1
|
Pandey PK, Jain M, Jha PK. Drug delivery from a ring implant attached to intraocular lens: An in-silico investigation. J Pharm Sci 2024; 113:3332-3343. [PMID: 39245324 DOI: 10.1016/j.xphs.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/10/2024]
Abstract
Multiple iterations required to design ocular implants, which will last for the desired operational period of months or even years, necessitate the use of in-silico models for ocular drug delivery. In this study, we developed an in-silico model to simulate the flow of Aqueous Humor (AH) and drug delivery from an implant to the Trabecular Meshwork (TM). The implant, attached to the side of the intraocular lens (IOL), and the TM are treated as porous media, with their effects on AH flow accounted for using the Darcy equation. This model accurately predicts the physiological values of Intraocular Pressure (IOP) for both healthy individuals and glaucoma patients, as reported in the literature. Results reveal that the effective diffusivity of the drug within the implant is the critical parameter that can alter the bioavailability time period (BTP) from a few days to months. Intuitively, BTP should increase as effective diffusivity decreases. However, we discovered that with lower levels of initial drug loading, BTP declines when effective diffusivity falls below a specific threshold. Our findings further reveal that, while AH flow has a minimal effect on the drug release profile at the implant site, it significantly impacts drug availability at the TM.
Collapse
Affiliation(s)
- Pawan Kumar Pandey
- Department of Chemical Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | | | - Prateek K Jha
- Department of Chemical Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
2
|
Xi X, Lei F, Gao K, Li J, Liu R, Karpf AR, Bronich TK. Ligand-installed polymeric nanocarriers for combination chemotherapy of EGFR-positive ovarian cancer. J Control Release 2023; 360:872-887. [PMID: 37478915 DOI: 10.1016/j.jconrel.2023.07.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Combination chemotherapeutic drugs administered via a single nanocarrier for cancer treatment provides benefits in reducing dose-limiting toxicities, improving the pharmacokinetic properties of the cargo and achieving spatial-temporal synchronization of drug exposure for maximized synergistic therapeutic effects. In an attempt to develop such a multi-drug carrier, our work focuses on functional multimodal polypeptide-based polymeric nanogels (NGs). Diblock copolymers poly (ethylene glycol)-b-poly (glutamic acid) (PEG-b-PGlu) modified with phenylalanine (Phe) were successfully synthesized and characterized. Self-assembly behavior of the resulting polymers was utilized for the synthesis of NGs with hydrophobic domains in cross-linked polyion cores coated with inert PEG chains. The resulting NGs were small (ca. 70 nm in diameter) and were able to encapsulate the combination of drugs with different physicochemical properties such as cisplatin and neratinib. Drug combination-loaded NGs exerted a selective synergistic cytotoxicity towards EGFR overexpressing ovarian cancer cells. Moreover, we developed ligand-installed EGFR-targeted NGs and tested them as an EGFR-overexpressing tumor-specific delivery system. Both in vitro and in vivo, ligand-installed NGs displayed preferential associations with EGFR (+) tumor cells. Ligand-installed NGs carrying cisplatin and neratinib significantly improved the treatment response of ovarian cancer xenografts. We also confirmed the importance of simultaneous administration of the dual drug combination via a single NG system which provides more therapeutic benefit than individual drug-loaded NGs administered at equivalent doses. This work illustrates the potential of our carrier system to mediate efficient delivery of a drug combination to treat EGFR overexpressing cancers.
Collapse
Affiliation(s)
- Xinyuan Xi
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Fan Lei
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Keliang Gao
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7363, USA
| | - Jingjing Li
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7363, USA
| | - Rihe Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7363, USA
| | - Adam R Karpf
- Eppley Institute for Research in Cancer and Allied Diseases and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
3
|
A pH-Responsive Asymmetric Microfluidic/Chitosan Device for Drug Release in Infective Bone Defect Treatment. Int J Mol Sci 2023; 24:ijms24054616. [PMID: 36902046 PMCID: PMC10003349 DOI: 10.3390/ijms24054616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/12/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
Bacterial infection is currently considered to be one of the major reasons that leads to the failure of guided bone regeneration (GBR) therapy. Under the normal condition, the pH is neutral, while the microenvironment will become acid at the sites of infection. Here, we present an asymmetric microfluidic/chitosan device that can achieve pH-responsive drug release to treat bacterial infection and promote osteoblast proliferation at the same time. On-demand release of minocycline relies on a pH-sensitive hydrogel actuator, which swells significantly when exposed to the acid pH of an infected region. The PDMAEMA hydrogel had pronounced pH-sensitive properties, and a large volume transition occurred at pH 5 and 6. Over 12 h, the device enabled minocycline solution flowrates of 0.51-1.63 µg/h and 0.44-1.13 µg/h at pH 5 and 6, respectively. The asymmetric microfluidic/chitosan device exhibited excellent capabilities for inhibiting Staphylococcus aureus and Streptococcus mutans growth within 24 h. It had no negative effect on proliferation and morphology of L929 fibroblasts and MC3T3-E1 osteoblasts, which indicates good cytocompatibility. Therefore, such a pH-responsive drug release asymmetric microfluidic/chitosan device could be a promising therapeutic approach in the treatment of infective bone defects.
Collapse
|
4
|
The emerging role of 3D-printing in ocular drug delivery: Challenges, current status, and future prospects. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
5
|
Amalia E, Diantini A, Endang Prabandari E, Waluyo D, Subarnas A. Caffeic Acid Phenethyl Ester as a DHODH Inhibitor and Its Synergistic Anticancer Properties in Combination with 5-Fluorouracil in a Breast Cancer Cell Line. J Exp Pharmacol 2022; 14:243-253. [PMID: 35910085 PMCID: PMC9329448 DOI: 10.2147/jep.s365159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction A combination of chemotherapy agents is the best choice in breast cancer treatment to increase the patient survival rate. 5-fluorouracil (5-FU) is one of the drugs applied in combination with other drugs to control and delay development of cancer cells. Nevertheless, the occurrence of multidrug resistance and dose-limiting cytotoxicity have limited the efficacy of 5-FU treatment. Therefore, the discovery of new anti-breast cancer drugs should be pursued. Objective To study potency of a promising naturally derived compound, caffeic acid phenethyl ester (CAPE), for breast cancer treatment in single and combination with 5-FU. Methods Cytotoxicity of CAPE, 5-FU, and 5-FU+CAPE was studied by in vitro MTT experiment in MCF-7 cell line, and RT-PCR analysis was used to evaluate the change in gene expression due to the treatment. Moreover, an enzymatic assay and molecular docking analysis were applied to evaluate the possible mechanism of substance-induced apoptosis. Results The study revealed that a single treatment of CAPE showed cytotoxicity with IC50 6.6 ± 1.0 µM and 6.5 ± 2.9 µM at 24 h and 48 h, respectively. Meanwhile, 5-FU showed cytostatic activity. The 5-FU + CAPE has a synergistic effect at 24 h treatment with a CI = 0.5 and an additive effect at 48 h treatment with CI = 1.0. CAPE was also found to enhances the mRNA expression of caspase-8 and BAX within 6 hours in combination with 5-FU compared to 5-FU treatment alone. Our study reveals a new mechanism of CAPE which is related to the inhibition of human dihydroorotate dehydrogenase (HsDHODH) with an IC50 of 120.7 ± 6.8 µM, by bound to the ubiquinone-binding site of the enzyme and could be responsible for inducing extrinsic and intrinsic apoptosis. Conclusion This study demonstrated the cytotoxicity of CAPE potential to induce apoptosis of breast cancer MCF-7 cell line single and cytotoxic-cytostatic combination with 5-FU. Therefore, further studies to develop CAPE and its derivatives will be required to discover new candidates for breast cancer agents.
Collapse
Affiliation(s)
- Eri Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia.,Department of Pharmacology, Faculty of Science and Technology, Department of Pharmacy, Muhammadiyah University, Bandung, Indonesia.,Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | | | - Danang Waluyo
- Research Center for Vaccine and Drug, National Research and Innovation Agency, Bogor, Indonesia
| | - Anas Subarnas
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
6
|
Larochelle RD, Mann SE, Ifantides C. 3D Printing in Eye Care. Ophthalmol Ther 2021; 10:733-752. [PMID: 34327669 PMCID: PMC8320416 DOI: 10.1007/s40123-021-00379-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Three-dimensional printing enables precise modeling of anatomical structures and has been employed in a broad range of applications across medicine. Its earliest use in eye care included orbital models for training and surgical planning, which have subsequently enabled the design of custom-fit prostheses in oculoplastic surgery. It has evolved to include the production of surgical instruments, diagnostic tools, spectacles, and devices for delivery of drug and radiation therapy. During the COVID-19 pandemic, increased demand for personal protective equipment and supply chain shortages inspired many institutions to 3D-print their own eye protection. Cataract surgery, the most common procedure performed worldwide, may someday make use of custom-printed intraocular lenses. Perhaps its most alluring potential resides in the possibility of printing tissues at a cellular level to address unmet needs in the world of corneal and retinal diseases. Early models toward this end have shown promise for engineering tissues which, while not quite ready for transplantation, can serve as a useful model for in vitro disease and therapeutic research. As more institutions incorporate in-house or outsourced 3D printing for research models and clinical care, ethical and regulatory concerns will become a greater consideration. This report highlights the uses of 3D printing in eye care by subspecialty and clinical modality, with an aim to provide a useful entry point for anyone seeking to engage with the technology in their area of interest.
Collapse
Affiliation(s)
- Ryan D Larochelle
- Department of Ophthalmology, University of Colorado, Sue Anschutz-Rodgers Eye Center, 1675 Aurora Court, F731, Aurora, CO, 80045, USA
| | - Scott E Mann
- Department of Otolaryngology, University of Colorado, Aurora, CO, USA
- Department of Surgery, Denver Health Medical Center, Denver, CO, USA
| | - Cristos Ifantides
- Department of Ophthalmology, University of Colorado, Sue Anschutz-Rodgers Eye Center, 1675 Aurora Court, F731, Aurora, CO, 80045, USA.
- Department of Surgery, Denver Health Medical Center, Denver, CO, USA.
| |
Collapse
|
7
|
Pinho LAG, Gratieri T, Gelfuso GM, Marreto RN, Cunha-Filho M. Three-dimensional printed personalized drug devices with anatomical fit: a review. J Pharm Pharmacol 2021; 74:1391-1405. [PMID: 34665263 DOI: 10.1093/jpp/rgab146] [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: 06/28/2021] [Accepted: 09/24/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVES Three-dimensional printing (3DP) has opened the era of drug personalization, promising to revolutionize the pharmaceutical field with improvements in efficacy, safety and compliance of the treatments. As a result of these investigations, a vast therapeutic field has opened for 3DP-loaded drug devices with an anatomical fit. Along these lines, innovative dosage forms, unimaginable until recently, can be obtained. This review explores 3DP-engineered drug devices described in recent research articles, as well as in patented inventions, and even devices already produced by 3DP with drug-loading potential. KEY FINDINGS 3D drug-loaded stents, implants and prostheses are reviewed, along with devices produced to fit hard-to-attach body parts such as nasal masks, vaginal rings or mouthguards. The most promising 3DP techniques for such devices and the complementary technologies surrounding these inventions are also discussed, particularly the scanners useful for mapping body parts. Health regulatory concerns regarding the new use of such technology are also analysed. SUMMARY The scenario discussed in this review shows that for wearable 3DP drug devices to become a tangible reality to users, it will be necessary to overcome the existing regulatory barriers, create new interfaces with electronic systems and improve the mapping mechanisms of body surfaces.
Collapse
Affiliation(s)
- Ludmila A G Pinho
- Laboratory of Food, Drug, and Cosmetics (LTMAC), School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Tais Gratieri
- Laboratory of Food, Drug, and Cosmetics (LTMAC), School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Guilherme M Gelfuso
- Laboratory of Food, Drug, and Cosmetics (LTMAC), School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Ricardo Neves Marreto
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Marcilio Cunha-Filho
- Laboratory of Food, Drug, and Cosmetics (LTMAC), School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| |
Collapse
|
8
|
García-Estrada P, García-Bon MA, López-Naranjo EJ, Basaldúa-Pérez DN, Santos A, Navarro-Partida J. Polymeric Implants for the Treatment of Intraocular Eye Diseases: Trends in Biodegradable and Non-Biodegradable Materials. Pharmaceutics 2021; 13:701. [PMID: 34065798 PMCID: PMC8151640 DOI: 10.3390/pharmaceutics13050701] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022] Open
Abstract
Intraocular/Intravitreal implants constitute a relatively new method to treat eye diseases successfully due to the possibility of releasing drugs in a controlled and prolonged way. This particularity has made this kind of method preferred over other methods such as intravitreal injections or eye drops. However, there are some risks and complications associated with the use of eye implants, the body response being the most important. Therefore, material selection is a crucial factor to be considered for patient care since implant acceptance is closely related to the physical and chemical properties of the material from which the device is made. In this regard, there are two major categories of materials used in the development of eye implants: non-biodegradables and biodegradables. Although non-biodegradable implants are able to work as drug reservoirs, their surgical requirements make them uncomfortable and invasive for the patient and may put the eyeball at risk. Therefore, it would be expected that the human body responds better when treated with biodegradable implants due to their inherent nature and fewer surgical concerns. Thus, this review provides a summary and discussion of the most common non-biodegradable and biodegradable materials employed for the development of experimental and commercially available ocular delivery implants.
Collapse
Affiliation(s)
- Paulina García-Estrada
- Departamento de Ingenieria de Proyectos-CUCEI, Universidad de Guadalajara, C.P. 45157 Zapopan, Mexico; (P.G.-E.); (M.A.G.-B.); (E.J.L.-N.); (D.N.B.-P.)
| | - Miguel A. García-Bon
- Departamento de Ingenieria de Proyectos-CUCEI, Universidad de Guadalajara, C.P. 45157 Zapopan, Mexico; (P.G.-E.); (M.A.G.-B.); (E.J.L.-N.); (D.N.B.-P.)
| | - Edgar J. López-Naranjo
- Departamento de Ingenieria de Proyectos-CUCEI, Universidad de Guadalajara, C.P. 45157 Zapopan, Mexico; (P.G.-E.); (M.A.G.-B.); (E.J.L.-N.); (D.N.B.-P.)
| | - Dulce N. Basaldúa-Pérez
- Departamento de Ingenieria de Proyectos-CUCEI, Universidad de Guadalajara, C.P. 45157 Zapopan, Mexico; (P.G.-E.); (M.A.G.-B.); (E.J.L.-N.); (D.N.B.-P.)
| | - Arturo Santos
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, C.P. 45138 Zapopan, Mexico;
| | - Jose Navarro-Partida
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, C.P. 45138 Zapopan, Mexico;
| |
Collapse
|
9
|
Brown T, McElroy T, Simmons P, Walters H, Ntagwabira F, Wang J, Byrum SD, Allen AR. Cognitive impairment resulting from treatment with docetaxel, doxorubicin, and cyclophosphamide. Brain Res 2021; 1760:147397. [PMID: 33705788 DOI: 10.1016/j.brainres.2021.147397] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 01/08/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer among women and it is estimated that about 30% of newly diagnosed cancers in women will be breast cancers. While advancements in treating breast cancer have led to an average 5-year survival rate of 90%, many survivors experience cognitive impairments as a result of chemotherapy treatment. Doxorubicin, cyclophosphamide, and docetaxel (TAC) are commonly administered as breast cancer treatments; however, there are few studies that have tested the cognitive effects of TAC. In the current study, 12-week-old female C57BL/6 mice received 4 weekly intraperitoneal injections of either saline or a combination therapy of doxorubicin and cyclophosphamide followed by 4 weekly docetaxel injections. Four weeks after the last injection, mice were tested for hippocampus-dependent cognitive performance in the Y-maze and the Morris water maze. During Y-maze testing, mice exposed to TAC exhibited impairment. During the water maze assessment, all animals were able to locate the visible and hidden platform locations. However, mice that received the TAC presented with a significant impairment in spatial memory retention on the probe trial days. TAC treatment significantly decreases the dendritic complexity of arborization in the dentate gyrus region of the hippocampus. In addition, comparative proteomic analysis revealed downregulation of proteins within key metabolic and signaling pathways associated with cognitive dysfunction, such as oxidative phosphorylation, ephrin signaling, and calcium signaling.
Collapse
Affiliation(s)
- Taurean Brown
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Huddoy Walters
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Fabio Ntagwabira
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Arkansas Children's Research Institute, Little Rock, AR 72202, United States
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
10
|
Fouling in ocular devices: implications for drug delivery, bioactive surface immobilization, and biomaterial design. Drug Deliv Transl Res 2021; 11:1903-1923. [PMID: 33454927 DOI: 10.1007/s13346-020-00879-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
The last 30 years has seen a proliferation of research on protein-resistant biomaterials targeted at designing bio-inert surfaces, which are prerequisite for optimal performance of implantable devices that contact biological fluids and tissues. These efforts have only been able to yield minimal results, and hence, the ideal anti-fouling biomaterial has remained elusive. Some studies have yielded biomaterials with a reduced fouling index among which high molecular weight polyethylene glycols have remained dominant. Interestingly, the field of implantable ocular devices has not experienced an outflow of research in this area, possibly due to the assumption that biomaterials tested in other body fluids can be translated for application in the ocular space. Unfortunately, progression in the molecular understanding of many ocular conditions has brought to the fore the need for treatment options that necessitates the use of anti-fouling biomaterials. From the earliest implanted horsehair and silk seton for glaucoma drainage to the recent mini telescopes for sight recovery, this review provides a concise incursion into the gradual evolution of biomaterials for the design of implantable ocular devices as well as approaches used to overcome the challenges with fouling. The implication of fouling for drug delivery, the design of immune-responsive biomaterials, as well as advanced surface immobilization approaches to support the overall performance of implantable ocular devices are also reviewed.
Collapse
|
11
|
Wei Z, Volkova E, Blatchley MR, Gerecht S. Hydrogel vehicles for sequential delivery of protein drugs to promote vascular regeneration. Adv Drug Deliv Rev 2019; 149-150:95-106. [PMID: 31421149 PMCID: PMC6889011 DOI: 10.1016/j.addr.2019.08.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/04/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
In recent years, as the mechanisms of vasculogenesis and angiogenesis have been uncovered, the functions of various pro-angiogenic growth factors (GFs) and cytokines have been identified. Therefore, therapeutic angiogenesis, by delivery of GFs, has been sought as a treatment for many vascular diseases. However, direct injection of these protein drugs has proven to have limited clinical success due to their short half-lives and systemic off-target effects. To overcome this, hydrogel carriers have been developed to conjugate single or multiple GFs with controllable, sustained, and localized delivery. However, these attempts have failed to account for the temporal complexity of natural angiogenic pathways, resulting in limited therapeutic effects. Recently, the emerging ideas of optimal sequential delivery of multiple GFs have been suggested to better mimic the biological processes and to enhance therapeutic angiogenesis. Incorporating sequential release into drug delivery platforms will likely promote the formation of neovasculature and generate vast therapeutic potential.
Collapse
Affiliation(s)
- Zhao Wei
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eugenia Volkova
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michael R Blatchley
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Nys G, Fillet M. Microfluidics contribution to pharmaceutical sciences: From drug discovery to post marketing product management. J Pharm Biomed Anal 2018; 159:348-362. [DOI: 10.1016/j.jpba.2018.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/18/2022]
|
13
|
Roy A, Sarker S, Upadhyay P, Pal A, Adhikary A, Jana K, Ray M. Methylglyoxal at metronomic doses sensitizes breast cancer cells to doxorubicin and cisplatin causing synergistic induction of programmed cell death and inhibition of stemness. Biochem Pharmacol 2018; 156:322-339. [PMID: 30170097 DOI: 10.1016/j.bcp.2018.08.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022]
Abstract
Potent anticancer activity coupled with absence of toxicity at therapeutic dose established the glycolytic metabolite, methylglyoxal, as a promising candidate against malignant neoplasia. In this preclinical study we illustrate the applicability of methylglyoxal in formulating an optimally designed combination regimen with chemotherapeutic drugs against breast cancer. Results demonstrated a synergistic augmentation in doxorubicin and cisplatin mediated cytotoxicity in human breast cancer cell lines MDA MB 231 & MCF 7 with methylglyoxal co-treatment at metronomic concentrations. The cell death due to combination treatment was significantly prevented by N-Acetylcysteine and the synergistic effects were attenuated in presence of inhibitors for apoptosis and necroptosis, in MDA MB 231 and MCF 7 cells, respectively. Additionally, acridine orange staining and immunoblotting with LC3B antibody indicated the suppression of doxorubicin induced autophagy flux with methylglyoxal co-treatment. This report documents for the first time the preferential targeting of breast cancer stem cells by methylglyoxal. Combination treatment with doxorubicin or cisplatin hindered mammosphere forming efficiency and inclusively eliminated both cancer stem as well as non-stem cancer cells. The synergistic effect was validated in Ehrlich mammary carcinoma cell induced murine ascites model and the combination advantage in vivo was achieved without any additional deleterious effect to liver and kidney. Our present study evidences the implications of methylglyoxal inclusion in adjuvant multimodal chemotherapeutics against breast cancer and offers noteworthy insights into the possible outcome.
Collapse
Affiliation(s)
- Anirban Roy
- Department of Biophysics, Bose Institute, P 1/12 CIT Scheme VII M, Kolkata 700 054, WB, India
| | - Sushmita Sarker
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, JD-2, Salt Lake City, Kolkata 700 098, WB, India
| | - Priyanka Upadhyay
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, JD-2, Salt Lake City, Kolkata 700 098, WB, India
| | - Aparajita Pal
- Department of Biophysics, Bose Institute, P 1/12 CIT Scheme VII M, Kolkata 700 054, WB, India
| | - Arghya Adhikary
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, JD-2, Salt Lake City, Kolkata 700 098, WB, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, P 1/12 CIT Scheme VII M, Kolkata 700 054, WB, India.
| | - Manju Ray
- Department of Biophysics, Bose Institute, P 1/12 CIT Scheme VII M, Kolkata 700 054, WB, India; Department of Chemistry, Institute of Applied Science & Humanities GLA University Mathura, 17km Stone, NH-2, Mathura-Delhi Road, Mathura 281 406, UP, India.
| |
Collapse
|
14
|
Chang CE, Hsieh CM, Chen LC, Su CY, Liu DZ, Jhan HJ, Ho HO, Sheu MT. Novel application of pluronic lecithin organogels (PLOs) for local delivery of synergistic combination of docetaxel and cisplatin to improve therapeutic efficacy against ovarian cancer. Drug Deliv 2018; 25:632-643. [PMID: 29463123 PMCID: PMC6058476 DOI: 10.1080/10717544.2018.1440444] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The synergistic combination of docetaxel (DTX) and cisplatin (CIS) by local drug delivery with a pluronic lecithin organogel (PLO) to facilitate high drug concentrations at tumor sites and less nonspecific distribution to normal organs is thought to be beneficial in chemotherapy. In this study, using Capryol-90 (C90) with the addition of lecithin as the oil phase was developed to carry DTX, which was then incorporated into a PLO-containing CIS to formulate a dual-drug injectable PLO for local delivery. An optimal PLO composite, P13L0.15O1.5, composed of PF127:lecithin:C90 at a 13:0.15:1.5 weight ratio was obtained. The sol-gel transition temperature of P13L0.15O1.5 was found to be 33 °C. Tumor inhibition studies illustrated that DTX/CIS-loaded P13L0.15O1.5 could efficiently suppress tumor growth by both intratumoral and peritumoral injections in SKOV-3 xenograft mouse model. Pharmacokinetic studies showed that subcutaneous administration of P13L0.15O1.5 was able to sustain the release of DTX and CIS leading to their slow absorption into the systemic circulation resulting in lower area under the plasma concentration curve at 0-72 h (AUC0-72) and maximum concentration (Cmax) values but longer half-life (T1/2) and mean residence time (MRT) values. An in vivo biodistribution study showed lower DTX and CIS concentrations in organs compared to other treatment groups after IT administration of the dual drug-loaded P13L0.15O1.5. It was concluded that the local co-delivery of DTX and CIS by PLOs may be a promising and effective platform for local anticancer drug delivery with minimal systemic toxicities.
Collapse
Affiliation(s)
- Chia-En Chang
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Chien-Ming Hsieh
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ling-Chun Chen
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC.,b Department of Biotechnology and Pharmaceutical Technology , Yuanpei University of Medical Technology , Hsinchu , Taiwan, ROC
| | - Chia-Yu Su
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Der-Zen Liu
- c Graduate Institute of Biomedical Materials and Engineering , Taipei Medical University , Taipei , Taiwan, ROC
| | - Hua-Jing Jhan
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Hsiu-O Ho
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ming-Thau Sheu
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC.,d Clinical Research Center and Traditional Herbal Medicine Research Center , Taipei Medical University Hospital , Taipei , Taiwan, ROC
| |
Collapse
|
15
|
Kothari IR, Italiya KS, Sharma S, Mittal A, Chitkara D. A Rapid and Precise Liquid Chromatographic Method for Simultaneous Determination of Alpha Lipoic Acid and Docetaxel in Lipid-Based Nanoformulations. J Chromatogr Sci 2018; 56:888-894. [DOI: 10.1093/chromsci/bmy064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 06/07/2018] [Indexed: 11/13/2022]
Affiliation(s)
- Ishit R Kothari
- Industrial Research Laboratory (IRL), Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan, India
| | - Kishan S Italiya
- Industrial Research Laboratory (IRL), Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan, India
| | - Saurabh Sharma
- Industrial Research Laboratory (IRL), Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan, India
| | - Anupama Mittal
- Industrial Research Laboratory (IRL), Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan, India
| | - Deepak Chitkara
- Industrial Research Laboratory (IRL), Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan, India
| |
Collapse
|
16
|
Bloomquist CJ, Mecham MB, Paradzinsky MD, Janusziewicz R, Warner SB, Luft JC, Mecham SJ, Wang AZ, DeSimone JM. Controlling release from 3D printed medical devices using CLIP and drug-loaded liquid resins. J Control Release 2018; 278:9-23. [PMID: 29596874 DOI: 10.1016/j.jconrel.2018.03.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/12/2018] [Accepted: 03/23/2018] [Indexed: 10/17/2022]
Abstract
Mass customization along with the ability to generate designs using medical imaging data makes 3D printing an attractive method for the fabrication of patient-tailored drug and medical devices. Herein we describe the application of Continuous Liquid Interface Production (CLIP) as a method to fabricate biocompatible and drug-loaded devices with controlled release properties, using liquid resins containing active pharmaceutical ingredients (API). In this work, we characterize how the release kinetics of a model small molecule, rhodamine B-base (RhB), are affected by device geometry, network crosslink density, and the polymer composition of polycaprolactone- and poly (ethylene glycol)-based networks. To demonstrate the applicability of using API-loaded liquid resins with CLIP, the UV stability was evaluated for a panel of clinically-relevant small molecule drugs. Finally, select formulations were tested for biocompatibility, degradation and encapsulation of docetaxel (DTXL) and dexamethasone-acetate (DexAc). Formulations were shown to be biocompatible over the course of 175 days of in vitro degradation and the clinically-relevant drugs could be encapsulated and released in a controlled fashion. This study reveals the potential of the CLIP manufacturing platform to serve as a method for the fabrication of patient-specific medical and drug-delivery devices for personalized medicine.
Collapse
Affiliation(s)
- Cameron J Bloomquist
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael B Mecham
- Lineberger Comprehensive Cancer Center Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark D Paradzinsky
- Lineberger Comprehensive Cancer Center Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rima Janusziewicz
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel B Warner
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA
| | - J Christopher Luft
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sue J Mecham
- Lineberger Comprehensive Cancer Center Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrew Z Wang
- Lineberger Comprehensive Cancer Center Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Joseph M DeSimone
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA; Department of Chemical and Biomedical Engineering, North Carolina State University, Raleigh, NC 27695, USA; Carbon, Redwood City, CA 94063, USA.
| |
Collapse
|
17
|
Bonde GV, Yadav SK, Chauhan S, Mittal P, Ajmal G, Thokala S, Mishra B. Lapatinib nano-delivery systems: a promising future for breast cancer treatment. Expert Opin Drug Deliv 2018. [DOI: 10.1080/17425247.2018.1449832] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Gunjan Vasant Bonde
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Sarita Kumari Yadav
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
- Department of Pharmacy, Moti Lal Nehru Medical College, Allahabad, India
| | - Sheetal Chauhan
- Department of Pharmacology, Melaka Manipal Medical College, Manipal University, Manipal, India
| | - Pooja Mittal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Gufran Ajmal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Sathish Thokala
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
18
|
Oduola WO, Li X, Duan C, Qian L, Wu F, Dougherty ER. Time-Based Switching Control of Genetic Regulatory Networks: Toward Sequential Drug Intake for Cancer Therapy. Cancer Inform 2017; 16:1176935117706888. [PMID: 28579741 PMCID: PMC5431492 DOI: 10.1177/1176935117706888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/26/2017] [Indexed: 11/17/2022] Open
Abstract
As cancer growth and development typically involves multiple genes and pathways, combination therapy has been touted as the standard of care in the treatment of cancer. However, drug toxicity becomes a major concern whenever a patient takes 2 or more drugs simultaneously at the maximum tolerable dosage. A potential solution would be administering the drugs in a sequential or alternating manner rather than concurrently. This study therefore examines the feasibility of such an approach from a switched system control perspective. Particularly, we study how genetic regulatory systems respond to sequential (switched) drug inputs using the time-based switching mechanism. The design of the time-driven drug switching function guarantees the stability of the genetic regulatory system and the repression of the diseased genes. Simulation results using proof-of-concept models and the proliferation and survival pathways with sequential drug inputs show the effectiveness of the proposed approach.
Collapse
Affiliation(s)
- Wasiu Opeyemi Oduola
- Department of Electrical and Computer Engineering, Prairie View A&M University, Prairie View, TX, USA
| | - Xiangfang Li
- Department of Electrical and Computer Engineering, Prairie View A&M University, Prairie View, TX, USA
| | - Chang Duan
- Department of Mechanical Engineering, Prairie View A&M University, Prairie View, TX, USA
| | - Lijun Qian
- Department of Electrical and Computer Engineering, Prairie View A&M University, Prairie View, TX, USA
| | - Fen Wu
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, USA
| | - Edward R Dougherty
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, TX, USA
| |
Collapse
|
19
|
Sauvage F, Messaoudi S, Fattal E, Barratt G, Vergnaud-Gauduchon J. Heat shock proteins and cancer: How can nanomedicine be harnessed? J Control Release 2017; 248:133-143. [PMID: 28088573 DOI: 10.1016/j.jconrel.2017.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 01/08/2017] [Indexed: 12/18/2022]
Abstract
Heat shock protein (hsp90) is an interesting target for cancer therapy because it is involved in the folding and stabilization of numerous proteins, including many that contribute to the development of cancer. It is part of the chaperone machinery that includes other heat shock proteins (hsp70, hsp27, hsp40) and is mainly localized in the cytosol, although many analogues or isoforms can be found in mitochondrion, endoplasmic reticulum and the cell membrane. Many potential inhibitors of hsp90 have been tested for cancer therapy but their usefulness is limited by their poor solubility in water and their ability to reach the target cells and the correct intracellular compartment. Nanomedicine, the incorporation of active molecules into an appropriate delivery system, could provide a solution to these drawbacks. In this review, we explain the rationale for using nanomedicine for this sort of cancer therapy, considering the properties of the chaperone machinery and of the different hsp90 analogues. We present some results that have already been obtained and put forward some strategies for delivery of hsp90 analogues to specific organelles.
Collapse
Affiliation(s)
- Félix Sauvage
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France
| | - Samir Messaoudi
- BioCIS-UMR 8076, Univ. Paris-Sud, CNRS, University Paris-Saclay, Châtenay-Malabry, 92296, France
| | - Elias Fattal
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France
| | - Gillian Barratt
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France
| | - Juliette Vergnaud-Gauduchon
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France.
| |
Collapse
|
20
|
Zakerzadeh E, Alizadeh E, Samadi Kafil H, Mohammad Hassanzadeh A, Salehi R, Mahkam M. Novel antibacterial polymeric nanocomposite for smart co-delivery of anticancer drugs. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:1509-1520. [DOI: 10.1080/21691401.2016.1260576] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Elham Zakerzadeh
- Chemistry Department, Azarbaijan Shahid Madani University, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Nanotechnology, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Roya Salehi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Nanotechnology, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Mahkam
- Chemistry Department, Azarbaijan Shahid Madani University, Tabriz, Iran
| |
Collapse
|
21
|
Polymer-lipid hybrid nanoparticles synchronize pharmacokinetics of co-encapsulated doxorubicin-mitomycin C and enable their spatiotemporal co-delivery and local bioavailability in breast tumor. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1279-90. [PMID: 26772427 DOI: 10.1016/j.nano.2015.12.383] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/13/2015] [Accepted: 12/22/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED Effective combination chemotherapy requires the delivery of drugs of synergism to tumor sites while sparing normal tissues. Herein we investigated whether coencapsulation of doxorubicin and mitomycin C within polymer-lipid hybrid nanoparticles (DMPLN) achieved this goal via ratiometric drugs in an orthotopic murine breast tumor model with nanocarrier-modified biodistribution, pharmacokinetics, local bioavailability and toxicity. Fluorescence imaging revealed quickened and extended tumor uptake but reduced cardiac accumulation of DMPLN. Quantitative drug analysis demonstrated prolonged systemic circulation, increased tumor accumulation and sustained synergistic ratios of doxorubicin and mitomycin C delivered by DMPLN over 24h. Higher levels of tumor cell apoptosis and reduced organ toxicity were obtained with DMPLN compared to free drug cocktails. DMPLN released DOX in tumors more efficiently than that from liposomal doxorubicin, as evidenced by a higher extent of the metabolite, doxorubicinol. These findings substantiate the importance of rational design of nanoparticles for synergistic drug combination therapy. FROM THE CLINICAL EDITOR The treatment of cancer usually involves using combination chemotherapeutic agents. In adopting a nanomedicine approach, one can in theory design combination therapy consisting of drugs of synergistic activities, with the aim to target tumor specifically while minimizing systemic toxicity. The authors in this study provided evidence for this rational design by co-encapsulation of doxorubicin and mitomycin C within polymer-lipid hybrid nanoparticles (DMPLN) in a breast cancer model.
Collapse
|
22
|
Parthasarathy R, Chow KM, Derafshi Z, Fautsch MP, Hetling JR, Rodgers DW, Hersh LB, Pepperberg DR. Reduction of amyloid-beta levels in mouse eye tissues by intra-vitreally delivered neprilysin. Exp Eye Res 2015; 138:134-44. [PMID: 26142956 DOI: 10.1016/j.exer.2015.06.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/18/2015] [Accepted: 06/30/2015] [Indexed: 01/11/2023]
Abstract
Amyloid-beta (Aβ) is a group of aggregation-prone, 38- to 43-amino acid peptides generated in the eye and other organs. Numerous studies suggest that the excessive build-up of low-molecular-weight soluble oligomers of Aβ plays a role in the progression of Alzheimer's disease and other brain degenerative diseases. Recent studies raise the hypothesis that excessive Aβ levels may contribute also to certain retinal degenerative diseases. These findings, together with evidence that a major portion of Aβ is released as monomer into the extracellular space, raise the possibility that a technology enabling the enzymatic break-down of monomeric Aβ in the living eye under physiological conditions could prove useful for research on ocular Aβ physiology and, perhaps ultimately, for therapeutic applications. Neprilysin (NEP), an endopeptidase known to cleave Aβ monomer into inactive products, is a membrane-associated protein. However, sNEP, a recombinant form of the NEP catalytic domain, is soluble in aqueous medium. With the aim of determining the Aβ-cleaving activity of exogenous sNEP in the microenvironment of the intact eye, we analyzed the effect of intra-vitreally delivered sNEP on ocular Aβ levels in mice that exhibit readily measurable, aqueous buffer-extractable Aβ40 and Aβ42, two principal forms of Aβ. Anesthetized 10-month wild-type (C57BL/6J) and 2-3-month 5XFAD transgenic mice received intra-vitreal injections of sNEP (0.004-10 μg) in one eye and were sacrificed at defined post-treatment times (30 min - 12 weeks). Eye tissues (combined lens, vitreous, retina, RPE and choroid) were homogenized in phosphate-buffered saline, and analyzed for Aβ40 and Aβ42 (ELISA) and for total protein (Bradford assay). The fellow, untreated eye of each mouse served as control, and concentrations of Aβ (pmol/g protein) in the treated eye were normalized to that of the untreated control eye. In C57BL/6J mice, as measured at 2 h after sNEP treatment, increasing amounts of injected sNEP yielded progressively greater reductions of Aβ40, ranging from 12% ± 3% (mean ± SEM; n = 3) with 4 ng sNEP to 85% ± 13% (n = 5) with 10 μg sNEP. At 4 ng sNEP the average Aβ40 reduction reached >70% by 24 h following treatment and remained near this level for about 8 weeks. In 5XFAD mice, 10 μg sNEP produced an Aβ40 decrease of 99% ± 1% (n = 4) and a substantial although smaller decrease in Aβ42 (42% ± 36%; n = 4) within 24 h. Electroretinograms (ERGs) were recorded from eyes of C57BL/6J and 5XFAD mice at 9 days following treatment with 4 ng or 10 μg sNEP, conditions that on average led, respectively, to an 82% and 91% Aβ40 reduction in C57BL/6J eyes, an 87% and 92% Aβ40 reduction in 5XFAD eyes, and a 23% and 52% Aβ42 reduction in 5XFAD eyes. In all cases, sNEP-treated eyes exhibited robust ERG responses, consistent with a general tolerance of the posterior eye tissues to the investigated conditions of sNEP treatment. The sNEP-mediated decrease of ocular Aβ levels reported here represents a possible approach for determining effects of Aβ reduction in normally functioning eyes and in models of retinal degenerative disease.
Collapse
Affiliation(s)
- Rajni Parthasarathy
- Lions of Illinois Eye Research Institute, Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - K Martin Chow
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Zahra Derafshi
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | | | - John R Hetling
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - David W Rodgers
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Louis B Hersh
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - David R Pepperberg
- Lions of Illinois Eye Research Institute, Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
23
|
Huyer LD, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj A, Radisic M. Biomaterial based cardiac tissue engineering and its applications. Biomed Mater 2015; 10:034004. [PMID: 25989939 PMCID: PMC4464787 DOI: 10.1088/1748-6041/10/3/034004] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide, necessitating the development of effective treatment strategies. A myocardial infarction involves the blockage of a coronary artery leading to depletion of nutrient and oxygen supply to cardiomyocytes and massive cell death in a region of the myocardium. Cardiac tissue engineering is the growth of functional cardiac tissue in vitro on biomaterial scaffolds for regenerative medicine application. This strategy relies on the optimization of the complex relationship between cell networks and biomaterial properties. In this review, we discuss important biomaterial properties for cardiac tissue engineering applications, such as elasticity, degradation, and induced host response, and their relationship to engineered cardiac cell environments. With these properties in mind, we also emphasize in vitro use of cardiac tissues for high-throughput drug screening and disease modelling.
Collapse
Affiliation(s)
- Locke Davenport Huyer
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Miles Montgomery
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yun Xiao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Genevieve Conant
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network and IBBME, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Vinay R, KusumDevi V. Potential of targeted drug delivery system for the treatment of bone metastasis. Drug Deliv 2014; 23:21-9. [PMID: 24839990 DOI: 10.3109/10717544.2014.913325] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bone metastasis is a devastating complication of cancer that requires an immediate attention. Although our understanding of the metastatic process has improved over the years, yet a number of questions still remain unanswered, and more research is required for complete understanding of the skeletal consequences of metastasis. Furthermore, as no effective treatments are available for some of the most common skeleton disorders such as arthritis, osteoarthritis, osteosarcoma and metastatic bone cancer, there is an urgent need to develop new drugs and drug delivery systems for safe and efficient clinical treatments. Hence this article describes the potential of targeted delivery platforms aimed specifically at bone metastasized tumors. The review gives a brief understanding of the proposed mechanisms of metastasis and focuses primarily on the targeting moieties such as bisphosphonates, which represent the current gold standard in bone metastasis therapies. Special focus has been given to the targeted nanoparticulate systems for treating bone metastasis and its future. Also highlighted are some of the therapeutic targets that can be exploited for designing therapies for bone metastasis. Some of the patented molecules for bone metastasis prevention and treatment have also been discussed. Recently proposed HIFU-CHEM, which utilizes High Intensity Focused ultrasound (HIFU) guided by MRI in combination with temperature-sensitive nanomedicines has also been briefed. The study has been concluded with a focus on the innovations requiring an immediate attention that could improve the treatment modality of bone metastasis.
Collapse
Affiliation(s)
- Raichur Vinay
- a Department of Pharmaceutics , Al-Ameen College of Pharmacy , Bangalore , Karnataka , India
| | - V KusumDevi
- a Department of Pharmaceutics , Al-Ameen College of Pharmacy , Bangalore , Karnataka , India
| |
Collapse
|
25
|
Promsote W, Veeranan-Karmegam R, Ananth S, Shen D, Chan CC, Lambert NA, Ganapathy V, Martin PM. L-2-oxothiazolidine-4-carboxylic acid attenuates oxidative stress and inflammation in retinal pigment epithelium. Mol Vis 2014; 20:73-88. [PMID: 24426777 PMCID: PMC3888500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 01/03/2014] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Oxidant- and inflammation-induced damage to retinal pigment epithelial (RPE) cells is central to the pathogenesis of age-related macular degeneration (AMD). Thus, developing novel strategies to protect these cells is important. We reported previously on the robust antioxidant and therefore cell-protective effects of the cysteine pro-drug L-2-oxothiazolidine-4-carboxylic acid (OTC) in cultured human RPE cells. New reports citing a novel anti-inflammatory role for OTC in addition to the known glutathione-stimulating and antioxidant properties emerged recently; however, this role has not been evaluated in RPE cells or in intact retina. Given the crucial causative roles of oxidative stress and inflammation in AMD pathogenesis, knowing whether OTC might exhibit a similar benefit in this cell and tissue type has high clinical relevance; thus, we evaluated OTC in the present study. METHODS ARPE-19 and primary RPE cells isolated from wild-type, Gpr109a(-/-) , or Slc5a8(-/-) mouse eyes were exposed to TNF-α in the presence or absence of OTC, followed by analysis of IL-6 and Ccl2 expression with real-time quantitative polymerase chain reaction or enzyme-linked immunosorbent assay. Cellular and molecular markers of inflammation and oxidative stress (i.e., IL-1β, TGF-β, ABCG1, ABCA1, reduced glutathione, and dihydroethidium) were evaluated in Ccl2(-/-)/Cx3cr1(-/-) double knockout mice on rd8 background (DKO rd8) treated with OTC (10 mg/ml) in drinking water for a period of 5 months. RESULTS OTC treatment significantly inhibited the expression and secretion of IL-6 and Ccl2 in TNF-α-stimulated ARPE-19 cells. Studies conducted using DKO rd8 animals treated with OTC in drinking water confirmed these findings. Cellular and molecular markers of inflammation were significantly suppressed in the retinas of the OTC-treated DKO rd8 animals. Subsequent in vitro and in vivo studies of the possible mechanism(s) to explain these actions revealed that although OTC is an agonist of the anti-inflammatory G-protein coupled receptor GPR109A and a transportable substrate of the sodium-coupled monocarboxylate transporter SMCT1 (SLC5A8), these properties may play a role but do not explain entirely the anti-inflammatory effects this compound elicits in cultured RPE cells and the intact mouse retina. CONCLUSIONS This study represents, to our knowledge, the first report of the suppressive effects of OTC on inflammation in cultured RPE cells and on inflammation and oxidative stress in the retina in vivo.
Collapse
Affiliation(s)
- Wanwisa Promsote
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA
| | | | - Sudha Ananth
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA
| | - Defen Shen
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Nevin A. Lambert
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA
| | - Vadivel Ganapathy
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA,James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, GA
| | - Pamela M. Martin
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA,Department of Ophthalmology, and the Georgia Regents University, Augusta, GA,James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, GA
| |
Collapse
|
26
|
Critical assessment of implantable drug delivery devices in glaucoma management. JOURNAL OF DRUG DELIVERY 2013; 2013:895013. [PMID: 24066234 PMCID: PMC3770064 DOI: 10.1155/2013/895013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/26/2013] [Indexed: 12/21/2022]
Abstract
Glaucoma is a group of heterogeneous disorders involving progressive optic neuropathy that can culminate into visual impairment and irreversible blindness. Effective therapeutic interventions must address underlying vulnerability of retinal ganglion cells (RGCs) to degeneration in conjunction with correcting other associated risk factors (such as elevated intraocular pressure). However, realization of therapeutic outcomes is heavily dependent on suitable delivery system that can overcome myriads of anatomical and physiological barriers to intraocular drug delivery. Development of clinically viable sustained release systems in glaucoma is a widely recognized unmet need. In this regard, implantable delivery systems may relieve the burden of chronic drug administration while potentially ensuring high intraocular drug bioavailability. Presently there are no FDA-approved implantable drug delivery devices for glaucoma even though there are several ongoing clinical studies. The paper critically assessed the prospects of polymeric implantable delivery systems in glaucoma while identifying factors that can dictate (a) patient tolerability and acceptance, (b) drug stability and drug release profiles, (c) therapeutic efficacy, and (d) toxicity and biocompatibility. The information gathered could be useful in future research and development efforts on implantable delivery systems in glaucoma.
Collapse
|
27
|
Combined treatment with low concentrations of decitabine and SAHA causes cell death in leukemic cell lines but not in normal peripheral blood lymphocytes. BIOMED RESEARCH INTERNATIONAL 2013; 2013:659254. [PMID: 24000324 PMCID: PMC3755446 DOI: 10.1155/2013/659254] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/28/2013] [Accepted: 07/13/2013] [Indexed: 12/15/2022]
Abstract
Epigenetic therapy reverting aberrant acetylation or methylation offers the possibility to target preferentially tumor cells and to preserve normal cells. Combination epigenetic therapy may further improve the effect of individual drugs. We investigated combined action of demethylating agent decitabine and histone deacetylase inhibitor SAHA (Vorinostat) on different leukemic cell lines in comparison with peripheral blood lymphocytes. Large decrease of viability, as well as huge p21WAF1 induction, reactive oxygen species formation, and apoptotic features due to combined decitabine and SAHA action were detected in leukemic cell lines irrespective of their p53 status, while essentially no effect was observed in response to the combined drug action in normal peripheral blood lymphocytes of healthy donors. p53-dependent apoptotic pathway was demonstrated to participate in the wtp53 CML-T1 leukemic cell line response, while significant influence of reactive oxygen species on viability decrease has been detected in p53-null HL-60 cell line.
Collapse
|
28
|
Shmueli RB, Ohnaka M, Miki A, Pandey NB, Lima e Silva R, Koskimaki JE, Kim J, Popel AS, Campochiaro PA, Green JJ. Long-term suppression of ocular neovascularization by intraocular injection of biodegradable polymeric particles containing a serpin-derived peptide. Biomaterials 2013; 34:7544-51. [PMID: 23849876 DOI: 10.1016/j.biomaterials.2013.06.044] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/23/2013] [Indexed: 12/27/2022]
Abstract
Aberrant angiogenesis can cause or contribute to a number of diseases such as neovascular age-related macular degeneration (NVAMD). While current NVAMD treatments target angiogenesis, these treatments are not effective for all patients and also require frequent intravitreal injections. New agents and delivery systems to treat NVAMD could be beneficial to many patients. We have recently developed a serpin-derived peptide as an anti-angiogenic agent. Here, this peptide is investigated for activity in human retinal endothelial cells in vitro and for reducing angiogenesis in a laser-induced choroidal neovascularization mouse model of NVAMD in vivo. While frequent intravitreal injections can be tolerated clinically, reducing the number of injections can improve patient compliance, safety, and outcomes. To achieve this goal, and to maximize the in vivo activity of injected peptide, we have developed biodegradable polymers and controlled release particle formulations to extend anti-angiogenic therapy. To create these devices, the anionic peptides are first self-assembled into nanoparticles using a biodegradable cationic polymer and then as a second step, these nanoparticles are encapsulated into biodegradable poly(lactic-co-glycolic acid) (PLGA) microparticles. In situ, these particles show approximately zero-order, linear release of the anionic peptide over 200 days. These particles are made of safe, hydrolytically degradable polymers and have low endotoxin. Long-term in vivo experiments in the laser-induced neovascularization model for NVAMD show that these peptide-releasing particles decrease angiogenesis for at least fourteen weeks in vivo following a single particle dose and therefore are a promising treatment strategy for NVAMD.
Collapse
Affiliation(s)
- Ron B Shmueli
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ligand-functionalized nanoparticles target endothelial cells in retinal capillaries after systemic application. Proc Natl Acad Sci U S A 2013; 110:6115-20. [PMID: 23530216 DOI: 10.1073/pnas.1220281110] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To date, diseases affecting vascular structures in the posterior eye are mostly treated by laser photocoagulation and multiple intraocular injections, procedures that destroy healthy tissue and can cause vision-threatening complications. To overcome these drawbacks, we investigate the feasibility of receptor-mediated nanoparticle targeting to capillary endothelial cells in the retina after i.v. application. Cell-binding studies using microvascular endothelial cells showed receptor-specific binding and cellular uptake of cyclo(RGDfC)-modified quantum dots via the αvβ3 integrin receptor. Conversely, Mueller cells and astrocytes, representing off-target cells located in the retina, revealed only negligible interaction with nanoparticles. In vivo experiments, using nude mice as the model organism, demonstrated a strong binding of the ligand-modified quantum dots in the choriocapillaris and intraretinal capillaries upon i.v. injection and 1-h circulation time. Nontargeted nanoparticles, in contrast, did not accumulate to a significant amount in the target tissue. The presented strategy of targeting integrin receptors in the retina could be of utmost value for future intervention in pathologies of the posterior eye, which are to date only accessible with difficulty.
Collapse
|