadatta Mohapatra
- Phytomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Dulla Naveen Kumar
- Nanomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Singh Shreya
- Phytomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Vivek Pandey
- Centre for Genetics Disorders, Institute of Science (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Pawan K Dubey
- Centre for Genetics Disorders, Institute of Science (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Ashish Kumar Agrawal
- Nanomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Alakh N Sahu
- Phytomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Uttar Pradesh, Varanasi, 221005, India.
| |
Collapse
|
7
|
Tang Z, Zhang Y, Yu Z, Luo Z. Metformin Suppresses Stemness of Non-Small-Cell Lung Cancer Induced by Paclitaxel through FOXO3a. Int J Mol Sci 2023; 24:16611. [PMID: 38068934 PMCID: PMC10705988 DOI: 10.3390/ijms242316611] [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: 09/18/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Cancer stem cells (CSCs) play a pivotal role in drug resistance and metastasis. Among the key players, Forkhead box O3a (FOXO3a) acts as a tumor suppressor. This study aimed to unravel the role of FOXO3a in mediating the inhibitory effect of metformin on cancer stemness derived from paclitaxel (PTX)-resistant non-small-cell lung cancer (NSCLC) cells. We showed that CSC-like features were acquired by the chronic induction of resistance to PTX, concurrently with inactivation of FOXO3a. In line with this, knockdown of FOXO3a in PTX-sensitive cells led to changes toward stemness, while overexpression of FOXO3a in PTX-resistant cells mitigated stemness in vitro and remarkably curbed the tumorigenesis of NSCLC/PTX cells in vivo. Furthermore, metformin suppressed the self-renewal ability of PTX-resistant cells, reduced the expression of stemness-related markers (c-MYC, Oct4, Nanog and Notch), and upregulated FOXO3a, events concomitant with the activation of AMP-activated protein kinase (AMPK). All these changes were recapitulated by silencing FOXO3a in PTX-sensitive cells. Intriguingly, the introduction of the AMPK dominant negative mutant offset the inhibitory effect of metformin on the stemness of PTX-resistant cells. In addition, FOXO3a levels were elevated by the treatment of PTX-resistant cells with MK2206 (an Akt inhibitor) and U0126 (a MEK inhibitor). Collectively, our findings indicate that metformin exerts its effect on FOXO3a through the activation of AMPK and the inhibition of protein kinase B (Akt) and MAPK/extracellular signal-regulated kinase (MEK), culminating in the suppression of stemness in paclitaxel-resistant NSCLC cells.
Collapse
Affiliation(s)
- Zhimin Tang
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China;
| | - Yilan Zhang
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330031, China; (Y.Z.); (Z.Y.)
| | - Zhengyi Yu
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330031, China; (Y.Z.); (Z.Y.)
| | - Zhijun Luo
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China;
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330031, China; (Y.Z.); (Z.Y.)
| |
Collapse
|
8
|
Diwan D, Cheng L, Usmani Z, Sharma M, Holden N, Willoughby N, Sangwan N, Baadhe RR, Liu C, Gupta VK. Microbial cancer therapeutics: A promising approach. Semin Cancer Biol 2022; 86:931-950. [PMID: 33979677 DOI: 10.1016/j.semcancer.2021.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 01/27/2023]
Abstract
The success of conventional cancer therapeutics is hindered by associated dreadful side-effects of antibiotic resistance and the dearth of antitumor drugs' selectivity and specificity. Hence, the conceptual evolution of anti-cancerous therapeutic agents that selectively target cancer cells without impacting the healthy cells or tissues, has led to a new wave of scientific interest in microbial-derived bioactive molecules. Such strategic solutions may pave the way to surmount the shortcomings of conventional therapies and raise the potential and hope for the cure of wide range of cancer in a selective manner. This review aims to provide a comprehensive summary of anti-carcinogenic properties and underlying mechanisms of bioactive molecules of microbial origin, and discuss the current challenges and effective therapeutic application of combinatorial strategies to attain minimal systemic side-effects.
Collapse
Affiliation(s)
- Deepti Diwan
- Washington University, School of Medicine, Saint Louis, MO, USA
| | - Lei Cheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 230032, China
| | - Zeba Usmani
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Minaxi Sharma
- Department of Food Technology, Akal College of Agriculture, Eternal University, Baru Sahib, Himachal Pradesh, 173101, India
| | - Nicola Holden
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK
| | - Nicholas Willoughby
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Neelam Sangwan
- Department of Biochemistry, Central University of Haryana, Mahendergarh, Haryana, 123031, India
| | - Rama Raju Baadhe
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Vijai Kumar Gupta
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK; Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK.
| |
Collapse
|
9
|
Yathindranath V, Safa N, Sajesh BV, Schwinghamer K, Vanan MI, Bux R, Sitar DS, Pitz M, Siahaan TJ, Miller DW. Spermidine/Spermine N1-Acetyltransferase 1 ( SAT1)-A Potential Gene Target for Selective Sensitization of Glioblastoma Cells Using an Ionizable Lipid Nanoparticle to Deliver siRNA. Cancers (Basel) 2022; 14:5179. [PMID: 36358597 PMCID: PMC9656607 DOI: 10.3390/cancers14215179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2023] Open
Abstract
Spermidine/spermine N1-acetyltransferase 1 (SAT1) responsible for cell polyamine catabolism is overexpressed in glioblastoma multiforme (GB). Its role in tumor survival and promoting resistance towards radiation therapy has made it an interesting target for therapy. In this study, we prepared a lipid nanoparticle-based siRNA delivery system (LNP-siSAT1) to selectively knockdown (KD) SAT1 enzyme in a human glioblastoma cell line. The LNP-siSAT1 containing ionizable DODAP lipid was prepared following a microfluidics mixing method and the resulting nanoparticles had a hydrodynamic size of around 80 nm and a neutral surface charge. The LNP-siSAT1 effectively knocked down the SAT1 expression in U251, LN229, and 42MGBA GB cells, and other brain-relevant endothelial (hCMEC/D3), astrocyte (HA) and macrophage (ANA-1) cells at the mRNA and protein levels. SAT1 KD in U251 cells resulted in a 40% loss in cell viability. Furthermore, SAT1 KD in U251, LN229 and 42MGBA cells sensitized them towards radiation and chemotherapy treatments. In contrast, despite similar SAT1 KD in other brain-relevant cells no significant effect on cytotoxic response, either alone or in combination, was observed. A major roadblock for brain therapeutics is their ability to cross the highly restrictive blood-brain barrier (BBB) presented by the brain microcapillary endothelial cells. Here, we used the BBB circumventing approach to enhance the delivery of LNP-siSAT1 across a BBB cell culture model. A cadherin binding peptide (ADTC5) was used to transiently open the BBB tight junctions to promote paracellular diffusion of LNP-siSAT1. These results suggest LNP-siSAT1 may provide a safe and effective method for reducing SAT1 and sensitizing GB cells to radiation and chemotherapeutic agents.
Collapse
Affiliation(s)
- Vinith Yathindranath
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0Z3, Canada
| | - Nura Safa
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0Z3, Canada
| | - Babu V. Sajesh
- Cancer Care Manitoba Research Institute—CCMRI, Winnipeg, MB R3E 0V9, Canada
| | - Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA
| | - Magimairajan Issai Vanan
- Cancer Care Manitoba Research Institute—CCMRI, Winnipeg, MB R3E 0V9, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Rashid Bux
- BioMark Diagnostics Inc., Richmond, BC V6X 2W2, Canada
| | - Daniel S. Sitar
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0Z3, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Marshall Pitz
- Cancer Care Manitoba Research Institute—CCMRI, Winnipeg, MB R3E 0V9, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA
| | - Donald W. Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0Z3, Canada
| |
Collapse
|
10
|
Combined Therapy with Dacarbazine and Hyperthermia Induces Cytotoxicity in A375 and MNT-1 Melanoma Cells. Int J Mol Sci 2022; 23:ijms23073586. [PMID: 35408947 PMCID: PMC8998307 DOI: 10.3390/ijms23073586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/16/2023] Open
Abstract
Melanoma is a drug-resistant cancer, representing a serious challenge in cancer treatment. Dacarbazine (DTIC) is the standard drug in metastatic melanoma treatment, despite the poor results. Hyperthermia has been proven to potentiate chemotherapy. Hence, this work analyzed the combined action of hyperthermia and DTIC on A375 and MNT-1 cell lines. First, temperatures between 40 °C and 45 °C were tested. The effect of DTIC on cell viability was also investigated after exposures of 24, 48, and 72 h. Then, cells were exposed to 43 °C and to the respective DTIC IC10 or IC20 of each time exposure. Overall, hyperthermia reduced cell viability, however, 45 °C caused an excessive cell death (>90%). Combinational treatment revealed that hyperthermia potentiates DTIC’s effect, but it is dependent on the concentration and temperature used. Also, it has different mechanisms from the treatments alone, delaying A375 cells at the G2/M phase and MNT-1 cells at the S and G2/M phases. Intracellular reactive oxygen species (ROS) levels increased after treatment with hyperthermia, but the combined treatment showed no additional differences. Also, hyperthermia highly increased the number of A375 early apoptotic cells. These results suggest that combining hyperthermia and DTIC should be more explored to improve melanoma treatment.
Collapse
|
11
|
Caffeine improves the cytotoxic effect of dacarbazine on B16F10 murine melanoma cells. Bioorg Chem 2021; 120:105576. [PMID: 34979447 DOI: 10.1016/j.bioorg.2021.105576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Caffeine has been studied as a potentiating agent in chemotherapy against some types of cancer, but there are few reports on its effects on melanoma. This study aimed to investigate caffeine's ability to enhance the effects of dacarbazine in vitro. MATERIALS AND METHODS Murine melanoma B16F10 cells were treated 24 h with 1-40 µM caffeine. We evaluated cytotoxicity, DNA damage, apoptosis, and oxidative lesion induced by dacarbazine associated with caffeine. The metabolization of these drugs, as well as immunocytochemical labeling, were also evaluated. CONCLUSIONS The pre-treatment with caffeine showed to be more effective. Caffeine potentiated dacarbazine-induced cytotoxic effects by increasing dacarbazine biotransformation, apoptosis, DNA damage, and malondialdehyde levels; also, caffeine reduced Ki67 and ERK1/2 nuclear labeling and increased p53 labeling in B16F10 cells. In our experiment, caffeine promoted modifications associated with dacarbazine metabolism by viable cells potentiating this antineoplastic drug. These promising results should be further evaluated in experimental models in vivo.
Collapse
|
12
|
Preclinical Evaluation of LVR01 Attenuated Salmonella as Neoadjuvant Intralesional Therapy in Combination with Chemotherapy for Melanoma Treatment. J Invest Dermatol 2021; 142:1435-1443.e2. [PMID: 34662563 DOI: 10.1016/j.jid.2021.08.442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/16/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022]
Abstract
Treatment of malignant melanoma has improved in the last few years owing to early detection and new therapeutic options. Still, management of advanced disease remains a challenge because it requires systemic treatment. In such cases, dacarbazine-based chemotherapy has been widely used, despite low efficacy. Neoadjuvant therapies emerge as alternative options that could help chemotherapy to achieve increased benefit. In this work, we evaluate LVR01, an attenuated Salmonella enterica serovar typhimurium, as neoadjuvant intralesional therapy in combination with dacarbazine in a preclinical melanoma model. B16F1 melanoma‒bearing mice received intraperitoneal administration of dacarbazine for 3 consecutive days. LVR01 treatment, consisting of one single intratumoral injection, was applied 1 day before chemotherapy began. This therapeutic approach retarded tumor growth and prolonged overall survival, revealing a strong synergistic antitumor effect. Dacarbazine induced a drastic reduction of secondary lymphoid organ cellularity, which was partially restored by Salmonella, particularly potentiating activated cytotoxic cell compartments. Systemic immune reactivation could be a consequence of the intense inflammatory tumor microenvironment induced by LVR01. We propose that the use of LVR01 as neoadjuvant intralesional therapy could be considered as an interesting strategy with close clinical application to boost chemotherapy effect in patients with melanoma.
Collapse
|
13
|
Gonçalves JP, da Cruz AF, Nunes ÁM, Meneghetti MR, de Barros HR, Borges BS, de Medeiros LCAS, Soares MJ, Dos Santos MP, Grassi MT, Rossi GR, Bellan DL, Biscaia SMP, Cristal AM, Buzzo JLA, Ribeiro YC, Acco A, Cardoso MB, Simas FF, Trindade ES, Riegel-Vidotti IC, de Oliveira CC. Biocompatible gum arabic-gold nanorod composite as an effective therapy for mistreated melanomas. Int J Biol Macromol 2021; 185:551-561. [PMID: 34216657 DOI: 10.1016/j.ijbiomac.2021.06.172] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/23/2021] [Accepted: 06/26/2021] [Indexed: 01/13/2023]
Abstract
Advanced melanoma patients that are not included in common genetic classificatory groups lack effective and safe therapeutic options. Chemotherapy and immunotherapy show unsatisfactory results and devastating adverse effects for these called triple wild-type patients. New approaches exploring the intrinsic antitumor properties of gold nanoparticles might reverse this scenario as a safer and more effective alternative. Therefore, we investigated the efficacy and safety of a composite made of gum arabic-functionalized gold nanorods (GA-AuNRs) against triple wild-type melanoma. The natural polymer gum arabic successfully stabilized the nanorods in the biological environment and was essential to improve their biocompatibility. In vivo results obtained from treating triple wild-type melanoma-bearing mice showed that GA-AuNRs remarkably reduced primary tumor growth by 45%. Furthermore, GA-AuNRs induced tumor histological features associated with better prognosis while also reducing superficial lung metastasis depth and the incidence of intrapulmonary metastasis. GA-AuNRs' efficacy comes from their capacity to reduce melanoma cells ability to invade the extracellular matrix and grow into colonies, in addition to a likely immunomodulatory effect induced by gum arabic. Additionally, a broad safety investigation found no evidence of adverse effects after GA-AuNRs treatment. Therefore, this study unprecedentedly reports GA-AuNRs as a potential nanomedicine for advanced triple wild-type melanomas.
Collapse
Affiliation(s)
- Jenifer P Gonçalves
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Anderson F da Cruz
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Ábner M Nunes
- Catalysis and Chemical Reactivity Group, Institute of Chemistry and Biotechnology, Universidade Federal de Alagoas, Av. Lourival de Melo Mota, s/n, CEP 57072-900 Maceió, AL, Brazil
| | - Mario R Meneghetti
- Catalysis and Chemical Reactivity Group, Institute of Chemistry and Biotechnology, Universidade Federal de Alagoas, Av. Lourival de Melo Mota, s/n, CEP 57072-900 Maceió, AL, Brazil
| | - Heloise R de Barros
- Macromolecules and Interfaces Research Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil; Laboratory of Electroactive Materials, Chemistry Institute, Universidade de São Paulo, Av Professor Lineu Prestes, 748, CEP 05513-970 São Paulo, SP, Brazil
| | - Beatriz S Borges
- Laboratory of Cell Biology, Instituto Carlos Chagas (ICC/Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775, CEP 81350-010 Curitiba, PR, Brazil
| | - Lia C A S de Medeiros
- Laboratory of Cell Biology, Instituto Carlos Chagas (ICC/Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775, CEP 81350-010 Curitiba, PR, Brazil
| | - Maurilio J Soares
- Laboratory of Cell Biology, Instituto Carlos Chagas (ICC/Fiocruz), Rua Prof. Algacyr Munhoz Mader, 3775, CEP 81350-010 Curitiba, PR, Brazil
| | - Mayara P Dos Santos
- Environmental Chemistry Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Marco T Grassi
- Environmental Chemistry Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Gustavo R Rossi
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Daniel L Bellan
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Stellee M P Biscaia
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Aline M Cristal
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - João L A Buzzo
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Yasmin C Ribeiro
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Alexandra Acco
- Department of Pharmacology, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Mateus B Cardoso
- Laboratório Nacional de Luz Síncrotron, Centro Nacional de Pesquisa em Energia e Materiais, Rua Giuseppe Máximo Scolfaro, 10000, CEP 13083-970, Campinas, SP, Brazil
| | - Fernanda F Simas
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Edvaldo S Trindade
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Izabel C Riegel-Vidotti
- Macromolecules and Interfaces Research Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Carolina C de Oliveira
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil.
| |
Collapse
|
14
|
Salinomycin-loaded injectable thermosensitive hydrogels for glioblastoma therapy. Int J Pharm 2021; 598:120316. [PMID: 33540001 DOI: 10.1016/j.ijpharm.2021.120316] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/18/2021] [Accepted: 01/23/2021] [Indexed: 12/20/2022]
Abstract
Local drug delivery approaches for treating brain tumors not only diminish the toxicity of systemic chemotherapy, but also circumvent the blood-brain barrier (BBB) which restricts the passage of most chemotherapeutics to the brain. Recently, salinomycin has attracted much attention as a potential chemotherapeutic agent in a variety of cancers. In this study, poly (ethylene oxide)/poly (propylene oxide)/poly (ethylene oxide) (PEO-PPO-PEO, Pluronic F127) and poly (dl-lactide-co-glycolide-b-ethylene glycol-b-dl-lactide-co-glycolide) (PLGA-PEG-PLGA), the two most common thermosensitive copolymers, were utilized as local delivery systems for salinomycin in the treatment of glioblastoma. The Pluronic and PLGA-PEG-PLGA hydrogels released 100% and 36% of the encapsulated salinomycin over a one-week period, respectively. While both hydrogels were found to be effective at inhibiting glioblastoma cell proliferation, inducing apoptosis and generating intracellular reactive oxygen species, the Pluronic formulation showed better biocompatibility, a superior drug release profile and an ability to further enhance the cytotoxicity of salinomycin, compared to the PLGA-PEG-PLGA hydrogel formulation. Animal studies in subcutaneous U251 xenograftednudemice also revealed that Pluronic + salinomycin hydrogel reduced tumor growth compared to free salinomycin- and PBS-treated mice by 4-fold and 6-fold, respectively within 12 days. Therefore, it is envisaged that salinomycin-loaded Pluronic can be utilized as an injectable thermosensitive hydrogel platform for local treatment of glioblastoma, providing a sustained release of salinomycin at the tumor site and potentially bypassing the BBB for drug delivery to the brain.
Collapse
|
15
|
Wang J, Zhuo J, Tao Y, Xu S, Chen Z, Yang F, Ke Q, Xie H, Zheng S, Wang H, Xu X. Salinomycin-Loaded Small-Molecule Nanoprodrugs Enhance Anticancer Activity in Hepatocellular Carcinoma. Int J Nanomedicine 2020; 15:6839-6854. [PMID: 32982236 PMCID: PMC7501963 DOI: 10.2147/ijn.s236928] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Background There is currently no effective treatment for advanced hepatocellular carcinoma (HCC), and chemotherapy has little effect on long-term survival of HCC patients, largely due to the cancer stem cell (CSC) chemoresistance of HCC. Methods We constructed a small-molecule nanometer-sized prodrug (nanoprodrug) loaded with salinomycin (SAL) for the treatment of HCC. SAL was encapsulated by the prodrug LA-SN38 (linoleic acid modified 7-ethyl-10-hydroxycamptothecin) to construct a self-assembled nanoprodrug further PEGylated with DSPE-PEG2000. We characterized this codelivered nanoprodrug and its antitumor activity both in vitro in human HCC cell lines and in vivo in mice. Results Delivery of the SAL- and LA-SN38-based nanoprodrugs effectively promoted apoptosis of HCC cells, exerted inhibition of HCC tumor-sphere formation as well as HCC cell motility and invasion, and reduced the proportion of CD133+ HCC-CSC cells. In nude mice, the nanoprodrug suppressed growth of tumor xenografts derived from human cell lines and patient. Conclusion Our results show that SAL-based nanoprodrugs are a promising platform for treating patients with HCC and a novel strategy for combination therapy of cancers.
Collapse
Affiliation(s)
- Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Jianyong Zhuo
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China
| | - Yaoye Tao
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China
| | - Shengjun Xu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China
| | - Zun Chen
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China
| | - Fan Yang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China
| | - Qinghong Ke
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Haiyang Xie
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou 310003, People's Republic of China
| | - Hangxiang Wang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou 310003, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| |
Collapse
|
16
|
Malla WA, Arora R, Khan RIN, Mahajan S, Tiwari AK. Apoptin as a Tumor-Specific Therapeutic Agent: Current Perspective on Mechanism of Action and Delivery Systems. Front Cell Dev Biol 2020; 8:524. [PMID: 32671070 PMCID: PMC7330108 DOI: 10.3389/fcell.2020.00524] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide in humans and animals. Conventional treatment regimens often fail to produce the desired outcome due to disturbances in cell physiology that arise during the process of transformation. Additionally, development of treatment regimens with no or minimum side-effects is one of the thrust areas of modern cancer research. Oncolytic viral gene therapy employs certain viral genes which on ectopic expression find and selectively destroy malignant cells, thereby achieving tumor cell death without harming the normal cells in the neighborhood. Apoptin, encoded by Chicken Infectious Anemia Virus' VP3 gene, is a proline-rich protein capable of inducing apoptosis in cancer cells in a selective manner. In normal cells, the filamentous Apoptin becomes aggregated toward the cell margins, but is eventually degraded by proteasomes without harming the cells. In malignant cells, after activation by phosphorylation by a cancer cell-specific kinase whose identity is disputed, Apoptin accumulates in the nucleus, undergoes aggregation to form multimers, and prevents the dividing cancer cells from repairing their DNA lesions, thereby forcing them to undergo apoptosis. In this review, we discuss the present knowledge about the structure of Apoptin protein, elaborate on its mechanism of action, and summarize various strategies that have been used to deliver it as an anticancer drug in various cancer models.
Collapse
Affiliation(s)
- Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Sonalika Mahajan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Ashok Kumar Tiwari
- Division of Biological Standardisation, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
17
|
Sen’kova AV, Savin IA, Kabilova TO, Zenkova MA, Chernolovskaya EL. Tumor-Suppressing, Immunostimulating, and Hepatotoxic Effects of Immunostimulatory RNA in Combination with Dacarbazine in a Murine Melanoma Model. Mol Biol 2020. [DOI: 10.1134/s0026893320020144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
18
|
Luchtel RA, Bhagat T, Pradhan K, Jacobs WR, Levine M, Verma A, Shenoy N. High-dose ascorbic acid synergizes with anti-PD1 in a lymphoma mouse model. Proc Natl Acad Sci U S A 2020; 117:1666-1677. [PMID: 31911474 PMCID: PMC6983418 DOI: 10.1073/pnas.1908158117] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Major efforts are underway to identify agents that can potentiate effects of immune checkpoint inhibition. Here, we show that ascorbic acid (AA) treatment caused genomewide demethylation and enhanced expression of endogenous retroviral elements in lymphoma cells. AA also increased 5-hydroxymethylcytosine (5hmC) levels of CD8+ T cells and enhanced their cytotoxic activity in a lymphoma coculture system. High-dose AA treatment synergized with anti-PD1 therapy in a syngeneic lymphoma mouse model, resulting in marked inhibition of tumor growth compared with either agent alone. Analysis of the intratumoral epigenome revealed increased 5hmC with AA treatment, consistent with in vitro findings. Analysis of the tumor immune microenvironment revealed that AA strikingly increased intratumoral infiltration of CD8+ T cells and macrophages, suggesting enhanced tumor immune recognition. The combination treatment markedly enhanced intratumoral infiltration of macrophages and CD8+ T lymphocytes, granzyme B production by cytotoxic cells (cytotoxic T cells and natural killer cells), and interleukin 12 production by antigen-presenting cells compared with single-agent anti-PD1. These data indicate that AA potentiates anti-PD1 checkpoint inhibition through synergistic mechanisms. The study provides compelling rationale for testing combinations of high-dose AA and anti-PD1 agents in patients with aggressive B cell lymphoma as well as in preclinical models of other malignancies.
Collapse
Affiliation(s)
- Rebecca A Luchtel
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461
| | - Tushar Bhagat
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461
| | - Kith Pradhan
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461
| | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461;
- Department of Molecular Genetics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Amit Verma
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461
| | - Niraj Shenoy
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461;
- Experimental Therapeutics Program, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
19
|
Cancer Treatment Goes Viral: Using Viral Proteins to Induce Tumour-Specific Cell Death. Cancers (Basel) 2019; 11:cancers11121975. [PMID: 31817939 PMCID: PMC6966515 DOI: 10.3390/cancers11121975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/24/2022] Open
Abstract
Cell death is a tightly regulated process which can be exploited in cancer treatment to drive the killing of the tumour. Several conventional cancer therapies including chemotherapeutic agents target pathways involved in cell death, yet they often fail due to the lack of selectivity they have for tumour cells over healthy cells. Over the past decade, research has demonstrated the existence of numerous proteins which have an intrinsic tumour-specific toxicity, several of which originate from viruses. These tumour-selective viral proteins, although from distinct backgrounds, have several similar and interesting properties. Though the mechanism(s) of action of these proteins are not fully understood, it is possible that they can manipulate several cell death modes in cancer exemplifying the intricate interplay between these pathways. This review will discuss our current knowledge on the topic and outstanding questions, as well as deliberate the potential for viral proteins to progress into the clinic as successful cancer therapeutics.
Collapse
|
20
|
Banerjee I, De M, Dey G, Bharti R, Chattopadhyay S, Ali N, Chakrabarti P, Reis RL, Kundu SC, Mandal M. A peptide-modified solid lipid nanoparticle formulation of paclitaxel modulates immunity and outperforms dacarbazine in a murine melanoma model. Biomater Sci 2019; 7:1161-1178. [PMID: 30652182 DOI: 10.1039/c8bm01403e] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Melanoma is a highly aggressive skin cancer. A paclitaxel formulation of solid lipid nanoparticles modified with Tyr-3-octreotide (PSM) is employed to treat melanoma that highly expresses somatostatin receptors (SSTRs). PSM exerts more apoptotic and anti-invasive effects in B16F10 mice melanoma cells as compared to dacarbazine (DTIC), an approved chemotherapeutic drug for treating aggressive melanoma. Besides, PSM induces one of the biomarkers of immunogenic cell death in vitro and in vivo as confirmed by calreticulin exposure on the B16F10 cell surface. We observed a significant number of CD8 positive T cells in the tumor bed of the PSM treated group. As a result, PSM effectively reduces tumor volume in vivo as compared to DTIC. PSM also induces a favorable systemic immune response as determined in the spleen and sera of the treated animals. Importantly, PSM can reduce the number of nodule formations in the experimental lung metastasis model. Our experimentations indicate that the metronomic PSM exhibits remarkable anti-melanoma activities without any observable toxicity. This immune modulation behavior of PSM can be exploited for the therapy of melanoma and probably for other malignancies.
Collapse
Affiliation(s)
- Indranil Banerjee
- Division of Infectious Diseases and Immunology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S C Mullick Road, Kolkata - 700032, West Bengal, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Peng JQ, Fumoto S, Suga T, Miyamoto H, Kuroda N, Kawakami S, Nishida K. Targeted co-delivery of protein and drug to a tumor in vivo by sophisticated RGD-modified lipid-calcium carbonate nanoparticles. J Control Release 2019; 302:42-53. [PMID: 30926479 DOI: 10.1016/j.jconrel.2019.03.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/02/2019] [Accepted: 03/25/2019] [Indexed: 01/26/2023]
Abstract
Synchronized bio-distribution of combination therapies has several merits such as synergistic effects and reduced side-effects. Co-delivery of a protein and small molecule drug using a single nanocarrier is challenging because they possess totally different characteristics. Herein, we report the development of sophisticated nanoparticles composed of lipids, calcium carbonate and RGD peptide ligands for the co-delivery of a protein and small molecule drug combination via a simple preparation method. A 'one-step' ethanol injection method was employed to prepare the highly organized nanoparticles. The nanoparticles exhibited a spherical shape with ca. 130 nm diameter, and clearly had an integrated lipid layer covering the periphery. As a ligand, an RGD-modified lipid was post-inserted into the nanoparticles, which was important to overcome the 'PEG dilemma'. The pH-sensitivity of the targeted nanoparticles contributed to the efficient intracellular co-delivery of a protein and drug combination in Colon26 tumor cells, and noticeably improved their accumulation in the tumor region of xenograft mice. Synchronized bio-distribution of the protein and drug was achieved, which was the foundation for the synergistic effects of the combination. The targeting capability of the nanoparticles along with their pH-sensitive drug release and the synchronized bio-distribution of their cargos led to the significant antitumor activity of the SOD and paclitaxel combination in mice. This study provides novel information for the design and preparation of functionalized nanoparticles for the delivery of a protein/drug combination in vivo.
Collapse
Affiliation(s)
- Jian Qing Peng
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan.
| | - Tadaharu Suga
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Hirotaka Miyamoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Naotaka Kuroda
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Shigeru Kawakami
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| |
Collapse
|
22
|
Cui CX, Li YQ, Sun YJ, Zhu YL, Fang JB, Bai B, Li WJ, Li SZ, Ma YZ, Li X, Wang WH, Jin NY. Antitumor effect of a dual cancer-specific oncolytic adenovirus on prostate cancer PC-3 cells. Urol Oncol 2019; 37:352.e1-352.e18. [PMID: 30665692 DOI: 10.1016/j.urolonc.2018.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/21/2018] [Accepted: 12/16/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE Apoptin can specifically kill cancer cells but has no toxicity to normal cells. Human telomerase reverse transcriptase (hTERT) acts as a tumor-specific promoter, triggering certain genes to replicate or express only in tumor cells, conferring specific replication and killing abilities. This study aimed to investigate the anticancer potential of the recombinant adenovirus Ad-apoptin-hTERTp-E1a (Ad-VT) in prostate cancer. METHODS The pGL4.51 plasmid was used to transfect PC-3 cells to construct tumor cells stably expressing luciferase (PC-3-luc). Crystal violet staining and MTS assays determined the ability of Ad-VT to inhibit cell proliferation. Ad-VT-induced apoptosis of PC-3-luc cells was detected using Hoechst, Annexin V, JC-1 staining, and caspases activity analysis. PC-3-luc cells invasion and migration were detected using cell-scratch and Transwell assays. In vivo tumor inhibition was detected using imaging techniques. RESULTS Crystal violet staining and MTS results showed that the proliferation ability of PC-3-luc cells decreased significantly. Hoechst, JC-1, and Annexin V experiments demonstrated that Ad-VT mainly induced apoptosis to inhibit PC-3-luc cell proliferation. Ad-VT could significantly inhibit the migration and invasion of PC-3-luc cells over a short period of time. In vivo, Ad-VT could effectively inhibit tumor growth and prolong survival of the mice. CONCLUSIONS The recombinant adenovirus, comprising the apoptin protein and the hTERTp promoter, was able to inhibit the growth of prostate cancer PC-3 cells and promote their apoptosis.
Collapse
Affiliation(s)
- Chuan-Xin Cui
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P. R. China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China
| | - Yi-Quan Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Yu-Jia Sun
- Department of Operating Room, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Yi-Long Zhu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Jin-Bo Fang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Bing Bai
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Wen-Jie Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Jiang su Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, P. R. China
| | - Shan-Zhi Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Yi-Zhen Ma
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Jiang su Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, P. R. China
| | - Xiao Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China; Jiang su Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, P. R. China.
| | - Wei-Hua Wang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P. R. China; Department of Urology Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P. R. China.
| | - Ning-Yi Jin
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, P. R. China; Changchun University of Chinese Medicine, Changchun, P. R. China; Jiang su Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, P. R. China.
| |
Collapse
|
23
|
Wang W, Nakashima S, Nakamura S, Oda Y, Matsuda H. Anti-proliferative effect of auriculataoside A on B16 melanoma 4A5 cells by suppression of Cdc42-Rac1-RhoA signaling protein levels. J Nat Med 2019; 73:450-455. [PMID: 30627935 DOI: 10.1007/s11418-018-01278-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 12/27/2018] [Indexed: 01/24/2023]
Abstract
Auriculataoside A, an anthracenone dimer glycoside isolated from Cassia auriculata seed, shows anti-proliferative effects on cell line B16 melanoma 4A5 cells with an IC50 value of 0.82 μM. However, it shows no such effect on normal human dermal fibroblast (HDF) cells. To evaluate the mode of action underlying the anti-proliferative effect of auriculataoside A on cells, we examined changes in whole protein expression after treatment with auriculataoside A and found that the expression Cdc42, RhoA, and Rac1, which are Rho family GTPases, was reduced. Auriculataoside A also arrested the cell cycle at G1 phase. These results suggest that the suppression of the above proteins induced G1 arrest. In addition, auriculataoside A also suppressed the expression of β-catenin and c-Myc proteins. This action of auriculataoside A could be one of the mechanisms underlying its selective anti-proliferative effect on B16 melanoma cells.
Collapse
Affiliation(s)
- Weicheng Wang
- Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Souichi Nakashima
- Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Seikou Nakamura
- Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Yoshimi Oda
- Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan.,N.T.H Co., Ltd., 4F Sky-Ebisu Bldg., 1-8-11 Ebisu, Shibuya-ku, Tokyo, 150-0013, Japan
| | - Hisashi Matsuda
- Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan.
| |
Collapse
|
24
|
Murer P, Kiefer JD, Plüss L, Matasci M, Blümich SL, Stringhini M, Neri D. Targeted Delivery of TNF Potentiates the Antibody-Dependent Cell-Mediated Cytotoxicity of an Anti-Melanoma Immunoglobulin. J Invest Dermatol 2018; 139:1339-1348. [PMID: 30543899 DOI: 10.1016/j.jid.2018.11.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/01/2018] [Accepted: 11/13/2018] [Indexed: 12/27/2022]
Abstract
The recombinant murine IgG2a antibody TA99, directed against a melanoma antigen, was used to study combination modalities that potentiate antibody-dependent cell cytotoxicity. As previously reported, IgG2a(TA99) was extremely efficacious in preventing the growth of B16 lung metastases. However, the same antibody mediated only minimal tumor growth retardation when used to treat established neoplastic masses. The therapeutic activity of IgG2a(TA99) could be substantially enhanced by co-administration with an antibody-cytokine fusion (TA99-murine tumor necrosis factor [mTNF]), consisting of the TA99 antibody in single-chain variable fragment format fused to murine TNF. This fusion protein efficiently killed endothelial cells in vitro and displayed only minimal activity against B16 melanoma cells. In vivo, TA99-mTNF boosted the influx of natural killer cells and macrophages into B16 melanoma lesions. Therapy studies with two different administration schedules showed that the combination of TA99-mTNF and IgG2a(TA99) was superior to the individual products used as single agents. The combination treatment converted most of the tumor mass into a necrotic lesion, but a vital tumor rim eventually regrew, even when dacarbazine was included in the therapeutic regimen. The treatment modality described in this article may be applicable to the treatment of melanoma patients, given the specificity of the gp75 antigen and its conservation across species.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antibodies, Monoclonal, Murine-Derived/genetics
- Antibodies, Monoclonal, Murine-Derived/isolation & purification
- Antigens, Neoplasm/immunology
- CHO Cells
- Cell Line, Tumor/transplantation
- Cricetulus
- Drug Administration Schedule
- Drug Screening Assays, Antitumor
- Female
- Humans
- Immunoconjugates/administration & dosage
- Immunoconjugates/genetics
- Immunoconjugates/isolation & purification
- Immunoglobulin G/immunology
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Membrane Glycoproteins/immunology
- Mice
- Oxidoreductases/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/isolation & purification
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Tumor Necrosis Factor-alpha/administration & dosage
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/isolation & purification
Collapse
Affiliation(s)
- Patrizia Murer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Jonathan D Kiefer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Louis Plüss
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Sandra L Blümich
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, University of Zurich, Zurich, Switzerland
| | - Marco Stringhini
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland.
| |
Collapse
|
25
|
Micelles with ultralow critical micelle concentration as carriers for drug delivery. Nat Biomed Eng 2018; 2:318-325. [PMID: 30936455 DOI: 10.1038/s41551-018-0234-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 04/05/2018] [Indexed: 12/27/2022]
Abstract
Conventional micellar carriers disassemble into free surfactants when diluted at concentrations below the critical micelle concentration (CMC). This limits the bioavailability in vivo of injected hydrophobic drugs encapsulated in micellar systems. Here, we show that a micelle comprising a superhydrophilic zwitterionic polymer domain and a superhydrophobic lipid domain has an undetectable CMC below 10-6 mM-a value that is orders of magnitude lower than the CMCs (>10-3 mM) of typical micellar systems. We also show that zwitterionic moieties or zwitterionic polymers added to a micelle solution stabilize the micelles at concentrations below their inherent CMC. In a mouse model of melanoma, ultralow-CMC micelles encapsulating docetaxel led to the complete eradication of tumours, whereas conventional docetaxel micellar formulations did not reverse tumour growth. Ultralow-CMC micelles might become next-generation carriers for drug delivery.
Collapse
|
26
|
Zhang Y, Yang B, Zhao J, Li X, Zhang L, Zhai Z. Proteasome Inhibitor Carbobenzoxy-L-Leucyl-L-Leucyl-L-Leucinal (MG132) Enhances Therapeutic Effect of Paclitaxel on Breast Cancer by Inhibiting Nuclear Factor (NF)-κB Signaling. Med Sci Monit 2018; 24:294-304. [PMID: 29332931 PMCID: PMC5779800 DOI: 10.12659/msm.908139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Carbobenzoxy-L-leucyl-L-leucyl-L-leucinal (MG132), a peptide aldehyde proteasome inhibitor, can inhibit tumor progression by inactivating nuclear factor (NF)-κB signaling. Paclitaxel (PTX) is part of a routine regimen for the treatment of breast cancer. However, activation of the NF-κB pathway after treatment with PTX confers insensitivity to this drug. This study investigated the potential effect of MG132 as a co-treatment with PTX against breast cancer, and clarifies the underlying molecular mechanisms. Material/Methods Breast cancer cells were treated with PTX, MG132, or PTX plus MG132, and the therapeutic effects were evaluated phenotypically. A mouse model of breast cancer was used to determine the combined effect of PTX plus MG132 in vivo. Results Treatment with PTX plus MG132 suppressed aggressive phenotypes of breast cancer cells more effectively than PTX alone. Consistently, MG132 also enhanced the suppressive effect of PTX on tumor growth in C57BL/6 mice. Significantly, activation of the NF-κB pathway by PTX was attenuated by MG132. Conclusions Based on our findings, we suggest the application of MG132 in clinical practice in combination with PTX for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yunjing Zhang
- The Laboratory of Tumor Angiogenesis and Microenvironment, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Bin Yang
- The Laboratory of Tumor Angiogenesis and Microenvironment, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Jinping Zhao
- The Laboratory of Tumor Angiogenesis and Microenvironment, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Xiaoli Li
- The Laboratory of Tumor Angiogenesis and Microenvironment, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Long Zhang
- The Laboratory of Tumor Angiogenesis and Microenvironment, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| | - Zhenhua Zhai
- The Laboratory of Tumor Angiogenesis and Microenvironment, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland).,Department of Oncology, Cancer Centre, The First Hospital Affiliated to Jinzhou Medical University, Jinzhou, Liaoning, China (mainland)
| |
Collapse
|
27
|
Agha Amiri S, Shahhosseini S, Zarei N, Khorasanizadeh D, Aminollahi E, Rezaie F, Zargari M, Azizi M, Khalaj V. A novel anti-CD22 scFv-apoptin fusion protein induces apoptosis in malignant B-cells. AMB Express 2017; 7:112. [PMID: 28582973 PMCID: PMC5457376 DOI: 10.1186/s13568-017-0410-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/17/2022] Open
Abstract
CD22 marker is a highly internalizing antigen which is located on the surface of B-cells and is being used as a promising target for treatment of B cell malignancies. Monoclonal antibodies targeting CD22 have been introduced and some are currently under investigation in clinical trials. Building on the success of antibody drug conjugates, we developed a fusion protein consisting of a novel anti-CD22 scFv and apoptin and tested binding and therapeutic effects in lymphoma cells. The recombinant protein was expressed in E. coli and successfully purified and refolded. In vitro binding analysis by immunofluorescence and flow cytometry demonstrated that the recombinant protein specifically binds to CD22 positive Raji cells but not to CD22 negative Jurkat cells. The cytotoxic properties of scFv–apoptin were assessed by an MTT assay and Annexin V/PI flow cytometry analysis and showed that the recombinant protein induced apoptosis preferentially in Raji cells with no detectable effects in Jurkat cells. Our findings indicated that the recombinant anti-CD22 scFv–apoptin fusion protein could successfully cross the cell membrane and induce apoptosis with high specificity, make it as a promising molecule for immunotherapy of B-cell malignancies.
Collapse
|
28
|
Adoptive cell therapy with CD4 + T helper 1 cells and CD8 + cytotoxic T cells enhances complete rejection of an established tumour, leading to generation of endogenous memory responses to non-targeted tumour epitopes. Clin Transl Immunology 2017; 6:e160. [PMID: 29114389 PMCID: PMC5671987 DOI: 10.1038/cti.2017.37] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 01/31/2023] Open
Abstract
The results of adoptive T-cell therapies (ACTs) are very encouraging and show clinical evidence that ACT can provide a cure for patients with metastatic disease. However, various response rates and long-term cancer remission have been observed in different ACT trials. The types of T cells, prior treatment with chemotherapy and co-administration of other immune-target therapies have been found to influence the efficacy of ACT. In this study, we investigate the ability of ACT using CD4+ T helper 1 (Th1) cells and CD8+ cytotoxic T lymphocytes (CTLs) to reject the growth of established B16-ovalbumin (OVA) melanoma. CD8+ CTLs were found to be the main effector T cells that mediated tumour regression. However, low tumour-free survival rates were observed in ACT with CD8+ CTLs only. Co-transferring CD4+ Th1 cells and CD8+ CTLs has been observed to induce a synergistic antitumour response, resulting in complete regression in 80% of the tumour-bearing mice. We also examined a prior Dacarbazine (DTIC) and after virus-like particle (VLP)-OVA vaccine treatment to enhance ACT, but no therapeutic benefit was observed during primary B16-OVA tumour growth. Nevertheless, the ACT-mediated antitumour response was able to generate memory responses to both B16-OVA and B16-gp33 tumours. VLP-OVA vaccination following ACT enhances the memory responses to tumours that express a heterogenic population of both B16-OVA and B16-gp33 cells; however, it abolished the memory response to tumours consisting of only gp33-expressing cells. These findings provide important information for designing therapeutic treatments for patients with metastatic disease and cancer relapse to achieve durable cancer remission.
Collapse
|
29
|
Al-Qatati A, Aliwaini S. Combined pitavastatin and dacarbazine treatment activates apoptosis and autophagy resulting in synergistic cytotoxicity in melanoma cells. Oncol Lett 2017; 14:7993-7999. [PMID: 29344241 DOI: 10.3892/ol.2017.7189] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 09/01/2017] [Indexed: 01/10/2023] Open
Abstract
Melanoma is an aggressive skin cancer and its incidence is increasing faster than any other type of cancer. Whilst dacarbazine (DTIC) is the standard chemotherapy for metastatic melanoma, it has limited success. Statins, including pitavastatin, have been demonstrated to have a range of anti-cancer effects in a number of human cancer cell lines. The present study therefore explored the anti-cancer activity of combined DTIC and pitavastatin in A375 and WM115 human melanoma cells. Cell survival assays demonstrated that combined DTIC and pitavastatin treatment resulted in synergistic cell death. Cell cycle analyses further revealed that this combined treatment resulted in a G1 cell cycle arrest, as well as a sub-G1 population, indicative of apoptosis. Activation of apoptosis was confirmed by Annexin V-fluorescein isothiocyanate/propidium iodide double-staining and an increase in the levels of active caspase 3 and cleaved poly (ADP-ribose) polymerase. Furthermore, it was demonstrated that apoptosis occurs through the intrinsic pathway, evident from the release of cytochrome c. Finally, combined DTIC and pitavastatin treatment was demonstrated to also activate autophagy as part of a cell death mechanism. The present study provides novel evidence to suggest that the combined treatment of DTIC and pitavastatin may be effective in the treatment of melanoma.
Collapse
Affiliation(s)
- Abeer Al-Qatati
- Faculty of Applied Medical Sciences, Al-Azhar University, Gaza 1277, Palestine
| | - Saeb Aliwaini
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Islamic University of Gaza, Gaza 108, Palestine
| |
Collapse
|
30
|
PTD4-apoptin induces Bcl-2-insensitive apoptosis in human cervical carcinoma in vitro and in vivo. Anticancer Drugs 2017; 27:979-87. [PMID: 27548349 DOI: 10.1097/cad.0000000000000415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Worldwide, cervix carcinoma is among the most dangerous cancer types, and novel therapies are under development. Cancer treatments are often hampered because of lack of specificity. The chicken anemia virus-derived apoptin induces apoptosis selectively in tumor cells and leaves normal cells unharmed. Here, we have carried out in-vitro and in-vivo studies on the cytotoxic effect of apoptin in a cervix carcinoma model. Apoptin was fused to the protein transduction domain 4 (PTD4), enabling delivery of the fusion protein across cellular membranes. PTD4-apoptin protein is located in the nuclei of human cervical carcinoma HeLa cells and in the cytoplasm of normal cells L02. By MTT and flow cytometry analysis, we have proven that PTD4-apoptin protein induced apoptosis in the cervical carcinoma cells. PTD4-apoptin enhanced the level of active executioner caspase-3. Neither caspase-3 activation nor apoptin-induced accumulation of the mitochondrial outer-membrane protein Mfn-2 was affected by ectopic Bcl-2 expression. In contrast, apoptin-mediated AKT activation was inhibited by Bcl-2. In vivo, cervix carcinoma xenografts were treated for 7 days with PTD4-apoptin protein. The PTD4-apoptin treatment induced a decrease in the cervix carcinoma, whereas the PTD4-GFP protein-treated controls expanded significantly. TUNEL analysis showed that PTD4-apoptin protein induced apoptosis in cervix carcinoma cells, in contrast to the control PTD-GFP-treated ones. Our results indicate that apoptin is a potential anticancer agent for treating cervix carcinoma.
Collapse
|
31
|
Ethanol extract of Ilex hainanensis Merr. exhibits anti-melanoma activity by induction of G 1/S cell-cycle arrest and apoptosis. Chin J Integr Med 2017; 24:47-55. [PMID: 28741062 DOI: 10.1007/s11655-017-2544-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To evaluate anti-melanoma effect of ethanol extract of Ilex hainanensis Merr. (IME) and elucidate its underlying mechanism. METHODS Thirty-six tumor-bearing mice were randomized into 6 groups (n=6) as follows: model group, IME 25, 50, 100, and 200 mg/kg groups and dacarbazine (DTIC) 70 mg/kg group. The mice in the IME treatment groups were intragastrically administered with IME 25, 50, 100 or 200 mg/kg per day, respectively. The mice in the DTIC group were intraperitoneally injected with DTIC 70 mg/kg every 2 days. The drug administration was lasting for 14 days. The cell viability was evaluated by 3-(4,5-dime-thylthylthiazol-2-yl)-2, 5-diphenyl-tetrazolium bromide (MTT) assay. Flow cytometry was employed to detect cell cycle and apoptosis. The gene and protein expressions of nuclear factor κB-p65 (NF-κB-p65), Bcl-2, B-cell lymphomaextra large (Bcl-xL) and Bax were detected by quantitative real-time polymerase chain reaction and Western blot analyses. Caspases-3, -8, and -9 activities were detected using the colorimetric method. In addition, a B16-F10 melanoma xenograft mouse model was used to evaluate the anti-cancer activity of IME in vivo. Furthermore, a survival experiment of tumor-bearing mice was also performed to evaluate the possible toxicity of IME. RESULTS IME significantly inhibited the proliferation of B16-F10 cells (P<0.01). Flow cytometric analysis showed that IME induced G1/S cell cycle arrest and apoptosis (both P<0.01). IME inhibited activation of NF-κB, decreased the gene and protein expressions of Bcl-2, Bcl-xL, and increased the gene and protein expressions of Bax (all P<0.01). In addition, IME induced the activation of Caspases-3, -8, and -9 in B16-F10 cells. The study in vivo showed that IME significantly reduced tumor volume (P<0.01), and the inhibitory rate came up to 68.62%. IME also induced large areas of necrosis and intra-tumoral apoptosis that correlated with a reduction in tumor volume. Survival experiment showed that treatment with IME for 14 days significantly prolonged survival time and 20% of mice in the IME 200 mg/kg group were still alive until the 50th day. Notably, IME showed no apparent side-effects during the treatment period. CONCLUSION IME exhibited significant anti-melanoma activity in vitro and in vivo, suggesting that IME might be a promising effective candidate with lower toxic for malignant melanoma therapy.
Collapse
|
32
|
Lee JJ, Kang JA, Ryu Y, Han SS, Nam YR, Rho JK, Choi DS, Kang SW, Lee DE, Kim HS. Genetically engineered and self-assembled oncolytic protein nanoparticles for targeted cancer therapy. Biomaterials 2016; 120:22-31. [PMID: 28024232 DOI: 10.1016/j.biomaterials.2016.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/14/2016] [Accepted: 12/18/2016] [Indexed: 12/31/2022]
Abstract
The integration of a targeted delivery with a tumour-selective agent has been considered an ideal platform for achieving high therapeutic efficacy and negligible side effects in cancer therapy. Here, we present engineered protein nanoparticles comprising a tumour-selective oncolytic protein and a targeting moiety as a new format for the targeted cancer therapy. Apoptin from chicken anaemia virus (CAV) was used as a tumour-selective apoptotic protein. An EGFR-specific repebody, which is composed of LRR (Leucine-rich repeat) modules, was employed to play a dual role as a tumour-targeting moiety and a fusion partner for producing apoptin nanoparticles in E. coli, respectively. The repebody was genetically fused to apoptin, and the resulting fusion protein was shown to self-assemble into supramolecular repebody-apoptin nanoparticles with high homogeneity and stability as a soluble form when expressed in E. coli. The repebody-apoptin nanoparticles showed a remarkable anti-tumour activity with negligible side effects in xenograft mice through a cooperative action of the two protein components with distinct functional roles. The repebody-apoptin nanoparticles can be developed as a systemic injectable and tumour-selective therapeutic protein for targeted cancer treatment.
Collapse
Affiliation(s)
- Joong-Jae Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Jung Ae Kang
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Jeonbuk 580-185, South Korea
| | - Yiseul Ryu
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Sang-Soo Han
- Predictive Model Research Center, Korea Institute of Toxicology (KIT), Daejeon 34114, South Korea
| | - You Ree Nam
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Jeonbuk 580-185, South Korea
| | - Jong Kook Rho
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Jeonbuk 580-185, South Korea
| | - Dae Seong Choi
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Jeonbuk 580-185, South Korea
| | - Sun-Woong Kang
- Predictive Model Research Center, Korea Institute of Toxicology (KIT), Daejeon 34114, South Korea; Department of Human and Environmental Toxicology, University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Dong-Eun Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Jeonbuk 580-185, South Korea.
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea.
| |
Collapse
|
33
|
Huang W, Zhou Q, Yuan X, Ge ZM, Ran FX, Yang HY, Qiang GL, Li RT, Cui JR. Proteasome Inhibitor YSY01A Enhances Cisplatin Cytotoxicity in Cisplatin-Resistant Human Ovarian Cancer Cells. J Cancer 2016; 7:1133-41. [PMID: 27326257 PMCID: PMC4911881 DOI: 10.7150/jca.14519] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/26/2016] [Indexed: 12/28/2022] Open
Abstract
Cisplatin is one of the most common drugs used for treatment of solid tumors such as ovarian cancer. Unfortunately, the development of resistance against this cytotoxic agent limits its clinical use. Here we report that YSY01A, a novel proteasome inhibitor, is capable of suppressing survival of cisplatin-resistant ovarian cancer cells by inducing apoptosis. And YSY01A treatment enhances the cytotoxicity of cisplatin in drug-resistant ovarian cancer cells. Specifically, YSY01A abrogates regulatory proteins important for cell proliferation and anti-apoptosis including NF-κB p65 and STAT3, resulting in down-regulation of Bcl-2. A dramatic increase in cisplatin uptake was also observed by inductively coupled plasma-mass spectrometry following exposure to YSY01A. Taken together, YSY01A serves as a potential candidate for further development as anticancer therapeutics targeting the proteasome.
Collapse
Affiliation(s)
- Wei Huang
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Quan Zhou
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xia Yuan
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ze-Mei Ge
- 2. Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fu-Xiang Ran
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua-Yu Yang
- 3. Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences and PUMC, Beijing 100730, China
| | - Guang-Liang Qiang
- 4. Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Run-Tao Li
- 2. Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jing-Rong Cui
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
34
|
Gupta SK, Gandham RK, Sahoo AP, Tiwari AK. Viral genes as oncolytic agents for cancer therapy. Cell Mol Life Sci 2015; 72:1073-94. [PMID: 25408521 PMCID: PMC11113997 DOI: 10.1007/s00018-014-1782-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 10/29/2014] [Accepted: 11/13/2014] [Indexed: 12/20/2022]
Abstract
Many viruses have the ability to modulate the apoptosis, and to accomplish it; viruses encode proteins which specifically interact with the cellular signaling pathways. While some viruses encode proteins, which inhibit the apoptosis or death of the infected cells, there are viruses whose encoded proteins can kill the infected cells by multiple mechanisms, including apoptosis. A particular class of these viruses has specific gene(s) in their genomes which, upon ectopic expression, can kill the tumor cells selectively without affecting the normal cells. These genes and their encoded products have demonstrated great potential to be developed as novel anticancer therapeutic agents which can specifically target and kill the cancer cells leaving the normal cells unharmed. In this review, we will discuss about the viral genes having specific cancer cell killing properties, what is known about their functioning, signaling pathways and their therapeutic applications as anticancer agents.
Collapse
Affiliation(s)
- Shishir Kumar Gupta
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - Ravi Kumar Gandham
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. P. Sahoo
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. K. Tiwari
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| |
Collapse
|
35
|
Rollano Peñaloza OM, Lewandowska M, Stetefeld J, Ossysek K, Madej M, Bereta J, Sobczak M, Shojaei S, Ghavami S, Łos MJ. Apoptins: selective anticancer agents. Trends Mol Med 2014; 20:519-28. [PMID: 25164066 DOI: 10.1016/j.molmed.2014.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022]
Abstract
Therapies that selectively target cancer cells for death have been the center of intense research recently. One potential therapy may involve apoptin proteins, which are able to induce apoptosis in cancer cells leaving normal cells unharmed. Apoptin was originally discovered in the Chicken anemia virus (CAV); however, human gyroviruses (HGyV) have recently been found that also harbor apoptin-like proteins. Although the cancer cell specific activity of these apoptins appears to be well conserved, the precise functions and mechanisms of action are yet to be fully elucidated. Strategies for both delivering apoptin to treat tumors and disseminating the protein inside the tumor body are now being developed, and have shown promise in preclinical animal studies.
Collapse
Affiliation(s)
- Oscar M Rollano Peñaloza
- Department Clinical & Experimental Medicine, Division of Cell Biology, and Integrative Regenerative Medical Center, Linköping University, Linköping, Sweden; Instituto de Biologia Molecular y Biotecnologia, La Paz, Bolivia
| | | | - Joerg Stetefeld
- Department of Chemistry, University of Manitoba, Winnipeg, Canada
| | - Karolina Ossysek
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mariusz Madej
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mateusz Sobczak
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, College of Medicine, Faculty of Health Sciences, and Manitoba Institute of Child Health, University of Manitoba, Winnipeg, Canada; Health Policy Research Centre, Shiraz University of Medical Science, Shiraz, Iran
| | - Marek J Łos
- Department Clinical & Experimental Medicine, Division of Cell Biology, and Integrative Regenerative Medical Center, Linköping University, Linköping, Sweden; Department of Pathology, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
36
|
In vitro treatment of melanoma brain metastasis by simultaneously targeting the MAPK and PI3K signaling pathways. Int J Mol Sci 2014; 15:8773-94. [PMID: 24840574 PMCID: PMC4057758 DOI: 10.3390/ijms15058773] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/04/2014] [Accepted: 05/06/2014] [Indexed: 01/13/2023] Open
Abstract
Malignant melanoma is the most lethal form of skin cancer, with a high propensity to metastasize to the brain. More than 60% of melanomas have the BRAFV600E mutation, which activates the mitogen-activated protein kinase (MAPK) pathway [1]. In addition, increased PI3K (phosphoinositide 3-kinase) pathway activity has been demonstrated, through the loss of activity of the tumor suppressor gene, PTEN [2]. Here, we treated two melanoma brain metastasis cell lines, H1_DL2, harboring a BRAFV600E mutation and PTEN loss, and H3, harboring WT (wild-type) BRAF and PTEN loss, with the MAPK (BRAF) inhibitor vemurafenib and the PI3K pathway associated mTOR inhibitor temsirolimus. Combined use of the drugs inhibited tumor cell growth and proliferation in vitro in H1_DL2 cells, compared to single drug treatment. Treatment was less effective in the H3 cells. Furthermore, a strong inhibitory effect on the viability of H1_DL2 cells, when grown as 3D multicellular spheroids, was seen. The treatment inhibited the expression of pERK1/2 and reduced the expression of pAKT and p-mTOR in H1_DL2 cells, confirming that the MAPK and PI3K pathways were inhibited after drug treatment. Microarray experiments followed by principal component analysis (PCA) mapping showed distinct gene clustering after treatment, and cell cycle checkpoint regulators were affected. Global gene analysis indicated that functions related to cell survival and invasion were influenced by combined treatment. In conclusion, we demonstrate for the first time that combined therapy with vemurafenib and temsirolimus is effective on melanoma brain metastasis cells in vitro. The presented results highlight the potential of combined treatment to overcome treatment resistance that may develop after vemurafenib treatment of melanomas.
Collapse
|
37
|
Backendorf C, Noteborn MHM. Apoptin Towards Safe and Efficient Anticancer Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:39-59. [DOI: 10.1007/978-1-4471-6458-6_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
38
|
Xu D, Wu F, Chen Y, Wei L, Yuan W. pH-sensitive degradable nanoparticles for highly efficient intracellular delivery of exogenous protein. Int J Nanomedicine 2013; 8:3405-14. [PMID: 24039423 PMCID: PMC3771747 DOI: 10.2147/ijn.s47701] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Encapsulating exogenous proteins into a nanosized particulate system for delivery into cells is a great challenge. To address this issue, we developed a novel nanoparticle delivery method that differs from the nanoparticles reported to date because its core was composed of cross-linked dextran glassy nanoparticles which had pH in endosome-responsive environment and the protein was loaded in the core of cross-linked dextran glassy nanoparticles. METHODS In this study, dextran in a poly(ethylene glycol) aqueous two-phase system created a different chemical environment in which proteins were encapsulated very efficiently (84.3% and 89.6% for enhanced green fluorescent protein and bovine serum albumin, respectively) by thermodynamically favored partition. The structures of the nanoparticles were confirmed by confocal laser scanning microscopy and scanning electron microscopy. RESULTS The nanoparticles had a normal size distribution and a mean diameter of 186 nm. MTT assays showed that the nanoparticles were nontoxic up to a concentration of 2000 μg/mL in human hepatocarcinoma cell line SMMC-7721, HeLa, and BRL-3A cells. Of note, confocal laser scanning microscopy studies showed that nanoparticles loaded with fluorescein isothiocyanate-bovine serum albumin were efficiently delivered and released proteins into the cytoplasm of HeLa cells. Flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling assays showed that nanoparticles with a functional protein (apoptin) efficiently induced significant tumor cell apoptosis, which was confirmed by DAPI staining. CONCLUSION Our findings indicate that these nanoparticles meet the high demands for delivering protein medicines and have great potential in protein therapy.
Collapse
Affiliation(s)
- Dan Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yinghui Chen
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Liangming Wei
- Key Laboratory for Thin Film and Microfabrication of the Ministry of Education, Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
39
|
Lanz HL, Zimmerman RME, Brouwer J, Noteborn MHM, Backendorf C. Mitotic catastrophe triggered in human cancer cells by the viral protein apoptin. Cell Death Dis 2013; 4:e487. [PMID: 23392175 PMCID: PMC3734808 DOI: 10.1038/cddis.2013.2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mitotic catastrophe is an oncosuppressive mechanism that senses mitotic failure leading to cell death or senescence. As such, it protects against aneuploidy and genetic instability, and its induction in cancer cells by exogenous agents is currently seen as a promising therapeutic end point. Apoptin, a small protein from Chicken Anemia Virus (CAV), is known for its ability to selectively induce cell death in human tumor cells. Here, we show that apoptin triggers p53-independent abnormal spindle formation in osteosarcoma cells. Approximately 50% of apoptin-positive cells displayed non-bipolar spindles, a 10-fold increase as compared to control cells. Besides, tumor cells expressing apoptin are greatly limited in their progress through anaphase and telophase, and a significant drop in mitotic cells past the meta-to-anaphase transition is observed. Time-lapse microscopy showed that mitotic osteosarcoma cells expressing apoptin displayed aberrant mitotic figures and/or had a prolonged cycling time during mitosis. Importantly, all dividing cells expressing apoptin eventually underwent cell death either during mitosis or during the following interphase. We infer that apoptin can efficiently trigger cell death in dividing human tumor cells through induction of mitotic catastrophe. However, the killing activity of apoptin is not only confined to dividing cells, as the CAV-derived protein is also able to trigger caspase-3 activation and apoptosis in non-mitotic cancer cells.
Collapse
Affiliation(s)
- H L Lanz
- Department of Molecular Genetics, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
40
|
Mechanisms of Apoptin-induced cell death. Med Oncol 2011; 29:2985-91. [DOI: 10.1007/s12032-011-0119-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 11/09/2011] [Indexed: 12/22/2022]
|