1
|
Kedjar N, Iannuzzi E, Kreuzer M, Alonso-Moreno C, Moya-Lopez C. Fine-Tuning the Physicochemical Properties of Poly(lactic Acid) Nanoparticles for the Controlled Release of the BET Inhibitor JQ1: Influence of PVA Concentration. Polymers (Basel) 2025; 17:123. [PMID: 39795526 PMCID: PMC11722895 DOI: 10.3390/polym17010123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
The compounds targeting the bromo and extra terminal domain proteins (BET), such as the JQ1, present potent anti-cancer activity in preclinical models, however, the application of JQ1 at the clinical level is limited by its short half-life, rapid clearance, and non-selective inhibition of BET family proteins, leading to off-target effects and resistance. To address these challenges, the optimization of JQ1 delivery has been accomplished through polylactide (PLA) nanoparticles. PLA derivatives with varying molecular weights were synthesized via ring-opening polymerization using a zinc-based initiator and characterized using thermogravimetric analysis, differential scanning calorimetry, and infrared spectroscopy. PLA nanoparticles (NPs) were subsequently formulated, and the effects of key parameters-including PLA molecular weight, organic phase concentration, and surfactant concentration-on particle size, polydispersity index (PDI), and encapsulation efficiency were systematically investigated. PLA molecular weight and organic phase concentration mainly influenced the NPs size whilst the thermodynamic state of the NPs was unaffected by these two parameters. The surfactant concentration is correlated to the encapsulation efficacy of JQ1 as well as the release profile, suggesting the potential tool that the variation of these parameters represent for customizing the release of JQ1 according to specific needs.
Collapse
Affiliation(s)
- Nedjla Kedjar
- Laboratory of Applied Chemistry (LAC), Faculty of Sciences Technology, University of Ain Temouchent Belhadj Bouchaib, Ain Temouchent 46000, Algeria;
| | - Eleonora Iannuzzi
- Facultad de Farmacia-Centro de Innovación en Química Avanzada (ORFEO-CINQA), Unidad nanoDrug, Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha, 02071 Albacete, Albacete, Spain;
| | - Martin Kreuzer
- ALBA Synchrotron, Carrer de la Llum 2-26, 08290 Cerdanyola del Vallès, Barcelona, Spain
| | - Carlos Alonso-Moreno
- Facultad de Farmacia-Centro de Innovación en Química Avanzada (ORFEO-CINQA), Unidad nanoDrug, Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha, 02071 Albacete, Albacete, Spain;
| | - Carmen Moya-Lopez
- Facultad de Farmacia-Centro de Innovación en Química Avanzada (ORFEO-CINQA), Unidad nanoDrug, Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha, 02071 Albacete, Albacete, Spain;
| |
Collapse
|
2
|
Wang X, Huang H, Xu W, Gong Y, Shi S, Wan X, Li P. TGF-β1 and FOXM1 siRNA co-loaded nanoparticles by disulfide crosslinked PEG-PDMAEMA for the treatment of triple-negative breast cancer and its bone metastases in vitro. Drug Dev Ind Pharm 2024:1-12. [PMID: 39286903 DOI: 10.1080/03639045.2024.2404979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is characterized by higher malignancy and mortality and is prone to distant metastasis, among which bone is the most common site. It's urgent to explore new strategies for the treatment of TNBC and its bone metastases. METHODS A tumor environment responsive vector, poly-(dimethylaminoethyl methacrylate)-SS-poly(ethylene glycol)-SS-poly-(dimethylaminoethyl methacrylate) (PDMAEMA-SS-PEG-SS-PDMAEMA), was constructed to co-delivery transforming growth factor-β1 (TGF-β1) siRNA and forkhead box M1 (FOXM1) siRNA in MDA-MB-231 cells. The preparation, characterization, in vitro release, stability, and transfection efficiency of nanoparticles were measured. Cell viability, migration, and invasion of MDA-MB-231 cells were determined. Cell chemotactic migration and cell heterogeneity adhesion of MDA-MB-231 cells to the human osteoblast-like cell line MG-63 were determined. RESULTS PDMAEMA-SS-PEG-SS-PDMAEMA self-assembled with siRNA at N/P of 15:1 into nanoparticles with a particle size of 122 nm. In vitro release exhibited redox and pH sensitivity, and the nanoparticles protected siRNA from degradation by RNase and serum protein, remaining stable at 4 °C with similar transfection efficiency with lipo2000. Nanoparticles co-loaded with TGF-β1 siRNA and FOXM1 siRNA inhibited the cell viability, migration and invasion of MDA-MB-231 cells, as well as chemotactic migration and heterogeneous adhesion of MDA-MB-231 cells to MG-63 cells, showing a synergetic effect. After gene silencing on TGF-β1 and FOXM1, the epithelial to mesenchymal transition (EMT) related molecules vimentin mRNA expression decreased while E-cadherin increased. CONCLUSIONS PDMAEMA-SS-PEG-SS-PDMAEMA was suitable for TGF-β1 siRNA and FOXM1 siRNA delivery, exhibiting a synergetic inhibition effect on TNBC and its bone metastases, which might be related to its synergetic inhibition on EMT.
Collapse
Affiliation(s)
- Xingbo Wang
- Department of Orthopedics, Gansu Provincial Hospital, Lanzhou, China
| | - Hong Huang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Wenxiu Xu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yanling Gong
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Songbo Shi
- Department of Orthopedics, Gansu Provincial Hospital, Lanzhou, China
| | - Xu Wan
- Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China Shanghai
| | - Pengbiao Li
- Department of Orthopedics, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
3
|
Saiz ML, Lozano-Chamizo L, Florez AB, Marciello M, Diaz-Bulnes P, Corte-Iglesias V, Bernet CR, Rodrigues-Diez RR, Martin-Martin C, Rodriguez-Santamaria M, Fernandez-Vega I, Rodriguez RM, Diaz-Corte C, Suarez-Alvarez B, Filice M, Lopez-Larrea C. BET inhibitor nanotherapy halts kidney damage and reduces chronic kidney disease progression after ischemia-reperfusion injury. Biomed Pharmacother 2024; 174:116492. [PMID: 38537579 DOI: 10.1016/j.biopha.2024.116492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
Targeting epigenetic mechanisms has emerged as a potential therapeutic approach for the treatment of kidney diseases. Specifically, inhibiting the bromodomain and extra-terminal (BET) domain proteins using the small molecule inhibitor JQ1 has shown promise in preclinical models of acute kidney injury (AKI) and chronic kidney disease (CKD). However, its clinical translation faces challenges due to issues with poor pharmacokinetics and side effects. Here, we developed engineered liposomes loaded with JQ1 with the aim of enhancing kidney drug delivery and reducing the required minimum effective dose by leveraging cargo protection. These liposomes efficiently encapsulated JQ1 in both the membrane and core, demonstrating superior therapeutic efficacy compared to freely delivered JQ1 in a mouse model of kidney ischemia-reperfusion injury. JQ1-loaded liposomes (JQ1-NPs) effectively targeted the kidneys and only one administration, one-hour after injury, was enough to decrease the immune cell (neutrophils and monocytes) infiltration to the kidney-an early and pivotal step to prevent damage progression. By inhibiting BRD4, JQ1-NPs suppress the transcription of pro-inflammatory genes, such as cytokines (il-6) and chemokines (ccl2, ccl5). This success not only improved early the kidney function, as evidenced by decreased serum levels of BUN and creatinine in JQ1-NPs-treated mice, along with reduced tissue expression of the damage marker, NGAL, but also halted the production of extracellular matrix proteins (Fsp-1, Fn-1, α-SMA and Col1a1) and the fibrosis development. In summary, this work presents a promising nanotherapeutic strategy for AKI treatment and its progression and provides new insights into renal drug delivery.
Collapse
Affiliation(s)
- Maria Laura Saiz
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Laura Lozano-Chamizo
- Nanobiotechnology for Life Sciences Laboratory, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, Madrid E-28040, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernández Almagro 3, Madrid E-28029, Spain; Atrys Health, Madrid E-28001, Spain
| | - Aida Bernardo Florez
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Marzia Marciello
- Nanobiotechnology for Life Sciences Laboratory, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, Madrid E-28040, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernández Almagro 3, Madrid E-28029, Spain
| | - Paula Diaz-Bulnes
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Viviana Corte-Iglesias
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain; Department of Immunology, Hospital Universitario Central de Asturias, Oviedo 33011, Spain
| | - Cristian Ruiz Bernet
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Raul R Rodrigues-Diez
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Cristina Martin-Martin
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain
| | - Mar Rodriguez-Santamaria
- Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain
| | - Ivan Fernandez-Vega
- Department of Pathology, Hospital Universitario Central de Asturias, Oviedo 33001, Spain; Biobank of Principality of Asturias, Oviedo 33011, Spain
| | - Ramon M Rodriguez
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa), Ctra. Valldemossa 79, Palma, Balearic Islands E-07120, Spain; Research Unit, University Hospital Son Espases, Ctra. Valldemossa79, Palma, Balearic Islands E-07120, Spain
| | - Carmen Diaz-Corte
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; Department of Nephrology, Hospital Universitario Central de Asturias, Oviedo 33001, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain.
| | - Marco Filice
- Nanobiotechnology for Life Sciences Laboratory, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, Madrid E-28040, Spain; Microscopy and Dynamic Imaging Unit, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle Melchor Fernández Almagro 3, Madrid E-28029, Spain.
| | - Carlos Lopez-Larrea
- Translational Immunology, Health Research Institute of the Principality of Asturias (ISPA), Avenida de Roma S/N, Oviedo, Asturias 33011, Spain; ISCIII RICORS2040 Kidney Disease Research Network, Madrid, Spain; Department of Immunology, Hospital Universitario Central de Asturias, Oviedo 33011, Spain
| |
Collapse
|
4
|
Masoudi E, Soleimani M, Zarinfard G, Homayoun M, Bakhtiari M. The effects of chitosan-loaded JQ1 nanoparticles on OVCAR-3 cell cycle and apoptosis-related gene expression. Res Pharm Sci 2024; 19:53-63. [PMID: 39006975 PMCID: PMC11244706 DOI: 10.4103/1735-5362.394820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/14/2023] [Accepted: 01/02/2024] [Indexed: 07/16/2024] Open
Abstract
Background and purpose Ovarian cancer is the deadliest gynecological cancer. Bromodomain and extra terminal domain (BET) proteins play major roles in the regulation of gene expression at the epigenetic level. Jun Qi (JQ1) is a potent inhibitor of BET proteins. Regarding the short half-life and poor pharmacokinetic profile, JQ1 was loaded into newly developed nano-carriers. Chitosan nanoparticles are one of the best and potential polymers in cancer treatment. The present study aimed to build chitosan-JQl nanoparticles (Ch-J-NPs), treat OVCAR-3 cells with Ch-J-NPs, and evaluate the effects of these nanoparticles on cell cycle and apoptosis-associated genes. Experimental approach Ch-J-NPs were synthesized and characterized. The size and morphology of Ch-J-NPs were defined by DLS and FE-SEM techniques. OVCAR-3 cells were cultured and treated with Ch-J-NPs. Then, IC50 was measured using MTT assay. The groups were defined and cells were treated with IC50 concentration of Ch-J-NPs, for 48 h. Finally, cells in different groups were assessed for the expression of genes of interest using quantitative RT-PCR. Findings/Results IC50 values for Ch-J-NPs were 5.625 μg/mL. RT-PCR results demonstrated that the expression of genes associated with cell cycle activity (c-MYC, hTERT, CDK1, CDK4, and CDK6) was significantly decreased following treatment of cancer cells with Ch-J-NPs. Conversely, the expression of caspase-3, and caspase-9 significantly increased. BAX (pro-apoptotic) to BCL2 (anti-apoptotic) expression ratio, also increased significantly after treatment of cells with Ch-J-NPs. Conclusion and implications Ch-J-NPs showed significant anti-cell cyclic and apoptotic effects on OVCAR-3 cells.
Collapse
Affiliation(s)
- Ehsan Masoudi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mitra Soleimani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Giti Zarinfard
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Homayoun
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bakhtiari
- Department of Anatomical Sciences, School of Medicine, Behbahan University of Medical Sciences, Behbahan, Iran
| |
Collapse
|
5
|
Yang L, Jing Y, Xia X, Yin X. ARV-825 Showed Antitumor Activity against BRD4-NUT Fusion Protein by Targeting the BRD4. JOURNAL OF ONCOLOGY 2023; 2023:9904143. [PMID: 38130463 PMCID: PMC10735731 DOI: 10.1155/2023/9904143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 10/07/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
Objective The bromodomain-containing 4 (BRD4) is a member of the bromodomain and extra terminal domain (BET) family, which is an important epigenetic reader. It is currently a promising oncology target. In some tumors, BET bromodomain inhibitors have demonstrated promising results. Proteolysis-targeting methods (PROTAC), which rapidly and effectively degrade BRD4, have displayed considerable potential in the treatment of tumors in recent years. The purpose of this study is to examine the potential impact of BRD4 PROTAC compounds ARV-825 on oncogene BRD4-NUT fused protein in NUT carcinoma. Methods The effectiveness of ARV-825 was evaluated at the cellular level using the cell counting kit 8 test, wound healing, cell transfection, western blotting analysis, and RNA sequencing. The effectiveness of ARV-825 was also examined in vivo using a xenograft model. Results The BRD4-NUT fusion gene was overexpressed in 3T3 cells, and the pathogenic fusion gene was simulated. The results showed that the overexpression of BRD4-NUT could promote the proliferation and migration of 3T3 cells, but the expression of BRD4 protein was degraded after the addition of the novel cereblon-based PROTAC compound ARV-825 against BRD4, resulting in inhibition of BRD4-NUT 3T3 cell proliferation and migration. Further RNA-seq analysis showed that overexpression of BRD4-NUT was accompanied by increased expression of gene (e.g., Myc, E2F, TRAFs, Wnt, Gadd45g, and Sox6) with significantly enriched pathway (e.g., small cell lung cancer, NF-kappa B signaling pathway, and breast cancer), promoted cell cycle from G 1 phase to S phase, and increased cell proliferation and migration, activated the antiapoptosisi signal, led to abnormal cell growth, and ultimately led to tumorigenesis. The addition of ARV-825 effectively rescued this process and effectively inhibited cell vitality, proliferation, and migration. In vivo studies demonstrated that treatment with ARV-825 greatly suppressed tumor growth without causing harmful side effects and downregulated the BRD4-NUT expression level. Conclusion Through the induction of BRD4 protein degradation, ARV-825 can successfully limit BRD4-NUT 3T3 cell proliferation in vitro and in vivo. These findings suggested that the BRD4 inhibitor ARV-825 would be an effective therapeutic strategy for treating NUT carcinoma that with the genetic feature of BRD4-NUT fusion event.
Collapse
Affiliation(s)
- Liu Yang
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Yue Jing
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Xia Xia
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Roc Rock Biotechnology (Shenzhen), Shenzhen 518118, China
| |
Collapse
|
6
|
Gagliardi A, Voci S, Ambrosio N, Fresta M, Duranti A, Cosco D. Characterization and Preliminary In Vitro Antioxidant Activity of a New Multidrug Formulation Based on the Co-Encapsulation of Rutin and the α-Acylamino-β-Lactone NAAA Inhibitor URB894 within PLGA Nanoparticles. Antioxidants (Basel) 2023; 12:antiox12020305. [PMID: 36829864 PMCID: PMC9951992 DOI: 10.3390/antiox12020305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
A biodegradable and biocompatible polymeric matrix made up of poly(d,l-lactide-co-glycolide) (PLGA) was used for the simultaneous delivery of rutin and the (S)-N-(2-oxo-3-oxetanyl)biphenyl-4-carboxamide derivative (URB894). The goal was to exploit the well-known radical scavenging properties of rutin and the antioxidant features recently reported for the molecules belonging to the class of N-acylethanolamine-hydrolyzing acid amidase (NAAA) inhibitors, such as URB894. The use of the compounds, both as single agents or in association promoted the development of negatively-charged nanosystems characterized by a narrow size distribution and an average diameter of ~200 nm when 0.2-0.6 mg/mL of rutin or URB894 were used. The obtained multidrug carriers evidenced an entrapment efficiency of ~50% and 40% when 0.4 and 0.6 mg/mL of rutin and URB894 were associated during the sample preparation, respectively. The multidrug formulation evidenced an improved in vitro dose-dependent protective effect against H2O2-related oxidative stress with respect to that of the nanosystems containing the active compounds as a single agent, confirming the rationale of using the co-encapsulation approach to obtain a novel antioxidant nanomedicine.
Collapse
Affiliation(s)
- Agnese Gagliardi
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, 88100 Catanzaro, Italy
| | - Silvia Voci
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, 88100 Catanzaro, Italy
| | - Nicola Ambrosio
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, 88100 Catanzaro, Italy
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, 88100 Catanzaro, Italy
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento 6, 61029 Urbino, Italy
- Correspondence: (A.D.); (D.C.); Tel.: +39-0722-303501 (A.D.); +39-0961-3694119 (D.C.)
| | - Donato Cosco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, 88100 Catanzaro, Italy
- Correspondence: (A.D.); (D.C.); Tel.: +39-0722-303501 (A.D.); +39-0961-3694119 (D.C.)
| |
Collapse
|
7
|
Celano M, Gagliardi A, Maggisano V, Ambrosio N, Bulotta S, Fresta M, Russo D, Cosco D. Co-Encapsulation of Paclitaxel and JQ1 in Zein Nanoparticles as Potential Innovative Nanomedicine. MICROMACHINES 2022; 13:1580. [PMID: 36295933 PMCID: PMC9609127 DOI: 10.3390/mi13101580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
The manuscript describes the development of zein nanoparticles containing paclitaxel (PTX) and the bromo-and extra-terminal domain inhibitor (S)-tertbutyl2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno(3,2-f)(1,2,4)triazolo(4,3-a)(1,4)diazepin-6-yl)acetate (JQ1) together with their cytotoxicity on triple-negative breast cancer cells. The rationale of this association is that of exploiting different types of cancer cells as targets in order to obtain increased pharmacological activity with respect to that exerted by the single agents. Zein, a protein found in the endosperm of corn, was used as a biomaterial to obtain multidrug carriers characterized by mean sizes of ˂200 nm, a low polydispersity index (0.1-0.2) and a negative surface charge. An entrapment efficiency of ~35% of both the drugs was obtained when 0.3 mg/mL of the active compounds were used during the nanoprecipitation procedure. No adverse phenomena such as sedimentation, macro-aggregation or flocculation occurred when the nanosystems were heated to 37 °C. The multidrug nanoformulation demonstrated significant in vitro cytototoxic activity against MDA-MB-157 and MDA-MB-231 cancer cells by MTT-test and adhesion assay which was stronger than that of the compounds encapsulated as single agents. The results evidence the potential application of zein nanoparticles containing PTX and JQ1 as a novel nanomedicine.
Collapse
Affiliation(s)
- Marilena Celano
- Correspondence: (M.C.); (D.C.); Tel.: +39-0961-369-4099 (M.C.); +39-0961-369-4119 (D.C.)
| | | | | | | | | | | | | | - Donato Cosco
- Correspondence: (M.C.); (D.C.); Tel.: +39-0961-369-4099 (M.C.); +39-0961-369-4119 (D.C.)
| |
Collapse
|
8
|
Enhanced Antitumoral Activity of Encapsulated BET Inhibitors When Combined with PARP Inhibitors for the Treatment of Triple-Negative Breast and Ovarian Cancers. Cancers (Basel) 2022; 14:cancers14184474. [PMID: 36139634 PMCID: PMC9496913 DOI: 10.3390/cancers14184474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Poly (adenosine diphosphate ribose) polymerase inhibitors (PARPis) have demonstrated antitumoral activity in several cancers harbouring germline and somatic BRCA1/2 mutations. The widespread use of these agents in clinical practice is restricted by the development of acquired resistance due to the presence of compensatory pathways. A strategy to deal with this is the use of combination therapies with drugs that act synergistically against the tumour. BETis can completely disrupt the HR pathway by repressing the expression of BRCA1 and could be aimed at generation combination regimes to overcome PARPi resistance and enhance PARPi efficacy. However, this strategy is hampered by the poor pharmacokinetic profile and short half-life of BETis. In this work and as a proof of concept, we discuss the potential preclinical benefit provided by the combination of the PARPi olaparib and the BET inhibitor JQ1 encapsulated into nanoparticles for the treatment of BRCAness tumours. Abstract BRCA1/2 protein-deficient or mutated cancers comprise a group of aggressive malignancies. Although PARPis have shown considerably efficacy in their treatment, the widespread use of these agents in clinical practice is restricted by various factors, including the development of acquired resistance due to the presence of compensatory pathways. BETis can completely disrupt the HR pathway by repressing the expression of BRCA1 and could be aimed at generation combination regimes to overcome PARPi resistance and enhance PARPi efficacy. Due to the poor pharmacokinetic profile and short half-life, the first-in-class BETi JQ1 was loaded into newly developed nanocarrier formulations to improve the effectivity of olaparib for the treatment of BRCAness cancers. First, polylactide polymeric nanoparticles were generated by double emulsion. Moreover, liposomes were prepared by ethanol injection and evaporation solvent method. JQ1-loaded drug delivery systems display optimal hydrodynamic radii between 60 and 120 nm, with a very low polydispersity index (PdI), and encapsulation efficiencies of 92 and 16% for lipid- and polymeric-based formulations, respectively. Formulations show high stability and sustained release. We confirmed that all assayed JQ1 formulations improved antiproliferative activity compared to the free JQ1 in models of ovarian and breast cancers. In addition, synergistic interaction between JQ1 and JQ1-loaded nanocarriers and olaparib evidenced the ability of encapsulated JQ1 to enhance antitumoral activity of PARPis.
Collapse
|
9
|
α-Acylamino-β-lactone N-Acylethanolamine-hydrolyzing Acid Amidase Inhibitors Encapsulated in PLGA Nanoparticles: Improvement of the Physical Stability and Protection of Human Cells from Hydrogen Peroxide-Induced Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11040686. [PMID: 35453371 PMCID: PMC9028182 DOI: 10.3390/antiox11040686] [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: 02/16/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022] Open
Abstract
N-Acylethanolamine acid amidase (NAAA) is an N-terminal cysteine hydrolase that preferentially catalyzes the hydrolysis of endogenous lipid mediators such as palmitoylethanolamide, which has been shown to exhibit neuroprotective and antinociceptive properties by engaging peroxisome proliferator-activated receptor-α. A few potent NAAA inhibitors have been developed, including α-acylamino-β-lactone derivatives, which are very strong and effective, but they have limited chemical and plasmatic stability, compromising their use as systemic agents. In the present study, as an example of a molecule belonging to the chemical class of N-(2-oxo-3-oxetanyl)amide NAAA inhibitors, URB866 was entrapped in poly(lactic-co-glycolic acid) nanoparticles in order to increase its physical stability. The data show a monomodal pattern and a significant time- and temperature-dependent stability of the molecule-loaded nanoparticles, which also demonstrated a greater ability to effectively retain the compound. The nanoparticles improved the photostability of URB866 with respect to that of the free molecule and displayed a better antioxidant profile on various cell lines at the molecule concentration of 25 μM. Overall, these results prove that the use of polymeric nanoparticles could be a useful strategy for overcoming the instability of α-acylamino-β-lactone NAAA inhibitors, allowing the maintenance of their characteristics and activity for a longer time.
Collapse
|
10
|
Liao X, Qian X, Zhang Z, Tao Y, Li Z, Zhang Q, Liang H, Li X, Xie Y, Zhuo R, Chen Y, Jiang Y, Cao H, Niu J, Xue C, Ni J, Pan J, Cui D. ARV-825 Demonstrates Antitumor Activity in Gastric Cancer via MYC-Targets and G2M-Checkpoint Signaling Pathways. Front Oncol 2021; 11:753119. [PMID: 34733788 PMCID: PMC8559897 DOI: 10.3389/fonc.2021.753119] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/27/2021] [Indexed: 01/20/2023] Open
Abstract
Objective Suppression of bromodomain and extra terminal (BET) proteins has a bright prospect to treat MYC-driven tumors. Bromodomain containing 4 (BRD4) is one of the BET proteins. ARV-825, consisting of a BRD4 inhibitor conjugated with a cereblon ligand using proteolysis-targeting chimera (PROTAC) technology, was proven to decrease the tumor growth effectively and continuously. Nevertheless, the efficacy and mechanisms of ARV-825 in gastric cancer are still poorly understood. Methods Cell counting kit 8 assay, lentivirus infection, Western blotting analysis, Annexin V/propidium iodide (PI) staining, RNA sequencing, a xenograft model, and immunohistochemistry were used to assess the efficacy of ARV-825 in cell level and animal model. Results The messenger RNA (mRNA) expression of BRD4 in gastric cancer raised significantly than those in normal tissues, which suggested poor outcome of patients with gastric cancer. ARV-825 displayed higher anticancer efficiency in gastric cancer cells than OTX015 and JQ1. ARV-825 could inhibit cell growth, inducing cell cycle block and apoptosis in vitro. ARV-825 induced degradation of BRD4, BRD2, BRD3, c-MYC, and polo-like kinase 1 (PLK1) proteins in four gastric cancer cell lines. In addition, cleavage of caspase 3 and poly-ADP-ribose polymerase (PARP) was elevated. Knockdown or overexpression CRBN could increase or decrease, respectively, the ARV-825 IC50 of gastric cancer cells. ARV-825 reduced MYC and PLK1 expression in gastric cancer cells. ARV-825 treatment significantly reduced tumor growth without toxic side effects and downregulated the expression of BRD4 in vivo. Conclusions High mRNA expression of BRD4 in gastric cancer indicated poor prognosis. ARV-825, a BRD4 inhibitor, could effectively suppress the growth and elevate the apoptosis of gastric cancer cells via transcription downregulation of c-MYC and PLK1. These results implied that ARV-825 could be a good therapeutic strategy to treat gastric cancer.
Collapse
Affiliation(s)
- Xinmei Liao
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqing Qian
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yanfang Tao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Qian Zhang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Liang
- Institute of Nanomedicine, National Engineering Research Centre for Nanotechnology, Shanghai, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yi Xie
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Ran Zhuo
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yanling Chen
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - You Jiang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Haibo Cao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jiaqi Niu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Cuili Xue
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Ni
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Institute of Nanomedicine, National Engineering Research Centre for Nanotechnology, Shanghai, China
| |
Collapse
|
11
|
Zhang Z, Zhang Q, Xie J, Zhong Z, Deng C. Enzyme-responsive micellar JQ1 induces enhanced BET protein inhibition and immunotherapy of malignant tumors. Biomater Sci 2021; 9:6915-6926. [PMID: 34524279 DOI: 10.1039/d1bm00724f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bromodomain and extra-terminal (BET) proteins are attractive targets for treating various malignancies including melanoma. The inhibition of BET bromodomains, e.g. with JQ1, is found to downregulate the expression of both c-MYC oncoprotein and programmed cell death ligand 1 (PD-L1), which play a crucial role in tumor growth and the immunosuppressive tumor microenvironment, respectively. The BET bromodomain inhibitors like JQ1 though exhibiting high selectivity and affinity show usually low bioavailability and efficacy in vivo due to fast clearance and inferior uptake by tumor cells. The therapeutic effect of JQ1 might further be lowered by drug resistance. Here, enzyme-responsive micellar JQ1 (mJQ1) was fabricated from a poly(ethylene glycol)-b-poly(L-tyrosine) copolypeptide to enhance JQ1 delivery and the immunotherapy of malignant melanoma. The in vitro results showed that mJQ1 induced clearly better repression of c-MYC and PD-L1 proteins, cell cycle arrest, cell inhibition, and apoptotic activity than free JQ1 in B16F10 cancer cells. The intratumoral administration of mJQ1 at 2.5 mg of JQ1 equiv. per kg was found to show better inhibition of B16F10 tumors in C57BL/6 mice than the intraperitoneal administration of free JQ1 at 50 mg kg-1. In particular, when combined with radiotherapy, mJQ1 effectively suppressed tumor growth and brought about strong local and systemic antitumor immunity as evidenced by elevated CD8+ T cells and increased ratios of CD8+ T cells to Tregs, affording significantly improved survival of B16F10 tumor-bearing mice than their JQ1 counterparts and marked growth suppression of distant tumors. The great potency of enzyme-responsive micellar JQ1 makes it interesting for immunotherapy of various tumors.
Collapse
Affiliation(s)
- Zhenqi Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Qiang Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Jiguo Xie
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Chao Deng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
12
|
Li W, Wu H, Sui S, Wang Q, Xu S, Pang D. Targeting Histone Modifications in Breast Cancer: A Precise Weapon on the Way. Front Cell Dev Biol 2021; 9:736935. [PMID: 34595180 PMCID: PMC8476812 DOI: 10.3389/fcell.2021.736935] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/16/2021] [Indexed: 12/27/2022] Open
Abstract
Histone modifications (HMs) contribute to maintaining genomic stability, transcription, DNA repair, and modulating chromatin in cancer cells. Furthermore, HMs are dynamic and reversible processes that involve interactions between numerous enzymes and molecular components. Aberrant HMs are strongly associated with tumorigenesis and progression of breast cancer (BC), although the specific mechanisms are not completely understood. Moreover, there is no comprehensive overview of abnormal HMs in BC, and BC therapies that target HMs are still in their infancy. Therefore, this review summarizes the existing evidence regarding HMs that are involved in BC and the potential mechanisms that are related to aberrant HMs. Moreover, this review examines the currently available agents and approved drugs that have been tested in pre-clinical and clinical studies to evaluate their effects on HMs. Finally, this review covers the barriers to the clinical application of therapies that target HMs, and possible strategies that could help overcome these barriers and accelerate the use of these therapies to cure patients.
Collapse
Affiliation(s)
- Wei Li
- Harbin Medical University Third Hospital: Tumor Hospital of Harbin Medical University, Harbin, China
| | - Hao Wu
- Harbin Medical University Third Hospital: Tumor Hospital of Harbin Medical University, Harbin, China
| | - Shiyao Sui
- Harbin Medical University Third Hospital: Tumor Hospital of Harbin Medical University, Harbin, China
| | - Qin Wang
- Harbin Medical University Third Hospital: Tumor Hospital of Harbin Medical University, Harbin, China
| | - Shouping Xu
- Harbin Medical University Third Hospital: Tumor Hospital of Harbin Medical University, Harbin, China
| | - Da Pang
- Harbin Medical University Third Hospital: Tumor Hospital of Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| |
Collapse
|
13
|
Gene Transcription as a Therapeutic Target in Leukemia. Int J Mol Sci 2021; 22:ijms22147340. [PMID: 34298959 PMCID: PMC8304797 DOI: 10.3390/ijms22147340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022] Open
Abstract
Blood malignancies often arise from undifferentiated hematopoietic stem cells or partially differentiated stem-like cells. A tight balance of multipotency and differentiation, cell division, and quiescence underlying normal hematopoiesis requires a special program governed by the transcriptional machinery. Acquisition of drug resistance by tumor cells also involves reprogramming of their transcriptional landscape. Limiting tumor cell plasticity by disabling reprogramming of the gene transcription is a promising strategy for improvement of treatment outcomes. Herein, we review the molecular mechanisms of action of transcription-targeted drugs in hematological malignancies (largely in leukemia) with particular respect to the results of clinical trials.
Collapse
|
14
|
Shibasaki H, Kinoh H, Cabral H, Quader S, Mochida Y, Liu X, Toh K, Miyano K, Matsumoto Y, Yamasoba T, Kataoka K. Efficacy of pH-Sensitive Nanomedicines in Tumors with Different c-MYC Expression Depends on the Intratumoral Activation Profile. ACS NANO 2021; 15:5545-5559. [PMID: 33625824 DOI: 10.1021/acsnano.1c00364] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Effective inhibition of the protein derived from cellular myelocytomatosis oncogene (c-Myc) is one of the most sought-after goals in cancer therapy. While several c-Myc inhibitors have demonstrated therapeutic potential, inhibiting c-Myc has proven challenging, since c-Myc is essential for normal tissues and tumors may present heterogeneous c-Myc levels demanding contrasting therapeutic strategies. Herein, we developed tumor-targeted nanomedicines capable of treating both tumors with high and low c-Myc levels by adjusting their ability to spatiotemporally control drug action. These nanomedicines loaded homologues of the bromodomain and extraterminal (BET) motif inhibitor JQ1 as epigenetic c-Myc inhibitors through pH-cleavable bonds engineered for fast or slow drug release at intratumoral pH. In tumors with high c-Myc expression, the fast-releasing (FR) nanomedicines suppressed tumor growth more effectively than the slow-releasing (SR) ones, whereas, in the low c-Myc tumors, the efficacy of the nanomedicines was the opposite. By studying the tumor distribution and intratumoral activation of the nanomedicines, we found that, despite SR nanomedicines achieved higher accumulation than the FR counterparts in both c-Myc high and low tumors, the antitumor activity profiles corresponded with the availability of activated drugs inside the tumors. These results indicate the potential of engineered nanomedicines for c-Myc inhibition and spur the idea of precision pH-sensitive nanomedicine based on cancer biomarker levels.
Collapse
Affiliation(s)
- Hitoshi Shibasaki
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Hiroaki Kinoh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Xueying Liu
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuki Miyano
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Otorhinolaryngology, Tokyo Yamate Medical Center, 3-22-1, Hyakunin-cho, Shinjuku-ku, Tokyo 169-0073, Japan
| | - Yu Matsumoto
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Policy Alternative Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
15
|
d'Avanzo N, Torrieri G, Figueiredo P, Celia C, Paolino D, Correia A, Moslova K, Teesalu T, Fresta M, Santos HA. LinTT1 peptide-functionalized liposomes for targeted breast cancer therapy. Int J Pharm 2021; 597:120346. [DOI: 10.1016/j.ijpharm.2021.120346] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
|
16
|
Gagliardi A, Giuliano E, Venkateswararao E, Fresta M, Bulotta S, Awasthi V, Cosco D. Biodegradable Polymeric Nanoparticles for Drug Delivery to Solid Tumors. Front Pharmacol 2021; 12:601626. [PMID: 33613290 PMCID: PMC7887387 DOI: 10.3389/fphar.2021.601626] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Advances in nanotechnology have favored the development of novel colloidal formulations able to modulate the pharmacological and biopharmaceutical properties of drugs. The peculiar physico-chemical and technological properties of nanomaterial-based therapeutics have allowed for several successful applications in the treatment of cancer. The size, shape, charge and patterning of nanoscale therapeutic molecules are parameters that need to be investigated and modulated in order to promote and optimize cell and tissue interaction. In this review, the use of polymeric nanoparticles as drug delivery systems of anticancer compounds, their physico-chemical properties and their ability to be efficiently localized in specific tumor tissues have been described. The nanoencapsulation of antitumor active compounds in polymeric systems is a promising approach to improve the efficacy of various tumor treatments.
Collapse
Affiliation(s)
- Agnese Gagliardi
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Elena Giuliano
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Eeda Venkateswararao
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Stefania Bulotta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Donato Cosco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
17
|
Gagliardi A, Paolino D, Costa N, Fresta M, Cosco D. Zein- vs PLGA-based nanoparticles containing rutin: A comparative investigation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111538. [DOI: 10.1016/j.msec.2020.111538] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/19/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
|
18
|
Minko T. Nanoformulation of BRD4-Degrading PROTAC: Improving Druggability To Target the 'Undruggable' MYC in Pancreatic Cancer. Trends Pharmacol Sci 2020; 41:684-686. [PMID: 32893006 DOI: 10.1016/j.tips.2020.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
In a recent study, Saraswat and colleagues identified a novel proteolysis targeting chimera (PROTAC), ARV-825 (ARV), that efficiently degrades bromodomain-containing protein 4 (BRD4) to drug the 'undruggable' MYC in pancreatic cancer. ARV-loaded polyethylene glycol-poly lactic acid-co-glycolic acid (PLGA-PEG) polymeric nanoparticles (ARV-NPs) showed promising anticancer activity in both 2D cell culture and 3D multicellular tumor spheroid models of pancreatic cancer. This study demonstrates a unique therapeutic strategy in which targeting BRD4 for degradation via the E3 ubiquitin ligase cereblon (CRBN) pathway leads to sustained inhibition of oncogenic MYC expression for effective treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
19
|
Andrikopoulou A, Liontos M, Koutsoukos K, Dimopoulos MA, Zagouri F. The emerging role of BET inhibitors in breast cancer. Breast 2020; 53:152-163. [PMID: 32827765 PMCID: PMC7451423 DOI: 10.1016/j.breast.2020.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/13/2020] [Accepted: 08/10/2020] [Indexed: 01/10/2023] Open
Abstract
Bromodomain and extraterminal domain (BET) proteins are epigenetic molecules that regulate the expression of multiple genes involved in carcinogenesis. Breast cancer is an heterogenous disease emerging from aberrant gene expression and epigenetic alteration patterns. Amplification or overexpression of BET proteins has been identified in breast tumors highlighting their clinical significance. Development of BET inhibitors that disrupt BET protein binding to acetylated lysine residues of chromatin and suppress transcription of various oncogenes has shown promising results in breast cancer cells and xenograft models. Currently, Phase I/II clinical trials explore safety and efficacy of BET inhibitors in solid tumors and breast cancer. Treatment-emergent toxicities have been reported, including thrombocytopenia and gastrointestinal disorders. Preliminary results demonstrated greater response rates to BET inhibitors in combination with already approved anticancer agents. Consistently, BET inhibition sensitized breast tumors to chemotherapy drugs, hormone therapy and PI3K inhibitors in vitro. This article aims to review all existing preclinical and clinical evidence regarding BET inhibitors in breast cancer.
Collapse
Affiliation(s)
- Angeliki Andrikopoulou
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Michalis Liontos
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Konstantinos Koutsoukos
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Meletios-Athanasios Dimopoulos
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Flora Zagouri
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
20
|
Zhang MY, Liu SL, Huang WL, Tang DB, Zheng WW, Zhou N, Zhou H, Abudureheman T, Tang ZH, Zhou BBS, Duan CW. Bromodomains and Extra-Terminal (BET) Inhibitor JQ1 Suppresses Proliferation of Acute Lymphocytic Leukemia by Inhibiting c-Myc-Mediated Glycolysis. Med Sci Monit 2020; 26:e923411. [PMID: 32266878 PMCID: PMC7165247 DOI: 10.12659/msm.923411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Acute lymphocytic leukemia (ALL) is a common blood cancer which induces high mortality in children. Bromodomains and extra-terminal (BET) protein inhibitors, such as JQ1 and ARV-825, are promising cancer therapeutic agents that can be used by targeting c-Myc. A recent work reported that JQ1 effectively attenuates ALL in vitro by suppressing cell proliferation and accelerating apoptosis. The purpose of this research was to probe into the potential mechanism of how JQ1 inhibits ALL cell proliferation in vitro. Material/Methods Cell viability of ALL cells were measured by CTG after treatment by JQ1. Cell cycle analysis was done by EdU and PI staining. Cell apoptosis was assessed by Annexin V/PI staining. Glycolysis was detected using Seahorse and LC-MS kits. The expression of glycolytic rate-limiting enzymes was assessed by RNA-seq, qRT-PCR, and Western blot. Results JQ1 suppressed cell proliferation by arresting the cell cycle and inducing the apoptosis of acute lymphocytic leukemia cells. JQ1 inhibited cell proliferation of B-ALL cells by restraining glycolysis. Conversely, the cell cycle block of B-ALL cells induced by JQ1 was partially abolished after pretreatment with 2-Deoxy-D-glucose (2-DG), an inhibitor of glycolysis. Furthermore, JQ1 restrained the glycolysis of B-ALL cell lines by remarkably downregulating the rate-limiting enzymes of glycolysis, such as hexokinase 2, phosphofructokinase, and lactate dehydrogenase A. Moreover, the cell cycle arrest was reversed in B-ALL cells with overexpressed c-Myc treated by JQ1, which is involved in the enhancement of glycolysis. Conclusions The BET inhibitor JQ1 suppresses the proliferation of ALL by inhibiting c-Myc-mediated glycolysis, thus providing a new strategy for the treatment of ALL.
Collapse
Affiliation(s)
- Meng-Yi Zhang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland).,Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Sheng-Li Liu
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Wen-Li Huang
- Department of Pathology, School of Basic Medical Science, Central South University, Shanghai, China (mainland)
| | - Da-Bin Tang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Wei-Wei Zheng
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Neng Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Hang Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Tuersunayi Abudureheman
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Zhong-Hua Tang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland).,Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Cai-Wen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland).,Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|