201
|
Abawi A, Wang X, Bompard J, Bérot A, Andretto V, Gudimard L, Devillard C, Petiot E, Joseph B, Lollo G, Granjon T, Girard-Egrot A, Maniti O. Monomethyl Auristatin E Grafted-Liposomes to Target Prostate Tumor Cell Lines. Int J Mol Sci 2021; 22:ijms22084103. [PMID: 33921088 PMCID: PMC8071391 DOI: 10.3390/ijms22084103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Novel nanomedicines have been engineered to deliver molecules with therapeutic potentials, overcoming drawbacks such as poor solubility, toxicity or short half-life. Lipid-based carriers such as liposomes represent one of the most advanced classes of drug delivery systems. A Monomethyl Auristatin E (MMAE) warhead was grafted on a lipid derivative and integrated in fusogenic liposomes, following the model of antibody drug conjugates. By modulating the liposome composition, we designed a set of particles characterized by different membrane fluidities as a key parameter to obtain selective uptake from fibroblast or prostate tumor cells. Only the fluid liposomes made of palmitoyl-oleoyl-phosphatidylcholine and dioleoyl-phosphatidylethanolamine, integrating the MMAE-lipid derivative, showed an effect on prostate tumor PC-3 and LNCaP cell viability. On the other hand, they exhibited negligible effects on the fibroblast NIH-3T3 cells, which only interacted with rigid liposomes. Therefore, fluid liposomes grafted with MMAE represent an interesting example of drug carriers, as they can be easily engineered to promote liposome fusion with the target membrane and ensure drug selectivity.
Collapse
Affiliation(s)
- Ariana Abawi
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Xiaoyi Wang
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Julien Bompard
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Anna Bérot
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Valentina Andretto
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (V.A.); (G.L.)
| | - Leslie Gudimard
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Chloé Devillard
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Emma Petiot
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Benoit Joseph
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Giovanna Lollo
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (V.A.); (G.L.)
| | - Thierry Granjon
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Agnès Girard-Egrot
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
| | - Ofelia Maniti
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France; (A.A.); (X.W.); (J.B.); (A.B.); (L.G.); (C.D.); (E.P.); (B.J.); (T.G.); (A.G.-E.)
- Correspondence: ; Tel.: +33-(0)4-72-44-82-14
| |
Collapse
|
202
|
Wu T, Gong Y, Li Z, Li Y, Xiong X. Preparation and in vitro/vivo evaluation of folate-conjugated pluronic F87-PLGA/TPGS mixed nanoparticles for targeted drug delivery. Curr Drug Deliv 2021; 18:1505-1514. [PMID: 33845742 DOI: 10.2174/1567201818666210412123210] [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: 01/06/2021] [Revised: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
AIM Folate-conjugated Pluronic F87-poly(lactic-co-glycolic acid) block copolymer (FA-F87-PLGA) was synthesized to encapsulate anticancer drug Paclitaxel (PTX) for targeted drug delivery. To further improve the curative effect, D-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS or Vitamin E TPGS) was added to form FA-F87-PLGA/TPGS mixed NPs. METHODS FA-F87-PLGA was synthesized by the ring-opening polymerization and the structure was characterized. PTX-loaded nanoparticles were prepared with the nanoprecipitation method. The physicochemical characteristics were studied to determine the appropriate dose ratio of the FA-F87-PLGA to TPGS. The cytotoxicity against Ovarian Cancer Cells (OVCAR-3) was determined by MTT assay. The Area-Under-the Curve (AUC) and half-life were measured in the vivo pharmacokinetic studies. RESULTS Based on the optimization of particle size and embedding rate of PTX-loaded mixed NPs, the appropriate dosage ratio of FA-F87-PLGA to TPGS was finally determined to be 5:3. According to in vitro release studies, the cumulative release rate of PTX-loaded FA-F87-PLGA/TPGS mixed NPs was 92.04%, which was higher than that of nanoparticles without TPGS. The cytotoxicity studies showed that the IC50 value of PTX-loaded FA-F87-PLGA/TPGS decreased by 75.4 times and 19.7 times after 72 h treatment compared with free PTX injections and PTX-loaded FA-F87-PLGA NPs, respectively. In vivo pharmacokinetic studies indicated that FA-F87-PLGA/TPGS mixed NPs had a longer drug metabolism time and a larger Area-Under-the-Curve (AUC) compared with free PTX injections. CONCLUSION FA-F87-PLGA/TPGS mixed NPs are potential candidates for targeted drug delivery systems.
Collapse
Affiliation(s)
- Tianyi Wu
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Yanchun Gong
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Ziling Li
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Yuping Li
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Xiangyuan Xiong
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| |
Collapse
|
203
|
Amelioration of Tumor Targeting and In Vivo Biodistribution of 99mTc-Methotrexate-Gold Nanoparticles ( 99mTc-Mex-AuNPs). J Pharm Sci 2021; 110:2955-2965. [PMID: 33812886 DOI: 10.1016/j.xphs.2021.03.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 02/01/2023]
Abstract
Gold nanoparticles (AuNPs) represent very attractive and promising drug delivery carriers due to their unique dimensions, adjustable surface functions, and controllable drug release. Therefore, AuNPs are used to overcome the limitations of conventional chemotherapy, for example methotrexate (Mex), one of the first-generation chemotherapy drugs for cancer treatment, whose usefulness has been restricted due to drug resistance and dose-dependent side effects. In the present study, the AuNPs drug delivery system was synthesized and loaded with technetium-99 m radiolabeled Methotrexate (99mTc-Mex) to produce new potential nanoradiopharmaceutical for tumor targeting and further imaging. The Methotrexate loaded gold nanoparticles (Mex-AuNPs) successfully prepared in small spherical particle size (20.3 nm), polydispersity index PDI (< 0.5) and a zeta potential (-17.6 mV) with loading efficiency% (93 ± 1.2%) of methotrexate at 30 min as an optimum stirring time and showed strong absorption peak for Mex-AuNPs at λmax, 525 nm. The in vitro release profile of Mex-AuNPs showed high release percent of methotrexate at pH 5; the Q0.5 h and Q8h were 21.2 ± 1.5% and 92.9 ± 3.4%, respectively. The in vitro cytotoxicity was investigated at different concentrations (0.024-50 μl/100 μl) of Mex-AuNPs (1 mg/ml) against MCF-7 (Michigan Cancer Foundation-7) breast cancer cells by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay technique. Mex-AuNPs showed higher anticancer activity with low inhibitory concentration (IC50 = 0.098 μl/100 μl) that was three times lower than the inhibitory concentration (IC50) of methotrexate (IC50 = 0.3 μl/100 μl). 99mTc-Mex complex prepared by direct reduction method at maximum radiochemical yield (RCY)% ̴ 98.3 ± 1.09 % was loaded in AuNPs to form 99mTc-Mex-AuNPs with loading efficiency% (93 ± 1.2 %) at 30 min of stirring time. 99mTc-Mex-AuNPs showed convenient in vitro stability in mice serum up to 24 h with RCY% > 90 %. The preclinical biodistribution studies of 99mTc-Mex-AuNPs were performed in 3 experimental groups A (intravenous (I.V.) injected normal mice), B and C (I.V. and intratumor (I.T.) injected tumor bearing mice, respectively). The 99mTc-Mex-AuNPs achieved highest tumor uptake (93 ± 0.39 %ID/g) and highest Target/NonTarget (T/NT) ratio (58.1 ± 0.91) with high Tumor/Blood (T/B) ratio (25.8 ± 0.11) at 10 min post I.T. injection and retained high tumor uptake (79 ± 0.65 %ID/g) up to 60 min post I.T. injection before escaping into blood stream. Consequently, 99mTc-Mex-AuNPs can be considered as new potential nanoradiopharmaceutical in tumor diagnosis.
Collapse
|
204
|
Jaiswal S, Dutta P, Kumar S, Chawla R. Chitosan modified by organo-functionalities as an efficient nanoplatform for anti-cancer drug delivery process. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
205
|
Ou YH, Zou S, Goh WJ, Wang JW, Wacker M, Czarny B, Pastorin G. Cell-Derived Nanovesicles as Exosome-Mimetics for Drug Delivery Purposes: Uses and Recommendations. Methods Mol Biol 2021; 2211:147-170. [PMID: 33336276 DOI: 10.1007/978-1-0716-0943-9_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Cell-derived Drug Delivery Systems (DDSs), particularly exosomes, have grown in popularity and have been increasingly explored as novel DDSs, due to their intrinsic targeting capabilities. However, clinical translation of exosomes is impeded by the tedious isolation procedures and poor yield. Cell-derived nanovesicles (CDNs) have recently been produced and proposed as exosome-mimetics. Various methods for producing exosome-mimetics have been developed. In this chapter, we present a simple, efficient, and cost-effective CDNs production method that uses common laboratory equipment (microcentrifuge) and spin cups. Through a series of extrusion and size exclusion steps, CDNs are produced from in vitro cell culture and are found to highly resemble the endogenous exosomes. Thus, we envision that this strategy holds great potential as a viable alternative to exosomes in the development of ideal DDS.
Collapse
Affiliation(s)
- Yi-Hsuan Ou
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Shui Zou
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Wei Jiang Goh
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences (CeLS), Singapore, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre, Singapore, Singapore
| | - Matthias Wacker
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Bertrand Czarny
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences (CeLS), Singapore, Singapore.
| |
Collapse
|
206
|
Longo R, Gorrasi G, Guadagno L. Electromagnetically Stimuli-Responsive Nanoparticles-Based Systems for Biomedical Applications: Recent Advances and Future Perspectives. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:848. [PMID: 33810343 PMCID: PMC8065448 DOI: 10.3390/nano11040848] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
Nanoparticles (NPs) in the biomedical field are known for many decades as carriers for drugs that are used to overcome biological barriers and reduce drug doses to be administrated. Some types of NPs can interact with external stimuli, such as electromagnetic radiations, promoting interesting effects (e.g., hyperthermia) or even modifying the interactions between electromagnetic field and the biological system (e.g., electroporation). For these reasons, at present these nanomaterial applications are intensively studied, especially for drugs that manifest relevant side effects, for which it is necessary to find alternatives in order to reduce the effective dose. In this review, the main electromagnetic-induced effects are deeply analyzed, with a particular focus on the activation of hyperthermia and electroporation phenomena, showing the enhanced biological performance resulting from an engineered/tailored design of the nanoparticle characteristics. Moreover, the possibility of integrating these nanofillers in polymeric matrices (e.g., electrospun membranes) is described and discussed in light of promising applications resulting from new transdermal drug delivery systems with controllable morphology and release kinetics controlled by a suitable stimulation of the interacting systems (nanofiller and interacting cells).
Collapse
Affiliation(s)
- Raffaele Longo
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy;
| | | | - Liberata Guadagno
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy;
| |
Collapse
|
207
|
Le Q, Kim D, Lee J, Shim G, Oh Y. Photosensitizer‐Free Phototherapy with Peptide Micelle Nanoadjuvants for Cancer Vaccine against Metastasis of Melanoma. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Quoc‐Viet Le
- College of Pharmacy and Research Institute of Pharmaceutical Sciences Seoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences Seoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences Seoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| | - Gayong Shim
- School of Systems Biomedical Science Soongsil University Seoul 06978 Republic of Korea
| | - Yu‐Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences Seoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| |
Collapse
|
208
|
Fe 3O 4-Au Core-Shell Nanoparticles as a Multimodal Platform for In Vivo Imaging and Focused Photothermal Therapy. Pharmaceutics 2021; 13:pharmaceutics13030416. [PMID: 33804636 PMCID: PMC8003746 DOI: 10.3390/pharmaceutics13030416] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 01/02/2023] Open
Abstract
In this study, we report the synthesis of gold-coated iron oxide nanoparticles capped with polyvinylpyrrolidone (Fe@Au NPs). The as-synthesized nanoparticles (NPs) exhibited good stability in aqueous media and excellent features as contrast agents (CA) for both magnetic resonance imaging (MRI) and X-ray computed tomography (CT). Additionally, due to the presence of the local surface plasmon resonances of gold, the NPs showed exploitable "light-to-heat" conversion ability in the near-infrared (NIR) region, a key attribute for effective photothermal therapies (PTT). In vitro experiments revealed biocompatibility as well as excellent efficiency in killing glioblastoma cells via PTT. The in vivo nontoxicity of the NPs was demonstrated using zebrafish embryos as an intermediate step between cells and rodent models. To warrant that an effective therapeutic dose was achieved inside the tumor, both intratumoral and intravenous routes were screened in rodent models by MRI and CT. The pharmacokinetics and biodistribution confirmed the multimodal imaging CA capabilities of the Fe@AuNPs and revealed constraints of the intravenous route for tumor targeting, dictating intratumoral administration for therapeutic applications. Finally, Fe@Au NPs were successfully used for an in vivo proof of concept of imaging-guided focused PTT against glioblastoma multiforme in a mouse model.
Collapse
|
209
|
Zhong Y, Dong Y, Chen T, Yang L, Yao M, Zhi Y, Yang H, Zhang J, Bi W. 808 nm NIR Laser-Excited Upconversion Nanoplatform for Combinatory Photodynamic and Chemotherapy with Deep Penetration and Acid Bursting Release Performance. ACS APPLIED BIO MATERIALS 2021; 4:2639-2653. [DOI: 10.1021/acsabm.0c01607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yingtao Zhong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Yun Dong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Tie Chen
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Lingzhi Yang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Min Yao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Yunshi Zhi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Haoyi Yang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Wenchuan Bi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
210
|
Guo D, Ji X, Luo J. Rational nanocarrier design towards clinical translation of cancer nanotherapy. Biomed Mater 2021; 16. [DOI: 10.1088/1748-605x/abe35a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
|
211
|
Pan C, Zhang T, Li S, Xu Z, Pan B, Xu S, Jin S, Lu G, Yang S, Xue Z, Chen P, Shen X, Wang F, Xu C. Hybrid Nanoparticles Modified by Hyaluronic Acid Loading an HSP90 Inhibitor as a Novel Delivery System for Subcutaneous and Orthotopic Colon Cancer Therapy. Int J Nanomedicine 2021; 16:1743-1755. [PMID: 33688189 PMCID: PMC7936682 DOI: 10.2147/ijn.s275805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
Background As a therapeutic target for cancer treatment, HSP90 has been explored extensively. However, the significant side effects of the HSP90 inhibitor 17AAG have limited its clinical use. Methods In this study, we used hyaluronic acid (HA)–decorated DOTAP–PLGA hybrid nanoparticles (HA-DOTAP-PLGA NPs) as 17AAG-delivery carriers for targeted colon cancer therapy. Results Different methods were used to characterize the successful fabrication of these hybrid PLGA NPs. Our results demonstrated that internalization of HA-NPs in colon cancer cells was governed by CD44receptor–mediated endocytosis. Annexin V–propidium iodide staining experiments revealed that cell apoptosis induced by HA-NPs-17AAG in colon cancer cells was more efficient than free 17AAG. In two animal models used to screen anticancer efficacy (Luc-HT29 subcutaneous xenograft and AOM/DSS-induced orthotopic tumor model), HA-NPs-17AAG significantly inhibited xenograft and orthotopic tumor growth, demonstrating HA-NPs-17AAG had much better therapeutic efficiency than free 17AAG. It is worth noting that great biocompatibility of HA-DOTAP-PLGA NPs was observed both in vitro and in vivo. Conclusion Our research offers a preclinical proof of concept for colon cancer therapy with DOTAP-PLGA NPs as a creative drug-delivery system.
Collapse
Affiliation(s)
- Chenwei Pan
- Department of Infectious Disease, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Tiaotiao Zhang
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Shaoxun Li
- Department of Infectious Disease, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Zhihua Xu
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Binhui Pan
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Sheng Xu
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Shuanghong Jin
- Department of Infectious Disease, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Guangrong Lu
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Shouxing Yang
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Zhanxiong Xue
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Ping Chen
- Department of Infectious Disease, Shulan Hospital, Hangzhou, Zhejiang, 310012, People's Republic of China
| | - Xian Shen
- Department of Gastrointestinal Surgery, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Changlong Xu
- Department of Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Center for Diagnostics and Therapeutics, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| |
Collapse
|
212
|
Laramie MD, Fouts BL, MacCuaig WM, Buabeng E, Jones MA, Mukherjee P, Behkam B, McNally LR, Henary M. Improved pentamethine cyanine nanosensors for optoacoustic imaging of pancreatic cancer. Sci Rep 2021; 11:4366. [PMID: 33623069 PMCID: PMC7902650 DOI: 10.1038/s41598-021-83658-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
Optoacoustic imaging is a new biomedical imaging technology with clear benefits over traditional optical imaging and ultrasound. While the imaging technology has improved since its initial development, the creation of dedicated contrast agents for optoacoustic imaging has been stagnant. Current exploration of contrast agents has been limited to standard commercial dyes that have already been established in optical imaging applications. While some of these compounds have demonstrated utility in optoacoustic imaging, they are far from optimal and there is a need for contrast agents with tailored optoacoustic properties. The synthesis, encapsulation within tumor targeting silica nanoparticles and applications in in vivo tumor imaging of optoacoustic contrast agents are reported.
Collapse
Affiliation(s)
- Matthew D Laramie
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - Benjamin L Fouts
- Department of Surgery, Oklahoma Health Science Center, Oklahoma City, 73104, USA
- Stephenson Cancer Center, Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
| | - William M MacCuaig
- Stephenson Cancer Center, Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK, 72073, USA
| | - Emmanuel Buabeng
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, 30303, USA
| | - Meredith A Jones
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK, 72073, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech University, Blacksburg, VA, 24061, USA
| | - Lacey R McNally
- Department of Surgery, Oklahoma Health Science Center, Oklahoma City, 73104, USA.
- Stephenson Cancer Center, Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA.
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK, 72073, USA.
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC, 27157, USA.
| | - Maged Henary
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA.
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
213
|
Bioactive Polymeric Materials for the Advancement of Regenerative Medicine. J Funct Biomater 2021; 12:jfb12010014. [PMID: 33672492 PMCID: PMC8006220 DOI: 10.3390/jfb12010014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Biopolymers are widely accepted natural materials in regenerative medicine, and further development of their bioactivities and discoveries on their composition/function relationships could greatly advance the field. However, a concise insight on commonly investigated biopolymers, their current applications and outlook of their modifications for multibioactivity are scarce. This review bridges this gap for professionals and especially freshmen in the field who are also interested in modification methods not yet in commercial use. A series of polymeric materials in research and development uses are presented as well as challenges that limit their efficacy in tissue regeneration are discussed. Finally, their roles in the regeneration of select tissues including the skin, bone, cartilage, and tendon are highlighted along with modifiable biopolymer moieties for different bioactivities.
Collapse
|
214
|
Li T, Yao F, An Y, Li X, Duan J, Yang XD. Novel Complex of PD-L1 Aptamer and Holliday Junction Enhances Antitumor Efficacy in Vivo. Molecules 2021; 26:1067. [PMID: 33670583 PMCID: PMC7921949 DOI: 10.3390/molecules26041067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Blocking the PD-1/PD-L1 pathway can diminish immunosuppression and enhance anticancer immunity. PD-1/PD-L1 blockade can be realized by aptamers, which have good biocompatibility and can be synthesized in quantity economically. For in vivo applications, aptamers need to evade renal clearance and nuclease digestion. Here we investigated whether DNA nanostructures could be used to enhance the function of PD-L1 aptamers. Four PD-L1 aptamers (Apt) were built into a Holliday Junction (HJ) to form a tetravalent DNA nanostructure (Apt-HJ). The average size of Apt-HJ was 13.22 nm, which was above the threshold for renal clearance. Apt-HJ also underwent partial phosphorothioate modification and had improved nuclease resistance. Compared with the monovalent PD-L1 aptamer, the tetravalent Apt-HJ had stronger affinity to CT26 colon cancer cells. Moreover, Apt-HJ markedly boosted the antitumor efficacy in vivo vs. free PD-L1 aptamers without raising systemic toxicity. The results indicate that multiple aptamers attached to a DNA nanostructure may significantly improve the function of PD-L1 aptamers in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Xian-Da Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (T.L.); (F.Y.); (Y.A.); (X.L.); (J.D.)
| |
Collapse
|
215
|
Ercan A, Çelebier M, Oncul S, Varan G, Kocak E, Benito JM, Bilensoy E. Polycationic cyclodextrin nanoparticles induce apoptosis and affect antitumoral activity in HepG2 cell line: An evaluation at the molecular level. Int J Pharm 2021; 598:120379. [PMID: 33592288 DOI: 10.1016/j.ijpharm.2021.120379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a highly metastatic primary liver cancer generating molecular alterations that end up escaping the apoptotic machinery and conferring multidrug resistance. Targeted medicines with increased and selective cytotoxicity and minimal drug resistance are essential for the treatment of HCC. In this study, a self-assembled polycationic (PC) amphiphilic β-cyclodextrin (βCDC6) nanoparticle formulation was characterized and its efficacy over HCC cell line HepG2 was evaluated in terms of cytotoxicity, apoptotic potential, chemosensitivity and mitochondrial balance utilizing biochemical, gene expression and proteomic approaches without encapsulating an anti-neoplastic agent. Blank PC βCDC6 exerted an anti-proliferative effect on 3D multicellular HepG2 spheroid tumors. These nanoparticles were able to trigger apoptosis proved by caspase 3/7 activity, gene expression and flow cytometry studies. The subjection of PC restored the chemosensitivity of HepG2 cells by suppressing the function of p-glycoprotein. The proteomic studies with Q-TOF LC/MS revealed 73 proteins that are aberrantly encoded after cells were treated with the blank PC. Metabolomic analysis further confirmed the shift in certain biological pathways. Thus, we confirmed that the hepatocellular carcinoma-targeting βCDC6 PC nanoparticles induce apoptosis, lower the rate of cell proliferation, hinder multidrug resistance and they are convenient carriers for eventual therapeutic administrations in HCC patients.
Collapse
Affiliation(s)
- Ayse Ercan
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, 06100 Ankara Turkey
| | - Mustafa Çelebier
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100 Ankara Turkey
| | - Selin Oncul
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, 06100 Ankara Turkey
| | - Gamze Varan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara Turkey
| | - Engin Kocak
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100 Ankara Turkey
| | - Juan M Benito
- Institute for Chemical Research, CSIC-University of Sevilla, Sevilla, Spain
| | - Erem Bilensoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara Turkey.
| |
Collapse
|
216
|
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, Ingham ES, Mahakian LM, Tam SM, Cheng RH, Borowsky AD, Ferrara KW. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release 2021; 330:1080-1094. [PMID: 33189786 PMCID: PMC7906914 DOI: 10.1016/j.jconrel.2020.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/01/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 μg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Wei-Lun Tang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Azadeh Kheirolomoom
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Kenneth Lau
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Marina Nura Raie
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Spencer K Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | | | - Katherine W Ferrara
- Molecular Imaging Program, Department of Radiology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
217
|
Lin S, Liu C, Han X, Zhong H, Cheng C. Viral Nanoparticle System: An Effective Platform for Photodynamic Therapy. Int J Mol Sci 2021; 22:ijms22041728. [PMID: 33572365 PMCID: PMC7916136 DOI: 10.3390/ijms22041728] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising therapy due to its efficiency and accuracy. The photosensitizer is delivered to the target lesion and locally activated. Viral nanoparticles (VNPs) have been explored as delivery vehicles for PDT in recent years because of their favorable properties, including simple manufacture and good safety profile. They have great potential as drug delivery carriers in medicine. Here, we review the development of PDT photosensitizers and discuss applications of VNP-mediated photodynamic therapies and the performance of VNPs in the treatment of tumor cells and antimicrobial therapy. Furthermore, future perspectives are discussed for further developing novel viral nanocarriers or improving existing viral vectors.
Collapse
Affiliation(s)
| | - Chun Liu
- Correspondence: (C.L.); (X.H.); (C.C.); Tel.: +86-591-8372-5260 (C.C.)
| | - Xiao Han
- Correspondence: (C.L.); (X.H.); (C.C.); Tel.: +86-591-8372-5260 (C.C.)
| | | | - Cui Cheng
- Correspondence: (C.L.); (X.H.); (C.C.); Tel.: +86-591-8372-5260 (C.C.)
| |
Collapse
|
218
|
Alsaab HO, Al-Hibs AS, Alzhrani R, Alrabighi KK, Alqathama A, Alwithenani A, Almalki AH, Althobaiti YS. Nanomaterials for Antiangiogenic Therapies for Cancer: A Promising Tool for Personalized Medicine. Int J Mol Sci 2021; 22:1631. [PMID: 33562829 PMCID: PMC7915670 DOI: 10.3390/ijms22041631] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the hallmarks of cancer. Several studies have shown that vascular endothelium growth factor (VEGF) plays a leading role in angiogenesis progression. Antiangiogenic medication has gained substantial recognition and is commonly administered in many forms of human cancer, leading to a rising interest in cancer therapy. However, this treatment method can lead to a deteriorating outcome of resistance, invasion, distant metastasis, and overall survival relative to its cytotoxicity. Furthermore, there are significant obstacles in tracking the efficacy of antiangiogenic treatments by incorporating positive biomarkers into clinical settings. These shortcomings underline the essential need to identify additional angiogenic inhibitors that target numerous angiogenic factors or to develop a new method for drug delivery of current inhibitors. The great benefits of nanoparticles are their potential, based on their specific properties, to be effective mechanisms that concentrate on the biological system and control various important functions. Among various therapeutic approaches, nanotechnology has emerged as a new strategy for treating different cancer types. This article attempts to demonstrate the huge potential for targeted nanoparticles and their molecular imaging applications. Notably, several nanoparticles have been developed and engineered to demonstrate antiangiogenic features. This nanomedicine could effectively treat a number of cancers using antiangiogenic therapies as an alternative approach. We also discuss the latest antiangiogenic and nanotherapeutic strategies and highlight tumor vessels and their microenvironments.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (A.H.A.); (Y.S.A.)
| | - Alanoud S. Al-Hibs
- Department of Pharmacy, King Fahad Medical City, Riyadh 11564, Saudi Arabia;
| | - Rami Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Khawlah K. Alrabighi
- Batterjee Medical College for Sciences and Technology, Jeddah 21577, Saudi Arabia;
| | - Aljawharah Alqathama
- Department of Pharmacognosy, Pharmacy College, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Akram Alwithenani
- Department of Laboratory Medicine, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Atiah H. Almalki
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (A.H.A.); (Y.S.A.)
- Department of Pharmaceutical Chemistry, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Yusuf S. Althobaiti
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (A.H.A.); (Y.S.A.)
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
219
|
Calori IR, Bi H, Tedesco AC. Expanding the Limits of Photodynamic Therapy: The Design of Organelles and Hypoxia-Targeting Nanomaterials for Enhanced Photokilling of Cancer. ACS APPLIED BIO MATERIALS 2021; 4:195-228. [PMID: 35014281 DOI: 10.1021/acsabm.0c00945] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Photodynamic therapy (PDT) is a minimally invasive clinical protocol that combines a nontoxic photosensitizer (PS), appropriate visible light, and molecular oxygen for cancer treatment. This triad generates reactive oxygen species (ROS) in situ, leading to different cell death pathways and limiting the arrival of nutrients by irreversible destruction of the tumor vascular system. Despite the number of formulations and applications available, the advancement of therapy is hindered by some characteristics such as the hypoxic condition of solid tumors and the limited energy density (light fluence) that reaches the target. As a result, the use of PDT as a definitive monotherapy for cancer is generally restricted to pretumor lesions or neoplastic tissue of approximately 1 cm in size. To expand this limitation, researchers have synthesized functional nanoparticles (NPs) capable of carrying classical photosensitizers with self-supplying oxygen as well as targeting specific organelles such as mitochondria and lysosomes. This has improved outcomes in vitro and in vivo. This review highlights the basis of PDT, many of the most commonly used strategies of functionalization of smart NPs, and their potential to break the current limits of the classical protocol of PDT against cancer. The application and future perspectives of the multifunctional nanoparticles in PDT are also discussed in some detail.
Collapse
Affiliation(s)
- Italo Rodrigo Calori
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo-Ribeirão Preto, São Paulo 14040-901, Brazil
| | - Hong Bi
- School of Chemistry and Chemical Engineering, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, 111 Jiulong Road, Hefei 230601, China
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo-Ribeirão Preto, São Paulo 14040-901, Brazil.,School of Chemistry and Chemical Engineering, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, 111 Jiulong Road, Hefei 230601, China
| |
Collapse
|
220
|
Park J, Lee YK, Park IK, Hwang SR. Current Limitations and Recent Progress in Nanomedicine for Clinically Available Photodynamic Therapy. Biomedicines 2021; 9:85. [PMID: 33467201 PMCID: PMC7830249 DOI: 10.3390/biomedicines9010085] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Photodynamic therapy (PDT) using oxygen, light, and photosensitizers has been receiving great attention, because it has potential for making up for the weakness of the existing therapies such as surgery, radiation therapy, and chemotherapy. It has been mainly used to treat cancer, and clinical tests for second-generation photosensitizers with improved physicochemical properties, pharmacokinetic profiles, or singlet oxygen quantum yield have been conducted. Progress is also being made in cancer theranostics by using fluorescent signals generated by photosensitizers. In order to obtain the effective cytotoxic effects on the target cells and prevent off-target side effects, photosensitizers need to be localized to the target tissue. The use of nanocarriers combined with photosensitizers can enhance accumulation of photosensitizers in the tumor site, owing to preferential extravasation of nanoparticles into the tumor vasculature by the enhanced permeability and retention effect. Self-assembly of amphiphilic polymers provide good loading efficiency and sustained release of hydrophobic photosensitizers. In addition, prodrug nanomedicines for PDT can be activated by stimuli in the tumor site. In this review, we introduce current limitations and recent progress in nanomedicine for PDT and discuss the expected future direction of research.
Collapse
Affiliation(s)
- Jooho Park
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea;
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea;
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju 61452, Korea
| |
Collapse
|
221
|
Wen W, Guo C, Guo J. Acid-Responsive Adamantane-Cored Amphiphilic Block Polymers as Platforms for Drug Delivery. NANOMATERIALS 2021; 11:nano11010188. [PMID: 33451051 PMCID: PMC7828523 DOI: 10.3390/nano11010188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/06/2021] [Accepted: 01/09/2021] [Indexed: 12/19/2022]
Abstract
Four-arm star-shaped (denoted as ‘S’) polymer adamantane-[poly(lactic-co-glycolic acid)-b-poly(N,N’-diethylaminoethyl methacrylate) poly(ethylene glycol) monomethyl ether]4 (S-PLGA-D-P) and its linear (denoted as ‘L’) counterpart (L-PLGA-D-P) were synthesized, then their self-assembled micelles were further developed to be platforms for anticancer drug delivery. Two types of polymeric micelles exhibited strong pH-responsiveness and good drug loading capacity (21.6% for S-PLGA-D-P and 22.9% for L-PLGA-D-P). Using doxorubicin (DOX) as the model drug, their DOX-loaded micelles displayed well controlled drug release behavior (18.5–19.0% of DOX release at pH 7.4 and 77.6–78.8% of DOX release at pH 5.0 within 80 h), good cytocompatibility against NIH-3T3 cells and effective anticancer efficacy against MCF-7 cells. However, the star-shaped polymeric micelles exhibited preferable stability, which was confirmed by the lower critical micelle concentration (CMC 0.0034 mg/mL) and decrease rate of particle sizes after 7 days incubation (3.5%), compared with the linear polymeric micelle L-PLGA-D-P (CMC 0.0070 mg/mL, decrease rate of particle sizes was 9.6%). Overall, these developed polymeric micelles have promising application as drug delivery system in cancer therapy.
Collapse
Affiliation(s)
- Weiqiu Wen
- School of Chemical Engineering & Light Industry, Guangdong University of Technology, Guangzhou 510006, China;
| | - Chong Guo
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China;
| | - Jianwei Guo
- School of Chemical Engineering & Light Industry, Guangdong University of Technology, Guangzhou 510006, China;
- Correspondence:
| |
Collapse
|
222
|
Saw WS, Anasamy T, Foo YY, Kwa YC, Kue CS, Yeong CH, Kiew LV, Lee HB, Chung LY. Delivery of Nanoconstructs in Cancer Therapy: Challenges and Therapeutic Opportunities. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000206] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wen Shang Saw
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Theebaa Anasamy
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Yiing Yee Foo
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur 50603 Malaysia
| | - Yee Chu Kwa
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Chin Siang Kue
- Department of Diagnostic and Allied Health Sciences Faculty of Health and Life Sciences Management and Science University Shah Alam Selangor 40100 Malaysia
| | - Chai Hong Yeong
- School of Medicine Faculty of Health and Medical Sciences Taylor's University Subang Jaya Selangor 47500 Malaysia
| | - Lik Voon Kiew
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur 50603 Malaysia
| | - Hong Boon Lee
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
- School of Biosciences Faculty of Health and Medical Sciences Taylor's University Subang Jaya Selangor 47500 Malaysia
| | - Lip Yong Chung
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| |
Collapse
|
223
|
Szczygieł A, Anger-Góra N, Węgierek-Ciura K, Mierzejewska J, Rossowska J, Goszczyński TM, Świtalska M, Pajtasz-Piasecka E. Immunomodulatory potential of anticancer therapy composed of methotrexate nanoconjugate and dendritic cell‑based vaccines in murine colon carcinoma. Oncol Rep 2021; 45:945-962. [PMID: 33432365 PMCID: PMC7859925 DOI: 10.3892/or.2021.7930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022] Open
Abstract
Chemotherapy with low-molecular weight compounds, despite elimination of cancer cells, entails adverse effects. To overcome this disadvantage, innovative drug delivery systems are being developed, including conjugation of macromolecular carriers with therapeutics, e.g. a nanoconjugate of hydroxyethyl starch and methotrexate (HES-MTX). The purpose of the present study was to determine whether HES-MTX, applied as a chemotherapeutic, is able to modulate the immune response and support the antitumor response generated by dendritic cells (DCs) used subsequently as immunotherapeutic vaccines. Therefore, MTX or HES-MTX was administered, as sole treatment or combined with DC-based vaccines, to MC38 colon carcinoma tumor-bearing mice. Alterations in antitumor immune response were evaluated by multiparameter flow cytometry analyses and functional assays. The results demonstrated that the nanoconjugate possesses greater immunomodulatory potential than MTX as reflected by changes in the landscape of immune cells infiltrating the tumor and increased cytotoxicity of splenic lymphocytes. In contrast to MTX, therapy with HES-MTX as sole treatment or combined with DC-based vaccines, contributed to significant tumor growth inhibition. However, only treatment with HES-MTX and DC-based vaccines activated the systemic specific antitumor response. In conclusion, due to its immunomodulatory properties, the HES-MTX nanoconjugate could become a potent anticancer agent used in both chemo- and chemoimmunotherapeutic treatment schemes.
Collapse
Affiliation(s)
- Agnieszka Szczygieł
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Natalia Anger-Góra
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Katarzyna Węgierek-Ciura
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Jagoda Mierzejewska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Tomasz M Goszczyński
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Marta Świtalska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Elżbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| |
Collapse
|
224
|
Sonju JJ, Dahal A, Singh SS, Jois SD. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J Control Release 2021; 329:624-644. [PMID: 33010333 PMCID: PMC8082750 DOI: 10.1016/j.jconrel.2020.09.055] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/26/2022]
Abstract
Clinically efficacious medication in anticancer therapy has been successfully designed with liposome-based nanomedicine. The liposomal formulation in cancer drug delivery can be facilitated with a functionalized peptide that mediates the specific drug delivery opportunities with increased drug penetrability, specific accumulation in the targeted site, and enhanced therapeutic efficacy. This review aims to focus on recent advances in peptide-functionalized liposomal formulation techniques in cancer diagnosis and treatment regarding recently published literature. It also will highlight different aspects of novel liposomal formulation techniques that incorporate surface functionalization with peptides for better anticancer effect and current challenges in peptide-functionalized liposomal drug formulation.
Collapse
Affiliation(s)
- Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
225
|
Pinzaru I, Tanase A, Enatescu V, Coricovac D, Bociort F, Marcovici I, Watz C, Vlaia L, Soica C, Dehelean C. Proniosomal Gel for Topical Delivery of Rutin: Preparation, Physicochemical Characterization and In Vitro Toxicological Profile Using 3D Reconstructed Human Epidermis Tissue and 2D Cells. Antioxidants (Basel) 2021; 10:antiox10010085. [PMID: 33435216 PMCID: PMC7827235 DOI: 10.3390/antiox10010085] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/22/2022] Open
Abstract
Rutin (Rut) is a natural flavonol, well-known for its broad-spectrum of therapeutic effects, including antioxidant and antitumoral activities; still, it has a reduced clinical outcome due to its limited solubility in aqueous solutions. To overcome this drawback, this study proposes a novel formulation for rutin as a proniosomal gel for cutaneous applications. The gel was prepared by coacervation phase-separation method and complies with the standard requirements in terms of particle size (140.5 ± 2.56 nm), zeta potential (−27.33 ± 0.09 mV), encapsulation capacity (> 50%), pH (7.002 ± 0.18) and rheological properties. The results showed high biocompatibility of the gel on the 3D reconstructed human epidermis model characterized by increased viability of the cells and a lack of irritant and phototoxic potential. The evaluations on 2D cells confirm the preferential cytotoxic effect of Rut on melanoma cells (IC50 value = 8.601 µM, nuclear fragmentation) compared to normal keratinocytes. Our data suggest that the proniosomal gel is a promising drug carrier for Rut in the management and prevention of skin disorders.
Collapse
Affiliation(s)
- Iulia Pinzaru
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Alina Tanase
- Faculty of Dental Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 9 Revolutiei Bv., Sq., 300041 Timișoara, Romania;
| | - Virgil Enatescu
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
- Correspondence: (V.E.); (D.C.); Tel.: +40-723-374896 (V.E.); +40-256-494604 (D.C.)
| | - Dorina Coricovac
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
- Correspondence: (V.E.); (D.C.); Tel.: +40-723-374896 (V.E.); +40-256-494604 (D.C.)
| | - Flavia Bociort
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Iasmina Marcovici
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Claudia Watz
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Lavinia Vlaia
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Codruta Soica
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Cristina Dehelean
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (I.P.); (I.M.); (C.W.); (L.V.); (C.S.); (C.D.)
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| |
Collapse
|
226
|
Balu R, Dutta NK, Dutta AK, Choudhury NR. Resilin-mimetics as a smart biomaterial platform for biomedical applications. Nat Commun 2021; 12:149. [PMID: 33420053 PMCID: PMC7794388 DOI: 10.1038/s41467-020-20375-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Intrinsically disordered proteins have dramatically changed the structure-function paradigm of proteins in the 21st century. Resilin is a native elastic insect protein, which features intrinsically disordered structure, unusual multi-stimuli responsiveness and outstanding resilience. Advances in computational techniques, polypeptide synthesis methods and modular protein engineering routines have led to the development of novel resilin-like polypeptides (RLPs) including modular RLPs, expanding their applications in tissue engineering, drug delivery, bioimaging, biosensors, catalysis and bioelectronics. However, how the responsive behaviour of RLPs is encoded in the amino acid sequence level remains elusive. This review summarises the milestones of RLPs, and discusses the development of modular RLP-based biomaterials, their current applications, challenges and future perspectives. A perspective of future research is that sequence and responsiveness profiling of RLPs can provide a new platform for the design and development of new modular RLP-based biomaterials with programmable structure, properties and functions.
Collapse
Affiliation(s)
- Rajkamal Balu
- Chemical and Environmental Engineering, School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia
| | - Naba K Dutta
- Chemical and Environmental Engineering, School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia.
| | - Ankit K Dutta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Namita Roy Choudhury
- Chemical and Environmental Engineering, School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
227
|
Kumari R, Sunil D. A mechanistic insight into benefits of aggregation induced emissive luminogens in cancer. J Drug Target 2021; 29:592-608. [PMID: 33399029 DOI: 10.1080/1061186x.2020.1868479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Exploration of advanced chemotheranostics that benefit from a combined in vivo strategy of cancer diagnosis and chemotherapy simultaneously is highly valued and will expose novel possibilities in modifying treatment and reduce side effects. In recent years, nanodrug delivery systems that incorporate aggregation-induced emissive luminogens (AIEgens) have been developed to track and monitor anticancer drug release, trace translocation processes and predict chemotherapeutic responses. There are several classes of AIEgen based chemotheranostics such us stimuli-responsive nanoprodrugs, pH-sensitive mesoporous silica nanocarriers, supramolecular polymer systems, drug encapsulated carriers, carrier-free nanodrugs, self-indicating drug delivery nanomachines and AIEgen-prodrug co-assembly. The present review conveys mechanistic insight into the benefits of AIEgens in the theranostic application by illustrating the recent breakthroughs in chemotheranostic nanomedicines that incorporate these unique fluorophores as signal reporters. The perspectives that can be further explored are also highlighted with the hope to instil more research interest in the advancement of AIE active cancer chemotheranostics for imaging and treatment in vivo.HIGHLIGHTSAggregation induced emissive materials (AIEgens) exhibit unique advantages over conventional luminogens for synergistic diagnosis and chemotherapy of cancer in vivo.The combination of AIE and nanotechnology offers an excellent platform to fabricate advanced chemotheranostics for cancer therapy.
Collapse
Affiliation(s)
- Rashmi Kumari
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
228
|
Hernández Millares R, Mirza JA, Lee J, Choi K, Eom M, Ye SJ. Radiosensitization by Au-nanofilm at micrometer level using confocal Raman spectroscopy. Med Phys 2021; 48:796-804. [PMID: 33128244 DOI: 10.1002/mp.14570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To measure the radiosensitization by an Au-nanofilm (GNF) at a micrometer level on a radiochromic film (RCF) using confocal Raman spectroscopy (CRS). METHODS Unlaminated radiochromic films were irradiated by 200 kVp x-ray from 0.3 to 50 Gy to obtain a calibration curve. Raman spectra of these films were measured by positioning the postirradiated RCF perpendicular to the CRS monochromatic beam and reading a depth profile of the film along the lateral axis. The Raman peak corresponding to the C ≡ C peak was obtained from a region of interest of 100 × 5 µm2 . To investigate the radiosensitization by GNF, two sets of RCF, one attached to a 100-nm thick GNF and the other without GNF were irradiated at 0.5 Gy by 50 and 120 kVp X-rays. The spatial resolution of the CRS on the RCF was quantified by the modulation transfer function method (MTF). Thus, in the spatial resolution determined by MTF, the doses deposited on the films were evaluated. The dose enhancement factor (DEF) was obtained in the measurable micro-size by comparing doses deposited on the RCFs with and without GNF. To verify the experimental results, Monte Carlo simulations following the experimental set up were performed using Geant4. In addition, analytical calculations for the radiosensitization by GNF were carried out. RESULTS The confocal Raman spectroscopy on the RCF achieved a spatial resolution of ~6 μm. An experimental DEF within the first 6 μm depth from the surface of RCF was found to be 17.9 for 50 kVp and 14.7 for 120 kVp. The DEF for the same depth obtained by MC and analytical calculations was 13.53 and 9.75 for 50 kVp, and 10.63 and 6.67 for 120 kVp, respectively. CONCLUSIONS The experimental DEF as a function of the distance from GNF was consistent with data from previous studies and the MC simulations, supporting that CRS in conjunction with the RCF is a feasible micrometer-resolution dosimeter.
Collapse
Affiliation(s)
- Rodrigo Hernández Millares
- Program in Biomedical Radiation Sciences, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
| | - Jamal Ahmad Mirza
- Program in Biomedical Radiation Sciences, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea.,Isotope Production Division, Pakistan Institute of Nuclear Science and Technology, Nilore, Islamabad, 44000, Pakistan
| | - Junyoung Lee
- Program in Biomedical Radiation Sciences, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
| | - Kwon Choi
- Program in Biomedical Radiation Sciences, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
| | - Mingi Eom
- Program in Biomedical Radiation Sciences, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
| | - Sung-Joon Ye
- Program in Biomedical Radiation Sciences, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, 03080, Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon, 16229, Korea
| |
Collapse
|
229
|
Tang SY, Wei H, Yu CY. Peptide-functionalized delivery vehicles for enhanced cancer therapy. Int J Pharm 2021; 593:120141. [DOI: 10.1016/j.ijpharm.2020.120141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 02/08/2023]
|
230
|
Liang X, Xie Y, Wu J, Wang J, Petković M, Stepić M, Zhao J, Ma J, Mi L. Functional titanium dioxide nanoparticle conjugated with phthalocyanine and folic acid as a promising photosensitizer for targeted photodynamic therapy in vitro and in vivo. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 215:112122. [PMID: 33433386 DOI: 10.1016/j.jphotobiol.2020.112122] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/27/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
Photodynamic therapy (PDT) is a promising cancer treatment that can be implemented using various agents. The conventional photosensitizer Al (III) phthalocyanine chloride tetrasulfonic acid (Pc) has limitations of selectivity in tumor targeting, low affinity to cancer cells, and low two-photon absorption. This study presents a novel photosensitizer FA-TiO2-Pc, which has the TiO2 nanoparticle conjugated with a tumor targeting agent of folic acid (FA), and Pc. FA-TiO2-Pc possessed high targeted photodynamic therapeutic activity and excellent biocompatibility. This promising photosensitizer showed high therapeutic drug efficiency in vitro at a low concentration dose and short incubation time under one-photon excitation (OPE). In vivo, when treated with a low dose of FA-TiO2-Pc and low light irradiation, the tumor growth was depressed in mice bearing HeLa xenograft tumors with minimal side effects. In addition, the two-photon absorption of FA-TiO2-Pc was also enhanced compared to Pc, proving that FA-TiO2-Pc system has a great potential to be used for the therapy of the folate receptor positive cancer cells in both OPE-PDT and two-photon excitation (TPE)-PDT agents.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Optical Science and Engineering, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Green Photoelectron Platform, Fudan University, 220 Handan Road, Shanghai 200433, China
| | - Yonghui Xie
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Street, Wuhan, Hubei 430014, China
| | - Junxin Wu
- Department of Optical Science and Engineering, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Green Photoelectron Platform, Fudan University, 220 Handan Road, Shanghai 200433, China
| | - Jing Wang
- State Key Laboratory of High Field Laser Physics, Shanghai Institute of Optics and Fine Mechanics, Chinese Academy of Sciences, 390 Qinghe Road, Shanghai 201800, China
| | - Marijana Petković
- Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Milutin Stepić
- Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China.
| | - Jiong Ma
- Department of Optical Science and Engineering, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Green Photoelectron Platform, Fudan University, 220 Handan Road, Shanghai 200433, China; Institute of Biomedical Engineering and Technology, Academy for Engineer and Technology, Fudan University, 220 Handan Road, Shanghai 200433, China; Shanghai Engineering Research Center of Industrial Microorganisms, The Multiscale Research Institute of Complex Systems (MRICS), School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, China.
| | - Lan Mi
- Department of Optical Science and Engineering, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Green Photoelectron Platform, Fudan University, 220 Handan Road, Shanghai 200433, China.
| |
Collapse
|
231
|
Fakhri KU, Sultan A, Mushtaque M, Hasan MR, Nafees S, Hafeez ZB, Zafaryab M, Rizwanullah M, Sharma D, Bano F, AlMalki WH, Ahmad FJ, Rizvi MMA. Obstructions in Nanoparticles Conveyance, Nano-Drug Retention, and EPR Effect in Cancer Therapies. HANDBOOK OF RESEARCH ON ADVANCEMENTS IN CANCER THERAPEUTICS 2021. [DOI: 10.4018/978-1-7998-6530-8.ch026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, the authors first review nano-devices that are mixtures of biologic molecules and synthetic polymers like nano-shells and nano-particles for the most encouraging applications for different cancer therapies. Nano-sized medications additionally spill especially into tumor tissue through penetrable tumor vessels and are then held in the tumor bed because of diminished lymphatic drainage. This procedure is known as the enhanced penetrability and retention (EPR) impact. Nonetheless, while the EPR impact is generally held to improve conveyance of nano-medications to tumors, it in certainty offers not exactly a 2-overlay increment in nano-drug conveyance contrasted with basic ordinary organs, bringing about medication concentration that is not adequate for restoring most malignant growths. In this chapter, the authors likewise review different obstructions for nano-sized medication conveyance and to make the conveyance of nano-sized medications to tumors progressively successful by expanding on the EPR impact..
Collapse
Affiliation(s)
| | | | | | | | | | | | - Md Zafaryab
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Md Rizwanullah
- School of Pharmaceutical Education and Research, Jamia Hamdard, India
| | - Deepti Sharma
- Institute of Nuclear Medicine and Allied Sciences, India
| | - Farhad Bano
- National Institute of Immunology, New Delhi, India
| | | | - Farhan Jalees Ahmad
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
232
|
Shen J, Chen J, Ma J, Fan L, Zhang X, Yue T, Yan Y, Zhang Y. Enhanced lysosome escape mediated by 1,2-dicarboxylic-cyclohexene anhydride-modified poly-l-lysine dendrimer as a gene delivery system. Asian J Pharm Sci 2020; 15:759-776. [PMID: 33363631 PMCID: PMC7750821 DOI: 10.1016/j.ajps.2019.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/07/2019] [Accepted: 12/10/2019] [Indexed: 12/23/2022] Open
Abstract
Antisense oligodeoxynucleotide (ASODN) can directly interfere a series of biological events of the target RNA derived from tumor cells through Watson-Crick base pairing, in turn, plays antitumor therapeutic roles. In the study, a novel HIF-1α ASODN-loaded nanocomposite was formulated to efficiently deliver gene to the target RNA. The physicochemical properties of nanocomposite were characterized using TEM, FTIR, DLS and zeta potentials. The mean diameter of resulting GEL-DGL-FA-ASODN-DCA nanocomposite was about 170-192 nm, and according to the agarose gel retardation assay, the loading amount of ASODN accounted for 166.7 mg/g. The results of cellular uptake showed that the nanocomposite could specifically target to HepG2 and Hela cells. The cytotoxicity assay demonstrated that the toxicity of vectors was greatly reduced by using DCA to reversibly block the cationic DGL. The subcellular distribution images clearly displayed the lysosomal escape ability of the DCA-modified nanocomposite. In vitro exploration of molecular mechanism indicated that the nanocomposite could inhibit mRNA expression and HIF-1α protein translation at different levels. In vivo optical images and quantitative assay testified that the formulation accumulated preferentially in the tumor tissue. In vivo antitumor efficacy research confirmed that this nanocomposite had significant antitumor activity and the tumor inhibitory rate was 77.99%. These results manifested that the GEL-DGL-FA-ASODN-DCA nanocomposite was promising in gene therapeutics for antitumor by interacting directly with target RNA.
Collapse
Affiliation(s)
- Jianmin Shen
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Jing Chen
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Jingbo Ma
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Linlan Fan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Xiaoli Zhang
- Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Ting Yue
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.,Shenzhen Following Precision Medical Research Institute, Shenzhen 518001, China
| | - Yaping Yan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Yuhang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
233
|
Poudel K, Banstola A, Gautam M, Soe Z, Phung CD, Pham LM, Jeong JH, Choi HG, Ku SK, Tran TH, Yong CS, Kim JO. Macrophage-Membrane-Camouflaged Disintegrable and Excretable Nanoconstruct for Deep Tumor Penetration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:56767-56781. [PMID: 33289550 DOI: 10.1021/acsami.0c17235] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The consolidation of nanovectors with biological membranes has recently been a subject of interest owing to the prolonged systemic circulation time and delayed clearance by the reticuloendothelial system of such systems. Among the different biomembranes, the macrophage membrane has a similar systemic circulation time, with an additional chemotactic aptitude, targeting integrin proteins. In this study, we aimed to establish a laser-activated, disintegrable, and deeply tumor-penetrative nanoplatform. We used a highly tumor-ablative and laser-responsive disintegrable copper sulfide nanoparticle, loaded it with paclitaxel, and camouflaged it with the macrophage membrane for the fabrication of PTX@CuS@MMNPs. The in vitro paclitaxel release profile was favorable for release in the tumor microenvironment, and the release was accelerated after laser exposure. Cellular internalization was improved by membrane encapsulation. Cellular uptake, cytotoxicity, reactive oxygen species generation, and apoptosis induction of PTX@CuS@MMNPs were further improved upon laser exposure, and boosted permeation was achieved by co-administration of the tumor-penetrating peptide iRGD. In vivo tumor accumulation, tumor inhibition rate, and apoptotic marker expression induced by PTX@CuS@MMNPs were significantly improved by laser irradiation and iRGD co-administration. PTX@CuS@MMNPs induced downregulation of cellular proliferation and angiogenic markers but no significant changes in body weight, survival, or significant toxicities in vital organs after laser exposure, suggesting their biocompatibility. The disintegrability of the nanosystem, accredited to biodegradability, favored efficient elimination from the body. In conclusion, PTX@CuS@MMNPs showed promising traits in combination therapies for excellent tumor eradication.
Collapse
Affiliation(s)
- Kishwor Poudel
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Zarchi Soe
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Cao Dai Phung
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Le Minh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong District, Hanoi 100803, Vietnam
- PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No. 167 Hoang Ngan, Cau Giay, Hanoi 11313, Vietnam
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
234
|
Chen W, He J, Li H, Li X, Tian W. A quinolone derivative-based organoplatinum(II) metallacycle supramolecular self-delivery nanocarrier for combined cancer therapy. Supramol Chem 2020. [DOI: 10.1080/10610278.2020.1846739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wenzhuo Chen
- MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, Shanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi’an, PR China
| | - Jia He
- MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, Shanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi’an, PR China
| | - Hui Li
- School of Materials Science and Engineering, Jiangxi University of Science and Technology, Ganzhou, P. R. China
| | - Xuefeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou, P. R. China
- State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, P. R. China
| | - Wei Tian
- MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, Shanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi’an, PR China
| |
Collapse
|
235
|
Zang X, Zhou J, Zhang X, Chen D, Han Y, Chen X. Dual-targeting tumor cells and tumor associated macrophages with lipid coated calcium zoledronate for enhanced lung cancer chemoimmunotherapy. Int J Pharm 2020; 594:120174. [PMID: 33338567 DOI: 10.1016/j.ijpharm.2020.120174] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/26/2020] [Accepted: 12/10/2020] [Indexed: 12/28/2022]
Abstract
Lung cancer is the leading cause of cancer death among both men and women, and non-small cell lung cancer (NSCLC) accounts for almost 80% of such death. Tumor associated macrophage (TAMs) are abundant components in NSCLC. TAMs play critical roles in angiogenesis, immune escape and chemoresistance. Here we developed a dual-targeting drug delivery system (CaZOL@BMNPs) of zoledronate, which could bind to both tumor cells with overexpressed biotin receptors and macrophage mannose receptor (MMR) positive TAMs. The biotin- and mannose-modified lipid coated calcium zoledronate nanoparticles were preferentially internalized in both tumor cells and TAMs, and thereby inhibited their survivals. Our studies demonstrated that CaZOl@BMNPs treatment obviously reduced angiogenesis, reprogrammed immunosuppressive tumor microenvironment and eventually restrained tumor progression with negligible systemic toxicity. Collectively, CaZOL@BMNPs could be a promising approach by dual-targeting tumor cells and TAMs for NSCLS chemoimmunotherapy.
Collapse
Affiliation(s)
- Xinlong Zang
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao, PR China
| | - Jingyi Zhou
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao, PR China
| | - Xiaoxu Zhang
- School of Pharmacy, Shenyang University, Wenhua Road 103, Shenyang, PR China
| | - Dawei Chen
- School of Pharmacy, Shenyang University, Wenhua Road 103, Shenyang, PR China
| | - Yantao Han
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao, PR China
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao, PR China.
| |
Collapse
|
236
|
Nagaya T, Choyke PL, Kobayashi H. Near-Infrared Photoimmunotherapy for Cancers of the Gastrointestinal Tract. Digestion 2020; 102:1-8. [PMID: 33316807 PMCID: PMC8200364 DOI: 10.1159/000513216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/20/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cancers of the gastrointestinal (GI) tract are the common leading cause of cancer-related death in the world. Recent advances in cancer therapies such as intensive multidrug chemotherapy and molecular targeted treatment have improved therapeutic efficacy; however, the outcomes are not satisfied. Moreover, these therapies also cause severe side effects. New type of cancer therapies is urgently needed to improve the outcomes and to reduce side effects of GI tract cancers. SUMMARY This account is a comprehensive review article on the newly developed, photochemistry-based cancer therapy named as near-infrared photoimmunotherapy (NIR-PIT). NIR-PIT is a highly selective tumor treatment that employs an antibody-photoabsorber conjugate, which is activated by near-infrared light. A world-wide phase 3 clinical trial of NIR-PIT against recurrent head and neck cancer patients is currently underway. NIR-PIT differs from conventional cancer therapies such as surgery, chemotherapy, and radiation in its selectivity for killing cancer cells and cells treated with NIR-PIT leading to immunogenic cell death. Preclinical research in animals with combining cancer-targeting NIR-PIT and other cancer immunotherapies could lead to responses not only in local tumor but also in distant metastases. NIR-PIT also leads to an immediate and dramatic increase in vascular permeability after therapy. From these aspects, NIR-PIT appears to be a promising new form of cancer therapy. NIR-PIT could be readily translated into clinical use for virtually any cancers in the near future provided suitable humanized antibodies are available. Here, we describe the specific advantages and applications of NIR-PIT in the GI tract. Key Messages: We believe that NIR-PIT with NIR excitation light, which can be delivered via a fiber optic diffuser through endoscopes, is a promising method for a new treatment of GI cancers.
Collapse
Affiliation(s)
- Tadanobu Nagaya
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA,
- Department of Gastroenterology, Shinshu University Hospital, Matsumoto, Japan,
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
237
|
Musib D, Raza MK, Pal M, Roy M. A red light‐activable Mn
I
(CO)
3
‐functionalized gold nanocomposite as the anticancer prodrug with theranostic potential. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.6110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Dulal Musib
- Department of Chemistry National Institute of Technology, Manipur Imphal India
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry Indian Institute of Science Bangalore India
| | - Mrityunjoy Pal
- Department of Chemistry National Institute of Technology, Manipur Imphal India
| | - Mithun Roy
- Department of Chemistry National Institute of Technology, Manipur Imphal India
| |
Collapse
|
238
|
Domiński A, Konieczny T, Duale K, Krawczyk M, Pastuch-Gawołek G, Kurcok P. Stimuli-Responsive Aliphatic Polycarbonate Nanocarriers for Tumor-Targeted Drug Delivery. Polymers (Basel) 2020; 12:E2890. [PMID: 33276597 PMCID: PMC7761607 DOI: 10.3390/polym12122890] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/28/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Nanoparticles based on amphiphilic copolymers with tunable physicochemical properties can be used to encapsulate delicate pharmaceutics while at the same time improving their solubility, stability, pharmacokinetic properties, reducing immune surveillance, or achieving tumor-targeting ability. Those nanocarriers based on biodegradable aliphatic polycarbonates are a particularly promising platform for drug delivery due to flexibility in the design and synthesis of appropriate monomers and copolymers. Current studies in this field focus on the design and the synthesis of new effective carriers of hydrophobic drugs and their release in a controlled manner by exogenous or endogenous factors in tumor-specific regions. Reactive groups present in aliphatic carbonate copolymers, undergo a reaction under the action of a stimulus: e.g., acidic hydrolysis, oxidation, reduction, etc. leading to changes in the morphology of nanoparticles. This allows the release of the drug in a highly controlled manner and induces a desired therapeutic outcome without damaging healthy tissues. The presented review summarizes the current advances in chemistry and methods for designing stimuli-responsive nanocarriers based on aliphatic polycarbonates for controlled drug delivery.
Collapse
Affiliation(s)
- Adrian Domiński
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowskiej St, 41-819 Zabrze, Poland; (A.D.); (T.K.); (K.D.)
| | - Tomasz Konieczny
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowskiej St, 41-819 Zabrze, Poland; (A.D.); (T.K.); (K.D.)
| | - Khadar Duale
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowskiej St, 41-819 Zabrze, Poland; (A.D.); (T.K.); (K.D.)
| | - Monika Krawczyk
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland; (M.K.); (G.P.-G.)
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
| | - Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland; (M.K.); (G.P.-G.)
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
| | - Piotr Kurcok
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowskiej St, 41-819 Zabrze, Poland; (A.D.); (T.K.); (K.D.)
| |
Collapse
|
239
|
Nanodelivery of immunogenic cell death-inducers for cancer immunotherapy. Drug Discov Today 2020; 26:651-662. [PMID: 33278602 DOI: 10.1016/j.drudis.2020.11.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/22/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022]
|
240
|
Hejmady S, Pradhan R, Alexander A, Agrawal M, Singhvi G, Gorain B, Tiwari S, Kesharwani P, Dubey SK. Recent advances in targeted nanomedicine as promising antitumor therapeutics. Drug Discov Today 2020; 25:2227-2244. [DOI: 10.1016/j.drudis.2020.09.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/29/2020] [Accepted: 09/26/2020] [Indexed: 12/18/2022]
|
241
|
Zhou S, Shang Q, Wang N, Li Q, Song A, Luan Y. Rational design of a minimalist nanoplatform to maximize immunotherapeutic efficacy: Four birds with one stone. J Control Release 2020; 328:617-630. [DOI: 10.1016/j.jconrel.2020.09.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/27/2020] [Accepted: 09/15/2020] [Indexed: 12/25/2022]
|
242
|
Nicolson F, Ali A, Kircher MF, Pal S. DNA Nanostructures and DNA-Functionalized Nanoparticles for Cancer Theranostics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001669. [PMID: 33304747 PMCID: PMC7709992 DOI: 10.1002/advs.202001669] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/27/2020] [Indexed: 05/12/2023]
Abstract
In the last two decades, DNA has attracted significant attention toward the development of materials at the nanoscale for emerging applications due to the unparalleled versatility and programmability of DNA building blocks. DNA-based artificial nanomaterials can be broadly classified into two categories: DNA nanostructures (DNA-NSs) and DNA-functionalized nanoparticles (DNA-NPs). More importantly, their use in nanotheranostics, a field that combines diagnostics with therapy via drug or gene delivery in an all-in-one platform, has been applied extensively in recent years to provide personalized cancer treatments. Conveniently, the ease of attachment of both imaging and therapeutic moieties to DNA-NSs or DNA-NPs enables high biostability, biocompatibility, and drug loading capabilities, and as a consequence, has markedly catalyzed the rapid growth of this field. This review aims to provide an overview of the recent progress of DNA-NSs and DNA-NPs as theranostic agents, the use of DNA-NSs and DNA-NPs as gene and drug delivery platforms, and a perspective on their clinical translation in the realm of oncology.
Collapse
Affiliation(s)
- Fay Nicolson
- Department of ImagingDana‐Farber Cancer Institute & Harvard Medical SchoolBostonMA02215USA
- Center for Molecular Imaging and NanotechnologyMemorial Sloan Kettering Cancer CenterNew YorkNY10065USA
| | - Akbar Ali
- Department of ChemistryIndian Institute of Technology‐ BhilaiRaipurChhattisgarh492015India
| | - Moritz F. Kircher
- Department of ImagingDana‐Farber Cancer Institute & Harvard Medical SchoolBostonMA02215USA
- Center for Molecular Imaging and NanotechnologyMemorial Sloan Kettering Cancer CenterNew YorkNY10065USA
- Department of RadiologyBrigham and Women's Hospital & Harvard Medical SchoolBostonMA02215USA
| | - Suchetan Pal
- Department of ChemistryIndian Institute of Technology‐ BhilaiRaipurChhattisgarh492015India
| |
Collapse
|
243
|
Ferreira BL, Martel F, Silva C, Santos T, Daniel-da-Silva A. Nanostructured functionalized magnetic platforms for the sustained delivery of cisplatin: Synthesis, characterization and in vitro cytotoxicity evaluation. J Inorg Biochem 2020; 213:111258. [DOI: 10.1016/j.jinorgbio.2020.111258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 01/09/2023]
|
244
|
Yu X, Wu H, Hu H, Dong Z, Dang Y, Qi Q, Wang Y, Du S, Lu Y. Zein nanoparticles as nontoxic delivery system for maytansine in the treatment of non-small cell lung cancer. Drug Deliv 2020; 27:100-109. [PMID: 31870183 PMCID: PMC6968508 DOI: 10.1080/10717544.2019.1704942] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 01/04/2023] Open
Abstract
Purpose: Maytansine (DM1) is a potent anticancer drug and limited in clinical application due to its poor water solubility and toxic side effects. Zein is widely used in nano drug delivery systems due to its good biocompatibility. In this study, we prepared DM1-loaded zein nanoparticles (ZNPs) to achieve tumor targeting and reduce toxic side effects of DM1. Methods: ZNPs were prepared by phase separation and Box-Behnken design was used to optimize the formulation. Then, confocal fluorescence microscope and flow cytometry were used to determine cellular uptake of ZNPs. A549 cells were cultured in vitro to study cytotoxicity and used to establish tumor xenografts in nude mice. Biodistribution and antitumor activity of ZNPs were performed in vivo experiments. In addition, we also performed histological and immunohistochemical examinations on tumors and viscera. Results: The optimal prescription was obtained by using 120 μL zein added to 2 mL water under stirring in 300 rpm. The encapsulation efficiency and drug loading were 82.97 ± 0.80% and 3.32 ± 0.03%, respectively. We found that DM1-loaded ZNPs have a strong inhibitory effect on A549 cells, which stemmed from the ability of ZNPs to enhance cellular uptake. Furthermore, we demonstrated that DM1-loaded ZNPs exhibits a better antitumor efficacy than DM1, which tumor inhibition rate were 97.3% and 92.7%, respectively. The biodistribution revealed that ZNPs could targeted to tumor. Finally, we confirmed by histological that DM1-loaded ZNPs are nontoxic. Conclusion: DM1-loaded ZNPs have considerable antitumor activity. Thus, DM1-loaded ZNPs are a promising treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Xianglong Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huichao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haiyan Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziyi Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yunni Dang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Qi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shouying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
245
|
Yang S, Chen C, Qiu Y, Xu C, Yao J. Paying attention to tumor blood vessels: Cancer phototherapy assisted with nano delivery strategies. Biomaterials 2020; 268:120562. [PMID: 33278682 DOI: 10.1016/j.biomaterials.2020.120562] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022]
Abstract
Cancer phototherapy has attracted increasing attention for its promising effectiveness and relative non-invasiveness. Over the past years, tremendous efforts have been made to develop better phototherapy strategies with various nano delivery systems. This review introduces cancer phototherapy strategies based on tumor blood vessels for improved therapeutic outcomes from the angle of direct tumor destruction and improved delivery process assisted with nano delivery designs. Latest directions and ideas of cancer phototherapy with translation potential are also discussed. Focusing on the double role of tumor vessels not only as an anti-tumor target but also as part of the delivery process, we highlight the crosstalk between photo-induced extensive effects and the complicated drug delivery process. Due to the heterogeneity of tumors, deeper investigations about the interconnection between tumor vessels and cancer phototherapy remain to be carried out. More delicate and intelligent nano delivery systems are expected to help realize the full potential of this therapeutic strategy.
Collapse
Affiliation(s)
- Shan Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Chen Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Yue Qiu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Cheng Xu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
246
|
Pan Y, Luo Z, Wang X, Chen Q, Chen J, Guan Y, Liu D, Xu H, Liu J. A versatile and multifunctional metal-organic framework nanocomposite toward chemo-photodynamic therapy. Dalton Trans 2020; 49:5291-5301. [PMID: 32242552 DOI: 10.1039/c9dt04804a] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Previously most of the applications of targeting components have been based on the enhanced permeability and retention effect achieved using folic acid, which consider the side effects of the targeting components to some extent. Herein, we report a new strategy to decorate the surface of MOFs using a pemetrexed (MTA) targeting molecule, affording a new drug delivery system of ALA@UIO-66-NH-FAM/MTA (ALA = 5-amino-levulinic acid and FAM = 5-carboxyfluorescein). The confocal microscopy and flow cytometry results showed that ALA@UIO-66-NH-FAM/MTA presented a better targeting effect compared to ALA@UIO-66-NH-FAM/FA (FA = folic acid) and indicated a gradually increasing tendency of the targeting effect with the increasing expression of folate receptors on the tumor cell cytomembrane. Furthermore, the cytotoxicity experiment indicates that the combination of chemotherapy and photodynamic therapy is a more effective therapy model than single chemotherapy and photodynamic therapy. This work demonstrates the first attempt at folic acid antagonist (MTA) modification for NMOFs, providing a new concept for the design of MOFs with folate receptor targeting capacity for clinical applications.
Collapse
Affiliation(s)
- Ying Pan
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Zhidong Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Xiaoxiong Wang
- School of Civil and Environmental Engineering, Shenzhen Polytechnic, Shenzhen, 518055, China.
| | - Qianyi Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Junhao Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Yucheng Guan
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Dong Liu
- Shenzhen Huachuang Bio-pharmaceutical Technology Co. Ltd., Shenzhen 518112, China.
| | - Hongjia Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Jianqiang Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
247
|
Paliwal SR, Kenwat R, Maiti S, Paliwal R. Nanotheranostics for Cancer Therapy and Detection: State of the Art. Curr Pharm Des 2020; 26:5503-5517. [PMID: 33200696 DOI: 10.2174/1381612826666201116120422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 08/09/2020] [Indexed: 11/22/2022]
Abstract
Nanotheranostics, an approach of combining both diagnosis and therapy, is one of the latest advances in cancer therapy particularly. Nanocarriers designed and derived from inorganic materials such as like gold nanoparticles, silica nanoparticles, magnetic nanoparticles and carbon nanotubes have been explored for tremendous applications in this area. Similarly, nanoparticles composed of some organic material alone or in combination with inorganic nano-cargos have been developed pre-clinically and possess excellent features desired. Photothermal therapy, MRI, simultaneous imaging and delivery, and combination chemotherapy with a diagnosis are a few of the known methods exploring cancer therapy and detection at organ/tissue/molecular/sub-cellular level. This review comprises an overview of the recent reports meant for nano theranostics purposes. Targeted cancer nanotheranostics have been included for understating tumor micro-environment or cell-specific targeting approach employed. A brief account of various strategies is also included for the readers highlighting the mechanism of cancer therapy.
Collapse
Affiliation(s)
- Shivani Rai Paliwal
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilapsur, CG, India
| | - Rameshroo Kenwat
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP, India
| | - Sabyasachi Maiti
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP, India
| | - Rishi Paliwal
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP, India
| |
Collapse
|
248
|
Jarak I, Varela CL, Tavares da Silva E, Roleira FFM, Veiga F, Figueiras A. Pluronic-based nanovehicles: Recent advances in anticancer therapeutic applications. Eur J Med Chem 2020; 206:112526. [PMID: 32971442 DOI: 10.1016/j.ejmech.2020.112526] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
Abstract
Pluronics are a class of amphiphilic tri-block copolymers with wide pharmaceutical applicability. In the past decades, the ability to form biocompatible nanosized micelles was exploited to formulate stable drug nanovehicles with potential use in antitumor therapy. Due to the great potential for tuning physical and structural properties of Pluronic unimers, a panoply of drug or polynucleotide-loaded micelles was prepared and tested for their antitumoral activity. The attractive inherent antitumor properties of Pluronic polymers in combination with cell targeting and stimuli-responsive ligands greatly improved antitumoral therapeutic effects of tested drugs. In spite of that, the extraordinary complexity of biological challenges in the delivery of micellar drug payload makes their therapeutic potential still not exploited to the fullest. In this review paper we attempt to present the latest developments in the field of Pluronic based nanovehicles and their application in anticancer therapy with an overview of the chemistry involved in the preparation of these nanovehicles.
Collapse
Affiliation(s)
- Ivana Jarak
- Univ. Coimbra, Department of Pharmaceutical Technology, Faculty of Pharmacy, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal
| | - Carla L Varela
- Univ. Coimbra, CIEPQPF, FFUC, Laboratory of Pharmaceutical Chemistry, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal
| | - Elisiário Tavares da Silva
- Univ. Coimbra, CIEPQPF, FFUC, Laboratory of Pharmaceutical Chemistry, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal
| | - Fernanda F M Roleira
- Univ. Coimbra, CIEPQPF, FFUC, Laboratory of Pharmaceutical Chemistry, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal
| | - Francisco Veiga
- Univ. Coimbra, Department of Pharmaceutical Technology, Faculty of Pharmacy, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal; Univ. Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal
| | - Ana Figueiras
- Univ. Coimbra, Department of Pharmaceutical Technology, Faculty of Pharmacy, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal; Univ. Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal.
| |
Collapse
|
249
|
Li W, Suarato G, Cathcart JM, Sargunas PR, Meng Y. Design, characterization, and intracellular trafficking of biofunctionalized chitosan nanomicelles. Biointerphases 2020; 15:061003. [PMID: 33187397 PMCID: PMC7666618 DOI: 10.1116/6.0000380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022] Open
Abstract
The hydrophobically modified glycol chitosan (HGC) nanomicelle has received increasing attention as a promising platform for the delivery of chemotherapeutic drugs. To improve the tumor selectivity of HGC, here an avidin and biotin functionalization strategy was applied. The hydrodynamic diameter of the biotin-avidin-functionalized HGC (cy5.5-HGC-B4F) was observed to be 104.7 nm, and the surface charge was +3.1 mV. Confocal and structured illumination microscopy showed that at 0.1 mg/ml, cy5.5-HGC-B4F nanomicelles were distributed throughout the cytoplasm of MDA-MB-231 breast cancer cells after 2 h of exposure without significant cytotoxicity. To better understand the intracellular fate of the nanomicelles, entrapment studies were performed and demonstrated that some cy5.5-HGC-B4F nanomicelles were capable of escaping endocytic vesicles, likely via the proton sponge effect. Quantitative analysis of the movements of endosomes in living cells revealed that the addition of HGC greatly enhanced the motility of endosomal compartments, and the nanomicelles were transported by early and late endosomes from cell periphery to the perinuclear region. Our results validate the importance of using live-cell imaging to quantitatively assess the dynamics and mechanisms underlying the complex endocytic pathways of nanosized drug carriers.
Collapse
Affiliation(s)
- Weiyi Li
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| | - Giulia Suarato
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| | - Jillian M. Cathcart
- Department of Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York 11794
| | - Paul R. Sargunas
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| | - Yizhi Meng
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| |
Collapse
|
250
|
Das U, Bhuniya A, Roy AK, Gmeiner WH, Ghosh S. Hairpin Oligonucleotide Can Functionalize Gold Nanorods for in Vivo Application Delivering Cytotoxic Nucleotides and Curcumin: A Comprehensive Study in Combination with Near-Infrared Laser. ACS OMEGA 2020; 5:28463-28474. [PMID: 33195896 PMCID: PMC7658950 DOI: 10.1021/acsomega.0c02288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/30/2020] [Indexed: 06/11/2023]
Abstract
We prepared a multimodality nanocomplex by functionalizing gold nanorods (GNRs) with a cytotoxic nucleoside, 5-fluoro-2'-deoxyuridine (FdU) containing a DNA hairpin, along with complexation of pleiotropic molecule curcumin. Conjugates were investigated for anti-tumor activity using an Ehrlich carcinoma model in combination with 808 nm laser irradiation. We demonstrated that hairpin-functionalized GNRs are suitable for intravenous administration, including delivery of cytotoxic nucleotides and curcumin. Curcumin binding with FdU-hairpin-functionalized GNRs displayed improved anti-tumor activity in part by inducing a lymphocyte-mediated immune response. The complex showed notable photothermal activity in vitro; however, 808 nm laser irradiation of the tumor following treatment with the complex did not increase the anti-tumor effect significantly. Biodistribution studies depicted that the nanoconjugates localized primarily in the sinusoidal structures of the liver and spleen with minimal tumor accumulation. Curcumin complexation alleviated the reduction in the RBC count that was observed for the conjugate without curcumin, especially in combination with laser irradiation. Localization of FdU-hairpin-GNR conjugates in the liver and spleen evoked an inflammatory response, which was mitigated by curcumin complexation. However, no functional abnormality was found in the liver in any case. Curcumin binding also notably decreased nanoconjugate accumulation in lungs and significantly reduced inflammation. Biodistribution studies were consistent with previous reports, suggesting that optimization of the GNR size and surface coating is required for more efficient tumor localization via the enhanced permeability and retention (EPR) effect. Our studies demonstrate that DNA/RNA hairpins are suitable for GNR surface functionalization and enable delivery of cytotoxic nucleotides as well as curcumin in vivo with potential for synergistic anti-cancer therapy.
Collapse
Affiliation(s)
- Upasana Das
- Department
of Anti-Cancer Drug Development and Chemotherapy, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata 700026, India
| | - Avishek Bhuniya
- Department
of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata 700026, India
| | - Anup K. Roy
- Department
of Pathology, Nil Ratan Sircar Medical College
and Hospital, Kolkata 700014, India
| | - William H. Gmeiner
- Department
of Cancer Biology, Wake Forest School of
Medicine, Winston-Salem, North Carolina 27157, United States
| | - Supratim Ghosh
- Department
of Anti-Cancer Drug Development and Chemotherapy, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata 700026, India
| |
Collapse
|