1
|
Daurat M, Gauthier C, El Cheikh K, Ali LMA, Morère E, Bettache N, Gary-Bobo M, Morère A, Garcia M, Maynadier M, Basile I. Engineered therapeutic antibodies with mannose 6-phosphate analogues as a tool to degrade extracellular proteins. Front Immunol 2024; 15:1273280. [PMID: 38533506 PMCID: PMC10964947 DOI: 10.3389/fimmu.2024.1273280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 02/14/2024] [Indexed: 03/28/2024] Open
Abstract
Inducing the degradation of pathological soluble antigens could be the key to greatly enhancing the efficacy of therapeutic monoclonal antibodies (mAbs), extensively used in the treatment of autoimmune and inflammatory disorders or cancer. Lysosomal targeting has gained increasing interest in recent years due to its pharmaceutical applications far beyond the treatment of lysosomal diseases, as a way to address proteins to the lysosome for eventual degradation. Mannose 6-phosphonate derivatives (M6Pn), called AMFA, are unique glycovectors that can significantly enhance the cellular internalization of the proteins conjugated to AMFA via the cation-independent mannose 6-phosphate receptor (M6PR) pathway. AMFA engineering of mAbs results in the generation of a bifunctional antibody that is designed to bind both the antigen and the M6PR. The improvement of the therapeutic potential by AMFA engineering was investigated using two antibodies directed against soluble antigens: infliximab (IFX), directed against tumor necrosis factor α (TNF-α), and bevacizumab (BVZ), directed against the vascular endothelial growth factor (VEGF). AMFA conjugations to the antibodies were performed either on the oligosaccharidic chains of the antibodies or on the lysine residues. Both conjugations were controlled and reproducible and provided a novel affinity for the M6PR without altering the affinity for the antigen. The grafting of AMFA to mAb increased their cellular uptake through an M6PR-dependent mechanism. The antigens were also 2.6 to 5.7 times more internalized by mAb-AMFA and rapidly degraded in the cells. Additional cell culture studies also proved the significantly higher efficacy of IFX-AMFA and BVZ-AMFA compared to their unconjugated counterparts in inhibiting TNF-α and VEGF activities. Finally, studies in a zebrafish embryo model of angiogenesis and in xenografted chick embryos showed that BVZ-AMFA was more effective than BVZ in reducing angiogenesis. These results demonstrate that AMFA grafting induces the degradation of soluble antigens and a significant increase in the therapeutic efficacy. Engineering with mannose 6-phosphate analogues has the potential to develop a new class of antibodies for autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
| | - Corentin Gauthier
- NanoMedSyn, Montpellier, France
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Lamiaa M. A. Ali
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
- Department of Biochemistry Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Elodie Morère
- NanoMedSyn, Montpellier, France
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Nadir Bettache
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Magali Gary-Bobo
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Alain Morère
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | | |
Collapse
|
2
|
Senrung A, Tripathi T, Aggarwal N, Janjua D, Yadav J, Chaudhary A, Chhokar A, Joshi U, Bharti AC. Phytochemicals Showing Antiangiogenic Effect in Pre-clinical Models and their Potential as an Alternative to Existing Therapeutics. Curr Top Med Chem 2024; 24:259-300. [PMID: 37867279 DOI: 10.2174/0115680266264349231016094456] [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: 05/27/2023] [Revised: 07/25/2023] [Accepted: 08/10/2023] [Indexed: 10/24/2023]
Abstract
Angiogenesis, the formation of new blood vessels from a pre-existing vascular network, is an important hallmark of several pathological conditions, such as tumor growth and metastasis, proliferative retinopathies, including proliferative diabetic retinopathy and retinopathy of prematurity, age-related macular degeneration, rheumatoid arthritis, psoriasis, and endometriosis. Putting a halt to pathology-driven angiogenesis is considered an important therapeutic strategy to slow down or reduce the severity of pathological disorders. Considering the attrition rate of synthetic antiangiogenic compounds from the lab to reaching the market due to severe side effects, several compounds of natural origin are being explored for their antiangiogenic properties. Employing pre-clinical models for the evaluation of novel antiangiogenic compounds is a promising strategy for rapid screening of antiangiogenic compounds. These studies use a spectrum of angiogenic model systems that include HUVEC two-dimensional culture, nude mice, chick chorioallantoic membrane, transgenic zebrafish, and dorsal aorta from rats and chicks, depending upon available resources. The present article emphasizes the antiangiogenic activity of the phytochemicals shown to exhibit antiangiogenic behavior in these well-defined existing angiogenic models and highlights key molecular targets. Different models help to get a quick understanding of the efficacy and therapeutics mechanism of emerging lead molecules. The inherent variability in assays and corresponding different phytochemicals tested in each study prevent their immediate utilization in clinical studies. This review will discuss phytochemicals discovered using suitable preclinical antiangiogenic models, along with a special mention of leads that have entered clinical evaluation.
Collapse
Affiliation(s)
- Anna Senrung
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Deshbandhu College, University of Delhi, Delhi, India
| | - Udit Joshi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| |
Collapse
|
3
|
Lim WJ, Chan PF, Hamid RA. A 1, 4-benzoquinone derivative isolated from Ardisia crispa (Thunb.) A. DC. root suppresses angiogenesis via its angiogenic signaling cascades. Saudi Pharm J 2024; 32:101891. [PMID: 38111673 PMCID: PMC10727947 DOI: 10.1016/j.jsps.2023.101891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023] Open
Abstract
The root hexane extract of Ardisia crispa (ACRH), which belongs to the Primulaceae family, has been reported to possess anti-inflammatory, chemopreventive, anti-arthritic, and antiangiogenic activities. In this study, we isolated a p-benzoquinone derivative, 2-methoxy-6-undecyl-1,4-benzoquinone (AC2), from ACRH and investigated its potential antiangiogenic activity in human umbilical vein endothelial cells (HUVECs) and zebrafish embryo models. Prior to this study, AC2 was characterized using 1H NMR spectroscopy and MS. AC2 significantly suppressed HUVEC proliferation in a time-independent manner, with an IC50 value of 1.35 ± 0.05, 1.15 ± 0.02, and 1.00 ± 0.01 µg/mL at 24, 48, and 72 h, respectively. AC2 also induced apoptosis in HUVECs and significantly suppressed their migration, invasion, and tube formation in a concentration-dependent manner. Additionally, AC2 significantly attenuated most of the analyzed protein markers, including pro-MMP-2, VEGF-C, VEGF-D, angiopoietin-2, endothelin-1, fibroblast growth factor (FGF)-1, FGF-2, follistatin, heparin-binding epidermal growth factor-like growth factor (HB-EGF), and hepatocyte growth factor (HGF) at all tested concentrations. Furthermore, AC2 significantly inhibited zebrafish embryo intersegmental vessels (ISVs), confirming its antiangiogenic role. In conclusion, AC2 exhibits a potential anti-angiogenic effect by suppressing several proangiogenic and growth factors. Further studies are needed to investigate their effects on other excessive angiogenic diseases.
Collapse
Affiliation(s)
- Wen Jun Lim
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Pit Foong Chan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Roslida Abd Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
4
|
Mohammadi M, Sefidgar M, Aghanajafi C, Kohandel M, Soltani M. Computational Multi-Scale Modeling of Drug Delivery into an Anti-Angiogenic Therapy-Treated Tumor. Cancers (Basel) 2023; 15:5464. [PMID: 38001724 PMCID: PMC10670623 DOI: 10.3390/cancers15225464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
The present study develops a numerical model, which is the most complex one, in comparison to previous research to investigate drug delivery accompanied by the anti-angiogenesis effect. This paper simulates intravascular blood flow and interstitial fluid flow using a dynamic model. The model accounts for the non-Newtonian behavior of blood and incorporates the adaptation of the diameter of a heterogeneous microvascular network derived from modeling the evolution of endothelial cells toward a circular tumor sprouting from two-parent vessels, with and without imposing the inhibitory effect of angiostatin on a modified discrete angiogenesis model. The average solute exposure and its uniformity in solid tumors of different sizes are studied by numerically solving the convection-diffusion equation. Three different methodologies are considered for simulating anti-angiogenesis: modifying the capillary network, updating the transport properties, and considering both microvasculature and transport properties modifications. It is shown that anti-angiogenic therapy decreases drug wash-out in the periphery of the tumor. Results show the decisive role of microvascular structure, particularly its distribution, and interstitial transport properties modifications induced via vascular normalization on the quality of drug delivery, such that it is improved by 39% in uniformity by the second approach in R = 0.2 cm.
Collapse
Affiliation(s)
- Mahya Mohammadi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19919-43344, Iran; (M.M.); (C.A.)
| | - Mostafa Sefidgar
- Department of Mechanical Engineering, Pardis Branch, Islamic Azad University, Pardis 16581-74583, Iran;
| | - Cyrus Aghanajafi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19919-43344, Iran; (M.M.); (C.A.)
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19919-43344, Iran; (M.M.); (C.A.)
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Centre for Sustainable Business, International Business University, Toronto, ON M5S 2V1, Canada
| |
Collapse
|
5
|
De Gaetano F, Margani F, Barbera V, D’Angelo V, Germanò MP, Pistarà V, Ventura CA. Characterization and In Vivo Antiangiogenic Activity Evaluation of Morin-Based Cyclodextrin Inclusion Complexes. Pharmaceutics 2023; 15:2209. [PMID: 37765179 PMCID: PMC10536596 DOI: 10.3390/pharmaceutics15092209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/11/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Morin (MRN) is a natural compound with antiangiogenic, antioxidant, anti-inflammatory, and anticancer activity. However, it shows a very low water solubility (28 μg/mL) that reduces its oral absorption, making bioavailability low and unpredictable. To improve MRN solubility and positively affect its biological activity, particularly its antiangiogenic activity, in this work, we prepared the inclusion complexes of MNR with sulfobutylether-β-cyclodextrin (SBE-β-CD) and hydroxypropyl-β-cyclodextrin (HP-β-CD). The inclusion complexes obtained by the freeze-drying method were extensively characterized in solution (phase-solubility studies, UV-Vis titration, and NMR spectroscopy) and in the solid state (TGA, DSC, and WAXD analysis). The complexation significantly increased the water solubility by about 100 times for MRN/HP-β-CD and 115 times for MRN/SBE-β-CD. Furthermore, quantitative dissolution of the complexes was observed within 60 min, whilst 1% of the free drug dissolved in the same experimental time. 1H NMR and UV-Vis titration studies demonstrated both CDs well include the benzoyl moiety of the drug. Additionally, SBE-β-CD could interact with the cinnamoyl moiety of MRN too. The complexes are stable in solution, showing a high value of association constant, that is, 3380 M-1 for MRN/HP-β-CD and 2870 M-1 for MRN/SBE-β-CD. In vivo biological studies on chick embryo chorioallantoic membrane (CAM) and zebrafish embryo models demonstrated the high biocompatibility of the inclusion complexes and the effective increase in antiangiogenic activity of complexed MRN with respect to the free drug.
Collapse
Affiliation(s)
- Federica De Gaetano
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche e Ambientali, Università di Messina, Viale Ferdinando Stagno d’Alcontres 31, I-98166 Messina, Italy; (F.D.G.); (V.D.); (M.P.G.)
| | - Fatima Margani
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Via Mancinelli 7, I-20131 Milano, Italy; (F.M.); (V.B.)
| | - Vincenzina Barbera
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Via Mancinelli 7, I-20131 Milano, Italy; (F.M.); (V.B.)
| | - Valeria D’Angelo
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche e Ambientali, Università di Messina, Viale Ferdinando Stagno d’Alcontres 31, I-98166 Messina, Italy; (F.D.G.); (V.D.); (M.P.G.)
| | - Maria Paola Germanò
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche e Ambientali, Università di Messina, Viale Ferdinando Stagno d’Alcontres 31, I-98166 Messina, Italy; (F.D.G.); (V.D.); (M.P.G.)
| | - Venerando Pistarà
- Dipartimento di Scienze del Farmaco e della Salute, Università di Catania, Viale A. Doria 6, I-95125 Catania, Italy
| | - Cinzia Anna Ventura
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche e Ambientali, Università di Messina, Viale Ferdinando Stagno d’Alcontres 31, I-98166 Messina, Italy; (F.D.G.); (V.D.); (M.P.G.)
| |
Collapse
|
6
|
An G, Kim M, Park J, Park H, Hong T, Lim W, Song G. Embryonic exposure to chloroxylenol induces developmental defects and cardiovascular toxicity via oxidative stress, inflammation, and apoptosis in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2023; 268:109617. [PMID: 36965842 DOI: 10.1016/j.cbpc.2023.109617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
Chloroxylenol is an extensively consumed anti-microbial compound. Since its usage is on the rise due to the coronavirus pandemic and ban on other antimicrobial ingredients, recent studies have suggested the necessity of estimating its potential for ecotoxicity. The detrimental effect of chloroxylenol on zebrafish (Danio rerio) viability has been reported; however, research on the mechanisms underlying its toxicity is quite limited. Therefore, we applied the zebrafish model for elucidating responses against chloroxylenol to predict its toxicity toward human health and ecology. Zebrafish exposed to chloroxylenol (0, 0.5, 1, 2.5, 5, and 10 mg/L) at the embryonic stage (from 6 h post-fertilization (hpf) to 96 hpf) showed impaired viability and hatchability, and pathological phenotypes. To address these abnormalities, cellular responses such as oxidative stress, inflammation, and apoptosis were confirmed via in vivo imaging of a fluorescent dye or measurement of the transcriptional changes related to each response. In particular, developmental defects in the cardiovascular system of zebrafish exposed to 0, 0.5, 1, and 2.5 mg/L of chloroxylenol from 6 to 96 hpf were identified by structural analyses of the system in the flk1:eGFP transgenic line. Additional experiments were conducted using human umbilical vein endothelial cells (HUVECs) to predict the adverse impacts of chloroxylenol on the human vascular system. Chloroxylenol impairs the viability and tube formation ability of HUVECs by modulating ERK signaling. The findings obtained using the zebrafish model provide evidence of the possible risks of chloroxylenol exposure and suggest the importance of more in-depth ecotoxicological studies.
Collapse
Affiliation(s)
- Garam An
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Miji Kim
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Junho Park
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hahyun Park
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
7
|
Singhal SS, Garg R, Mohanty A, Garg P, Ramisetty SK, Mirzapoiazova T, Soldi R, Sharma S, Kulkarni P, Salgia R. Recent Advancement in Breast Cancer Research: Insights from Model Organisms-Mouse Models to Zebrafish. Cancers (Basel) 2023; 15:cancers15112961. [PMID: 37296923 DOI: 10.3390/cancers15112961] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Animal models have been utilized for decades to investigate the causes of human diseases and provide platforms for testing novel therapies. Indeed, breakthrough advances in genetically engineered mouse (GEM) models and xenograft transplantation technologies have dramatically benefited in elucidating the mechanisms underlying the pathogenesis of multiple diseases, including cancer. The currently available GEM models have been employed to assess specific genetic changes that underlay many features of carcinogenesis, including variations in tumor cell proliferation, apoptosis, invasion, metastasis, angiogenesis, and drug resistance. In addition, mice models render it easier to locate tumor biomarkers for the recognition, prognosis, and surveillance of cancer progression and recurrence. Furthermore, the patient-derived xenograft (PDX) model, which involves the direct surgical transfer of fresh human tumor samples to immunodeficient mice, has contributed significantly to advancing the field of drug discovery and therapeutics. Here, we provide a synopsis of mouse and zebrafish models used in cancer research as well as an interdisciplinary 'Team Medicine' approach that has not only accelerated our understanding of varied aspects of carcinogenesis but has also been instrumental in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Sharad S Singhal
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Rachana Garg
- Department of Surgery, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Atish Mohanty
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Sravani Keerthi Ramisetty
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Raffaella Soldi
- Translational Genomics Research Institute, Phoenix, AZ 85338, USA
| | - Sunil Sharma
- Translational Genomics Research Institute, Phoenix, AZ 85338, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
8
|
Deshmukh D, Hsu YF, Chiu CC, Jadhao M, Hsu SCN, Hu SY, Yang SH, Liu W. Antiangiogenic potential of Lepista nuda extract suppressing MAPK/p38 signaling-mediated developmental angiogenesis in zebrafish and HUVECs. Biomed Pharmacother 2023; 159:114219. [PMID: 36621144 DOI: 10.1016/j.biopha.2023.114219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
The medicinal properties of natural/edible plant products and their use are popular in traditional practice owing to their nutritional contents with little to no side effects. Lepista nuda (L. nuda), an edible mushroom (Clitocybe nuda, commonly known as blewit), has attracted researchers to evaluate its contents and the mechanism of its activities. In the current study, we focused on evaluating the antiangiogenic effects of L. nuda water extract on zebrafish development and in vitro human umbilical vein endothelial cell (HUVEC) tube formation. Bioactive components such as ergothioneine, eritadenine, and adenosine were identified and quantified by HPLC analysis. The L. nuda extract showed antiangiogenic properties and inhibited intersegmental vessel (ISV), caudal vein plexus (CVP), hyaloid vessel (HV), and subintestinal vessel (SIV) development in Tg (fli1: EGFP) zebrafish embryos. The expression of angiogenesis-related genes (vegfaa, kdrl, vegfba, flt1, kdr) was affected following L. nuda extract treatment. L. nuda extract attenuated in vitro HUVEC tube formation, migration, and invasion. Furthermore, inhibition of MAPK/p38 signaling and depletion of proangiogenic genes, including growth factors (fgf, ang2, and vegfa); primary and accessory receptors (tie2, vegfr2, and eng); MMPs (mmp1 and mmp2); and cytokines (il-1α, il-1β, il-6, and tnf-α) was observed in HUVECs following L. nuda treatment. An in vivo zebrafish xenograft assay showed that L. nuda extract inhibited HuCCT1 cell-induced SIV sprouting in HuCCT1-injected embryos. Collectively, the results suggest that L. nuda could be a potential inhibitor of angiogenesis limiting cancer progression.
Collapse
Affiliation(s)
- Dhanashri Deshmukh
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ya Fen Hsu
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan; National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115, Taiwan.
| | - Mahendra Jadhao
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shao-Yang Hu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 912, Taiwan.
| | - Shu-Hui Yang
- Department of Management and Utilization, Fengshan Tropical Horticultural Experimental Branch, Taiwan Agricultural Research Institute, Kaohsiung 807, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
9
|
Huo L, Liu X, Jaiswal Y, Xu H, Chen R, Lu R, Nong Y, Williams L, Liang Y, Jia Z. Design and Synthesis of Acridine-Triazole and Acridine-Thiadiazole Derivatives and Their Inhibitory Effect against Cancer Cells. Int J Mol Sci 2022; 24:ijms24010064. [PMID: 36613504 PMCID: PMC9820444 DOI: 10.3390/ijms24010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
We report herein the design and synthesis of a series of novel acridine-triazole and acridine-thiadiazole derivatives. The newly synthesized compounds and the key intermediates were all evaluated for their antitumor activities against human foreskin fibroblasts (HFF), human gastric cancer cells-803 (MGC-803), hepatocellular carcinoma bel-7404 (BEL-7404), large cell lung cancer cells (NCI-H460), and bladder cancer cells (T24). Most of the compounds exhibited high levels of antitumor activity against MGC-803 and T24 but low toxicity against human normal liver cells (LO2), and their effect was even better than the commercial anticancer drugs, 5-fluorouracil (5-FU) and cis-platinum. Further, pharmacological mechanisms such as topo I, cell cycle, cell apoptosis, and neovascularization were all evaluated. Only a few compounds exhibited potent topo I inhibitory activity at 100 μM. In addition, the most active compounds with an IC50 value of 5.52-8.93 μM were chosen, and they could induce cell apoptosis in the G2 stage of MGC-803 or mainly arrest T24 cells in the S stage. To our delight, most of the compounds exhibited lower zebrafish cytotoxicity but could strongly inhibit the formation of zebrafish sub-intestinal veins, indicating a potential for clinical application.
Collapse
Affiliation(s)
- Lini Huo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
| | - Xiaochen Liu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
| | - Yogini Jaiswal
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, USA
| | - Hao Xu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
| | - Rui Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
- Correspondence: (R.C.); (R.L.)
| | - Rumei Lu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
- Correspondence: (R.C.); (R.L.)
| | - Yaqin Nong
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
| | - Leonard Williams
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis, NC 28081, USA
| | - Yan Liang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Zhiruo Jia
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
| |
Collapse
|
10
|
Li X, Xu J, Xie J, Yang W. Research progress in targeted therapy and immunotherapy for gastric cancer. Chin Med J (Engl) 2022; 135:1299-1313. [PMID: 35830242 PMCID: PMC9433086 DOI: 10.1097/cm9.0000000000002185] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Gastric cancer (GC) is one of the most common malignant tumors worldwide. Its incidence ranks the 5th among all malignant tumors globally, and it is the 3rd leading cause of death among patients with cancer. Surgical treatment is the first choice in clinical practice. However, targeted therapy, immunotherapy, and other treatment methods have also become research hotspots at home and abroad with the development of individualized precision therapy in recent years, besides traditional radiotherapy and chemotherapy. At present, targeted therapy and immunotherapy are methods used for treating GC, and they have important clinical application value and prospects. This study aimed to review the research progress of targeted therapy and immunotherapy for GC, focusing on its mechanism of action and related important clinical trials, hoping to provide references for the clinical treatment of GC.
Collapse
Affiliation(s)
- Xuewei Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jun Xu
- Department of Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Wenhui Yang
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China
| |
Collapse
|
11
|
Cascallar M, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, Piñeiro R, de la Fuente M. What Zebrafish and Nanotechnology Can Offer for Cancer Treatments in the Age of Personalized Medicine. Cancers (Basel) 2022; 14:cancers14092238. [PMID: 35565373 PMCID: PMC9099873 DOI: 10.3390/cancers14092238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer causes millions of deaths each year and thus urgently requires the development of new therapeutic strategies. Nanotechnology-based anticancer therapies are a promising approach, with several formulations already approved and in clinical use. The evaluation of these therapies requires efficient in vivo models to study their behavior and interaction with cancer cells, and to optimize their properties to ensure maximum efficacy and safety. In this way, zebrafish is an important candidate due to its high homology with the human genoma, its large offspring, and the ease in developing specific cancer models. The role of zebrafish as a model for anticancer therapy studies has been highly evidenced, allowing researchers not only to perform drug screenings but also to evaluate novel therapies such as immunotherapies and nanotherapies. Beyond that, zebrafish can be used as an “avatar” model for performing patient-derived xenografts for personalized medicine. These characteristics place zebrafish in an attractive position as a role model for evaluating novel therapies for cancer treatment, such as nanomedicine.
Collapse
Affiliation(s)
- María Cascallar
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Roberto Piñeiro
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
- Correspondence: ; Tel.: +34-981-955-704
| |
Collapse
|
12
|
Kasica N, Święch A, Saładziak K, Mackiewicz J, Osęka M. The Inhibitory Effect of Selected D2 Dopaminergic Receptor Agonists on VEGF-Dependent Neovascularization in Zebrafish Larvae: Potential New Therapy in Ophthalmic Diseases. Cells 2022; 11:cells11071202. [PMID: 35406766 PMCID: PMC8997652 DOI: 10.3390/cells11071202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 01/01/2023] Open
Abstract
Pathological angiogenesis is correlated with many ophthalmic diseases. The most common are exudative age-related macular degeneration and proliferative diabetic retinopathy. The current treatment for these diseases is based on regularly administered anti-VEGF antibodies injections. In the study, we investigated selected D2 dopaminergic receptor agonists, namely bromocriptine, cabergoline and pergolide, on hypoxia-induced neovascularization. We used the zebrafish laboratory model, specifically three-day post fertilization (dpf) Tg(fli-1: EGFP) zebrafish larvae. To induce abnormal angiogenesis of hyaloid-retinal vessels (HRVs) and intersegmental vessels (ISVs), the larvae were treated with cobalt chloride (II) (CoCl2) (a hypoxia-inducing agent) from 24 h post fertilization. The inhibitory role of D2 dopaminergic receptor agonists was investigated using confocal microscopy and qPCR. Additionally, the results were compared to those obtained in the group treated with CoCl2 followed by bevacizumab, the well-known antiangiogenic agent. Confocal microscopy analyses revealed severe deformation of vessels in the CoCl2 treated group, while co-incubation with bromocriptine, cabergoline, pergolide and bevacizumab, respectively, significantly inhibited abnormalities of angiogenesis. The qPCR analyses supported the protective role of the chosen dopaminergic agonists by demonstrating their influence on CoCl2-derived upregulation of vegfaa expression. The present results suggest that the D2 receptor agonists can be considered as a new direction in research for antiangiogenic therapy.
Collapse
Affiliation(s)
- Natalia Kasica
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13 Street, Box 105J, 10-719 Olsztyn, Poland
- Correspondence:
| | - Anna Święch
- Department of Retina and Vitreus Surgery, Medical University in Lublin, Chmielna 1 Street, 20-079 Lublin, Poland; (A.Ś.); (K.S.); (J.M.)
| | - Katarzyna Saładziak
- Department of Retina and Vitreus Surgery, Medical University in Lublin, Chmielna 1 Street, 20-079 Lublin, Poland; (A.Ś.); (K.S.); (J.M.)
| | - Jerzy Mackiewicz
- Department of Retina and Vitreus Surgery, Medical University in Lublin, Chmielna 1 Street, 20-079 Lublin, Poland; (A.Ś.); (K.S.); (J.M.)
| | - Maciej Osęka
- Oftalabs Sp. z o.o., Wrocławska 130, 58-306 Wałbrzych, Poland;
| |
Collapse
|
13
|
Effect of non-repetitive linker on in vitro and in vivo properties of an anti-VEGF scFv. Sci Rep 2022; 12:5449. [PMID: 35361822 PMCID: PMC8971466 DOI: 10.1038/s41598-022-09324-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/03/2022] [Indexed: 11/08/2022] Open
Abstract
Single chain antibody fragments (scFvs) are favored in diagnostic and therapeutic fields thanks to their small size and the availability of various engineering approaches. Linker between variable heavy (VH) and light (VL) chains of scFv covalently links these domains and it can affect scFv’s bio-physical/chemical properties and in vivo activity. Thus, scFv linker design is important for a successful scFv construction, and flexible linkers are preferred for a proper pairing of VH–VL. The flexibility of the linker is determined by length and sequence content and glycine-serine (GS) linkers are commonly preferred for scFvs based on their highly flexible profiles. Despite the advantage of this provided flexibility, GS linkers carry repeated sequences which can cause problems for PCR-based engineering approaches and immunogenicity. Here, two different linkers, a repetitive GS linker and an alternative non-repetitive linker with similar flexibility but lower immunogenicity are employed to generate anti-Vascular Endothelial Growth Factor scFvs derived from bevacizumab. Our findings highlight a better in vitro profile of the non-repetitive linker such as a higher monomer ratio, higher thermal stability while there was no significant difference in in vivo efficacy in a zebrafish embryonic angiogenesis model. This is the first study to compare in vivo efficacy of scFvs with different linkers in a zebrafish model.
Collapse
|
14
|
Oxidative Stress and AKT-Associated Angiogenesis in a Zebrafish Model and Its Potential Application for Withanolides. Cells 2022; 11:cells11060961. [PMID: 35326412 PMCID: PMC8946239 DOI: 10.3390/cells11060961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress and the AKT serine/threonine kinase (AKT) signaling pathway are essential regulators in cellular migration, metastasis, and angiogenesis. More than 300 withanolides were discovered from the plant family Solanaceae, exhibiting diverse functions. Notably, the relationship between oxidative stress, AKT signaling, and angiogenesis in withanolide treatments lacks comprehensive understanding. Here, we summarize connecting evidence related to oxidative stress, AKT signaling, and angiogenesis in the zebrafish model. A convenient vertebrate model monitored the in vivo effects of developmental and tumor xenograft angiogenesis using zebrafish embryos. The oxidative stress and AKT-signaling-modulating abilities of withanolides were highlighted in cancer treatments, which indicated that further assessments of their angiogenesis-modulating potential are necessary in the future. Moreover, targeting AKT for inhibiting AKT and its AKT signaling shows the potential for anti-migration and anti-angiogenesis purposes for future application to withanolides. This particularly holds for investigating the anti-angiogenetic effects mediated by the oxidative stress and AKT signaling pathways in withanolide-based cancer therapy in the future.
Collapse
|
15
|
Yang Y, Meng WJ, Wang ZQ. Cancer Stem Cells and the Tumor Microenvironment in Gastric Cancer. Front Oncol 2022; 11:803974. [PMID: 35047411 PMCID: PMC8761735 DOI: 10.3389/fonc.2021.803974] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/08/2021] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) remains one of the leading causes of cancer-related death worldwide. Cancer stem cells (CSCs) might be responsible for tumor initiation, relapse, metastasis and treatment resistance of GC. The tumor microenvironment (TME) comprises tumor cells, immune cells, stromal cells and other extracellular components, which plays a pivotal role in tumor progression and therapy resistance. The properties of CSCs are regulated by cells and extracellular matrix components of the TME in some unique manners. This review will summarize current literature regarding the effects of CSCs and TME on the progression and therapy resistance of GC, while emphasizing the potential for developing successful anti-tumor therapy based on targeting the TME and CSCs.
Collapse
Affiliation(s)
| | - Wen-Jian Meng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
16
|
Basnet RM, Zizioli D, Muscò A, Finazzi D, Sigala S, Rossini E, Tobia C, Guerra J, Presta M, Memo M. Caffeine Inhibits Direct and Indirect Angiogenesis in Zebrafish Embryos. Int J Mol Sci 2021; 22:ijms22094856. [PMID: 34063734 PMCID: PMC8124397 DOI: 10.3390/ijms22094856] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, we report the effects of caffeine on angiogenesis in zebrafish embryos both during normal development and after exposure to Fibroblast Growth Factor 2 (FGF2). As markers of angiogenesis, we measured the length and width of intersegmental vessels (ISVs), performed whole-mount in situ hybridization with fli1 and cadh5 vascular markers, and counted the number of interconnecting vessels (ICVs) in sub-intestinal venous plexus (SIVP). In addition, we measured angiogenesis after performing zebrafish yolk membrane (ZFYM) assay with microinjection of fibroblast growth factor 2 (FGF2) and perivitelline tumor xenograft assay with microinjection of tumorigenic FGF2-overexpressing endothelial (FGF2-T-MAE) cells. The results showed that caffeine treatment causes a shortening and thinning of ISVs along with a decreased expression of the vascular marker genes and a decrease in the number of ICVs in the SIVP. Caffeine was also able to block angiogenesis induced by exogenous FGF2 or FGF2-producing cells. Overall, our results are suggestive of the inhibitory effect of caffeine in both direct and indirect angiogenesis.
Collapse
Affiliation(s)
- Ram Manohar Basnet
- Unit of Pharmacology, DMMT, University of Brescia, 25123 Brescia, Italy; (R.M.B.); (A.M.); (S.S.); (E.R.)
| | - Daniela Zizioli
- Unit of Biotechnology, DMMT, University of Brescia, 25123 Brescia, Italy; (D.Z.); (D.F.)
| | - Alessia Muscò
- Unit of Pharmacology, DMMT, University of Brescia, 25123 Brescia, Italy; (R.M.B.); (A.M.); (S.S.); (E.R.)
| | - Dario Finazzi
- Unit of Biotechnology, DMMT, University of Brescia, 25123 Brescia, Italy; (D.Z.); (D.F.)
- Laboratorio Centrale Analisi Chimico-Cliniche, ASST Spedali Civili, 25123 Brescia, Italy
| | - Sandra Sigala
- Unit of Pharmacology, DMMT, University of Brescia, 25123 Brescia, Italy; (R.M.B.); (A.M.); (S.S.); (E.R.)
| | - Elisa Rossini
- Unit of Pharmacology, DMMT, University of Brescia, 25123 Brescia, Italy; (R.M.B.); (A.M.); (S.S.); (E.R.)
| | - Chiara Tobia
- Unit of Experimental Oncology and Immunology, DMMT, University of Brescia, 25123 Brescia, Italy; (C.T.); (J.G.); (M.P.)
| | - Jessica Guerra
- Unit of Experimental Oncology and Immunology, DMMT, University of Brescia, 25123 Brescia, Italy; (C.T.); (J.G.); (M.P.)
| | - Marco Presta
- Unit of Experimental Oncology and Immunology, DMMT, University of Brescia, 25123 Brescia, Italy; (C.T.); (J.G.); (M.P.)
| | - Maurizio Memo
- Unit of Pharmacology, DMMT, University of Brescia, 25123 Brescia, Italy; (R.M.B.); (A.M.); (S.S.); (E.R.)
- Correspondence:
| |
Collapse
|
17
|
Vandetanib versus Cabozantinib in Medullary Thyroid Carcinoma: A Focus on Anti-Angiogenic Effects in Zebrafish Model. Int J Mol Sci 2021; 22:ijms22063031. [PMID: 33809722 PMCID: PMC8002338 DOI: 10.3390/ijms22063031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/20/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Medullary thyroid carcinoma (MTC) is a tumor deriving from the thyroid C cells. Vandetanib (VAN) and cabozantinib (CAB) are two tyrosine kinase inhibitors targeting REarranged during Transfection (RET) and other kinase receptors and are approved for the treatment of advanced MTC. We aim to compare the in vitro and in vivo anti-tumor activity of VAN and CAB in MTC. The effects of VAN and CAB on viability, cell cycle, and apoptosis of TT and MZ-CRC-1 cells are evaluated in vitro using an MTT assay, DNA flow cytometry with propidium iodide, and Annexin V-FITC/propidium iodide staining, respectively. In vivo, the anti-angiogenic potential of VAN and CAB is evaluated in Tg(fli1a:EGFP)y1 transgenic fluorescent zebrafish embryos by analyzing the effects on the physiological development of the sub-intestinal vein plexus and the tumor-induced angiogenesis after TT and MZ-CRC-1 xenotransplantation. VAN and CAB exert comparable effects on TT and MZ-CRC-1 viability inhibition and cell cycle perturbation, and stimulated apoptosis with a prominent effect by VAN in MZ-CRC-1 and CAB in TT cells. Regarding zebrafish, both drugs inhibit angiogenesis in a dose-dependent manner, in particular CAB shows a more potent anti-angiogenic activity than VAN. To conclude, although VAN and CAB show comparable antiproliferative effects in MTC, the anti-angiogenic activity of CAB appears to be more relevant.
Collapse
|
18
|
Yang J, Pan S, Gao S, Li T, Xu H. CO/chemosensitization/antiangiogenesis synergistic therapy with H 2O 2-responsive diselenide-containing polymer. Biomaterials 2021; 271:120721. [PMID: 33631653 DOI: 10.1016/j.biomaterials.2021.120721] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/11/2020] [Accepted: 02/11/2021] [Indexed: 02/08/2023]
Abstract
Carbon monoxide (CO) therapy and antiangiogenesis therapy (AAT) are regarded as promising approaches for cancer treatment. However, the poor tumor targeting ability and inevitable side effects prevent their clinical application. In this study, we developed H2O2-responsive diselenide-containing micelles that combined CO therapy with chemosensitization therapy and AAT in a single system. Under the interaction of intratumoral H2O2, CO and gemcitabine (GEM) were released in situ from the micelles to reduce side effects, and CO significantly sensitized the chemotherapeutic effect of GEM by elevating the level of reactive oxygen species (ROS) in human gastric cancer AGS cells. Furthermore, diselenide bonds in the micelles were oxidized to seleninic acid in organic form, which suppressed the expressions of vascular endothelial growth factor (VEGF) and matrix metalloproteinase-2 (MMP-2) to realize AAT. This study provides an integrated solution to combine CO therapy with chemosensitization therapy and AAT together with good biocompatibility.
Collapse
Affiliation(s)
- Jichun Yang
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Beijing, 100084, China
| | - Shuojiong Pan
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shiqian Gao
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Beijing, 100084, China
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Huaping Xu
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
19
|
Wu JQ, Fan RY, Zhang SR, Li CY, Shen LZ, Wei P, He ZH, He MF. A systematical comparison of anti-angiogenesis and anti-cancer efficacy of ramucirumab, apatinib, regorafenib and cabozantinib in zebrafish model. Life Sci 2020; 247:117402. [PMID: 32035930 DOI: 10.1016/j.lfs.2020.117402] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/26/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
Abstract
AIMS Gastric cancer (GC) is one of the most common malignant tumors in the world. Anti-angiogenic therapy is a useful strategy for the treatment of advanced GC. This study was aimed to systemically compare the anti-angiogenesis, anti-cancer efficacy, as well as the safety of four known anti-angiogenic drugs, namely ramucirumab, apatinib, regorafenib and cabozantinib. MAIN METHODS Anti-angiogenic effect was evaluated for the intersegmental vessels (ISVs) and subintestinal veins (SIVs) formation in the Tg (fli-1: EGFP) zebrafish embryos. Anti-cancer efficacy was tested for the in vivo cell proliferation in cell line derived tumor xenograft (CDX) model based on Tg (fli-1: EGFP) zebrafish embryos. KEY FINDINGS All four drugs exhibited anti-angiogenic abilities and tumor inhibition effects in fli-1: EGFP transgenic zebrafish. Using zebrafish xenografted model, we found that effectiveness of ramucirumab in anti-GC-proliferation is better than apatinib, regorafenib and cabozantinib. The combination of anti-angiogenic drugs and cisplatin showed no significant benefit in tumors. Meanwhile, toxicity assay showed that all tested anti-angiogenic drugs could cause cardiovascular-related side effects. The therapeutic index (LD50/ED50) of cabozantinib is higher than apatinib and regorafenib, suggesting a potential as an anti-GC drug. SIGNIFICANCE The comparison of GC-related anti-angiogenic drugs was first reported. It was found that cabozantinib had a potential as an anti-GC drug. Zebrafish model was an ideal animal model for the research of anti-angiogenic behaviors.
Collapse
Affiliation(s)
- Jia-Qi Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Ruo-Yue Fan
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Shi-Ru Zhang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Chong-Yong Li
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Li-Zong Shen
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Pin Wei
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zhi-Heng He
- School of Medicine, Yale University, New Haven CT06511, USA
| | - Ming-Fang He
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
20
|
Kalla D, Kind A, Schnieke A. Genetically Engineered Pigs to Study Cancer. Int J Mol Sci 2020; 21:E488. [PMID: 31940967 PMCID: PMC7013672 DOI: 10.3390/ijms21020488] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Recent decades have seen groundbreaking advances in cancer research. Genetically engineered animal models, mainly in mice, have contributed to a better understanding of the underlying mechanisms involved in cancer. However, mice are not ideal for translating basic research into studies closer to the clinic. There is a need for complementary information provided by non-rodent species. Pigs are well suited for translational biomedical research as they share many similarities with humans such as body and organ size, aspects of anatomy, physiology and pathophysiology and can provide valuable means of developing and testing novel diagnostic and therapeutic procedures. Porcine oncology is a new field, but it is clear that replication of key oncogenic mutation in pigs can usefully mimic several human cancers. This review briefly outlines the technology used to generate genetically modified pigs, provides an overview of existing cancer models, their applications and how the field may develop in the near future.
Collapse
Affiliation(s)
| | | | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany; (D.K.); (A.K.)
| |
Collapse
|
21
|
Anticancer activity of grassy Hystrix brachyura bezoar and its mechanisms of action: An in vitro and in vivo based study. Biomed Pharmacother 2019; 114:108841. [PMID: 30981106 DOI: 10.1016/j.biopha.2019.108841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 01/17/2023] Open
Abstract
Porcupine bezoar (PB) is a calcified undigested material generally found in porcupine's (Hystrix brachyura) gastrointestinal tract. The bezoar is traditionally used in South East Asia and Europe for the treatment of cancer, poisoning, dengue, typhoid, etc. However, limited scientific studies have been performed to verify its anticancer potential to substantiate its traditional claims in the treatment of cancers. Hence, this study was aimed at investigating the in vitro and in vivo anticancer properties of two grassy PB aqueous extract (PB-A and PB-B) using A375 cancer cell line and zebrafish model, respectively. This paper presents the first report on in vitro A375 cell viability assay, apoptosis assay, cell cycle arrest assay, migration assay, invasion assay, qPCR experimental assay and in vivo anti-angiogenesis assay using the grassy PBs. Experimental findings revealed IC50 value are 26.59 ± 1.37 μg/mL and 30.12 ± 3.25 μg/mL for PB-A and PB-B respectively. PBs showed anti-proliferative activity with no significant cytotoxic effect on normal human dermal fibroblast (NHDF). PBs were also found to induce apoptosis via intrinsic pathway and arrest cell cycle at G2/M phase. Additionally, the findings indicated its ability to debilitate migration and invasion of A375 cells. Further evaluation using embryo zebrafish model revealed LC50 = 450.0 ± 2.50 μg/mL and 58.7 ± 5.0 μg/mL for PB-A and PB-B which also exerted anti-angiogenesis effect in zebrafish. Moreover, stearic acid, ursodeoxycholic acid and pregnenolone were identified as possible metabolites that might contribute to the anticancer effect of the both PBs. Overall, this study demonstrated that PB-A and PB-B possess potential in vitro and in vivo anticancer effects which are elicited through selective cytotoxic effect, induction of apoptosis, inhibition of migration and invasion and anti-angiogenesis. This study provides scientific evidence that the porcupine bezoar do possess anti-cancer efficacy and further justifies its traditional utility. However, more experiments with higher vertebrae models are still warranted to validate its traditional claims as an anticancer agent.
Collapse
|