1
|
Liang D, Walker J, Schwendeman PS, Chandrashekar A, Ackermann R, Olsen KF, Beck-Broichsitter M, Schwendeman SP. Effect of PLGA raw materials on in vitro and in vivo performance of drug-loaded microspheres. Drug Deliv Transl Res 2025; 15:185-202. [PMID: 38643259 DOI: 10.1007/s13346-024-01577-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 04/22/2024]
Abstract
Poly(lactide-co-glycolide) and poly(lactic-co-glycolic acids) (PLGAs) play a critical role in the development of commercial long-acting injectable microsphere formulations. However, very little information is available describing the impact of PLGA manufacturer and monomer distribution along the polymer chain (e.g., glycolic blockiness (Rc) and average lactic block length (LL)) on the degradation and release behavior of PLGA drug carriers in vitro and in vivo. Here, we compared the in vitro and in vivo performance of (a) four leuprolide-loaded microsphere formulations prepared from similar low-molecular-weight acid-capped PLGAs (10-14 kD, i.e., Expansorb® DLG 75-2A, Purasorb® PDLG 7502A, Resomer® RG 752H and Wako® 7515) and (b) two triamcinolone acetonide-loaded (Tr-A) microsphere formulations from similar medium-molecular-weight ester-capped PLGAs (i.e., Expansorb® DLG 75-4E and Resomer® RG 753S). Lupron Depot® and Zilretta® were used as reference commercial products. The six 75/25 PLGAs displayed block lengths that were either above or below values expected from a random copolymer. Drug release and polymer degradation were monitored simultaneously in vitro and in vivo using a cage implant system. The four leuprolide-loaded formulations showed similar release and degradation patterns with some notable differences between each other. Microspheres from the Expansorb® polymer displayed lower LL and higher Rc relative to the other 3 PLGA 75/25 microspheres, and likewise exhibited distinct peptide release and degradation behavior compared to the other 3 formulations. For each formulation, leuprolide release was erosion-controlled up to about 30% release after the initial burst followed by a faster than erosion release phase. In vitro release was similar as that in vivo over the first phase but notably different from the latter release phase, particularly for the most blocky Expansorb® formulation. The Purasorb® and Wako® formulations displayed highly similar performance in release, degradation, and erosion analysis. By contrast, the two ester-capped Expansorb® DLG 75-4E and Resomer® RG 753S used to prepare Tr-A microspheres shared essentially identical LL and higher Rc and behaved similarly although the Expansorb® degraded and released the steroid faster in vivo, suggestive of other factors responsible (e.g., residual monomer). The in vivo release performance for both drugs from the six microsphere formulations was similar to that of the commercial reference products. In summary, this work details information on comparing the similarities and differences in in vitro and in vivo performance of drug-loaded microspheres as a function of manufacturing and microstructural variables of different types of PLGA raw materials utilized and could, therefore, be meaningful in guiding the source control during development and manufacturing of PLGA microsphere-based drug products. Future work will expand the analysis to include a broader range of LL and higher Rc, and add additional important formulation metrics (e.g., thermal analysis, and residual monomer, moisture, and organic solvent levels).
Collapse
Affiliation(s)
- Desheng Liang
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Jennifer Walker
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Peter S Schwendeman
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Ave., Ann Arbor, MI, 48109, USA
| | - Aishwarya Chandrashekar
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Rose Ackermann
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Karl F Olsen
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA
| | - Moritz Beck-Broichsitter
- MilliporeSigma a Business of Merck Life Science KGaA, Frankfurter Strasse 250, D-64293, Darmstadt, Germany
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
Almasri RM, Ladouceur F, Mawad D, Esrafilzadeh D, Firth J, Lehmann T, Poole-Warren LA, Lovell NH, Al Abed A. Emerging trends in the development of flexible optrode arrays for electrophysiology. APL Bioeng 2023; 7:031503. [PMID: 37692375 PMCID: PMC10491464 DOI: 10.1063/5.0153753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Optical-electrode (optrode) arrays use light to modulate excitable biological tissues and/or transduce bioelectrical signals into the optical domain. Light offers several advantages over electrical wiring, including the ability to encode multiple data channels within a single beam. This approach is at the forefront of innovation aimed at increasing spatial resolution and channel count in multichannel electrophysiology systems. This review presents an overview of devices and material systems that utilize light for electrophysiology recording and stimulation. The work focuses on the current and emerging methods and their applications, and provides a detailed discussion of the design and fabrication of flexible arrayed devices. Optrode arrays feature components non-existent in conventional multi-electrode arrays, such as waveguides, optical circuitry, light-emitting diodes, and optoelectronic and light-sensitive functional materials, packaged in planar, penetrating, or endoscopic forms. Often these are combined with dielectric and conductive structures and, less frequently, with multi-functional sensors. While creating flexible optrode arrays is feasible and necessary to minimize tissue-device mechanical mismatch, key factors must be considered for regulatory approval and clinical use. These include the biocompatibility of optical and photonic components. Additionally, material selection should match the operating wavelength of the specific electrophysiology application, minimizing light scattering and optical losses under physiologically induced stresses and strains. Flexible and soft variants of traditionally rigid photonic circuitry for passive optical multiplexing should be developed to advance the field. We evaluate fabrication techniques against these requirements. We foresee a future whereby established telecommunications techniques are engineered into flexible optrode arrays to enable unprecedented large-scale high-resolution electrophysiology systems.
Collapse
Affiliation(s)
- Reem M. Almasri
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | | | - Damia Mawad
- School of Materials Science and Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Josiah Firth
- Australian National Fabrication Facility, UNSW, Sydney, NSW 2052, Australia
| | - Torsten Lehmann
- School of Electrical Engineering and Telecommunications, UNSW, Sydney, NSW 2052, Australia
| | | | | | - Amr Al Abed
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| |
Collapse
|
3
|
Hong JKY, Schutzman R, Olsen K, Chandrashekar A, Schwendeman SP. Mapping in vivo microclimate pH distribution in exenatide-encapsulated PLGA microspheres. J Control Release 2022; 352:438-449. [PMID: 36030989 DOI: 10.1016/j.jconrel.2022.08.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/30/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022]
Abstract
The pH inside the aqueous pores of poly(lactic-co-glycolic acid) (PLGA) microspheres, often termed microclimate pH (μpH), has been widely evaluated in vitro and shown to commonly be deleterious to pH-labile encapsulated drug molecules. However, whether the in vitro μpH is representative of the actual in vivo values has long been remained a largely unresolved issue. Herein we quantitatively mapped, for the first time, the in vivo μpH distribution kinetics inside degrading PLGA microspheres by combining two previously validated techniques, a cage implant system and confocal laser scanning microscopy. PLGA (50/50, Mw = 24-38 kDa, acid-end capped and ester-capped) microsphere formulations with and without encapsulating exenatide, a pH-labile peptide that is known to be unstable when pH > 4.5, were administered to rats subcutaneously via cage implants for up to 6 weeks. The results were compared with two different in vitro conditions. Strikingly, the in vivo μpH developed similarly to the low microsphere concentration in vitro condition with 1-μm nylon bags but very different from conventional high microsphere concentration sample-and-separate conditions. Improved maintenance of stable external pH in the release media for the former condition may have been one important factor. Stability of exenatide remaining inside microspheres was evaluated by mass spectrometry and found that it was steadily degraded primarily via pH-dependent acylation with a trend that slightly paralleled the changes in μpH. This methodology may be useful to elucidate pH-triggered instability of PLGA encapsulated drugs in vivo and for improving in vivo-predictive in vitro conditions for assessing general PLGA microsphere performance.
Collapse
Affiliation(s)
- Justin K Y Hong
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Richard Schutzman
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Karl Olsen
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Aishwarya Chandrashekar
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA.
| |
Collapse
|
4
|
Gupta R, Chen Y, Sarkar M, Xie H. Surfactant Mediated Accelerated and Discriminatory In Vitro Drug Release Method for PLGA Nanoparticles of Poorly Water-Soluble Drug. Pharmaceuticals (Basel) 2022; 15:ph15121489. [PMID: 36558940 PMCID: PMC9787738 DOI: 10.3390/ph15121489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/20/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
In vitro drug release testing is an important quality control tool for formulation development. However, the literature has evidence that poly-lactide-co-glycolide (PLGA)-based formulations show a slower in vitro drug release than a real in vivo drug release. Much longer in vitro drug release profiles may not be reflective of real in vivo performances and may significantly affect the timeline for a formulation development. The objective of this study was to develop a surfactant mediated accelerated in vitro drug release method for the PLGA nanoparticles (NPs) of a novel chemotherapeutic agent AC1LPSZG, a model drug with a poor solubility. The Sotax USP apparatus 4 was used to test in vitro drug release in a phosphate buffer with a pH value of 6.8. The sink conditions were improved using surfactants in the order of sodium lauryl sulfate (SLS) < Tween 80 < cetyltrimethylammonium bromide (CTAB). The dissolution efficiency (DE) and area under the dissolution curve (AUC) were increased three-fold when increasing the CTAB concentration in the phosphate buffer (pH 6.8). Similar Weibull release kinetics and good linear correlations (R2~0.99) indicated a good correlation between the real-time in vitro release profile in the phosphate buffer (pH 6.8) and accelerated release profiles in the optimized medium. This newly developed accelerated and discriminatory in vitro test can be used as a quality control tool to identify critical formulation and process parameters to ensure a batch-to-batch uniformity. It may also serve as a surrogate for bioequivalence studies if a predictive in vitro in vivo correlation (IVIVC) is obtained. The results of this study are limited to AC1LPSZG NPs, but a similar consideration can be extended to other PLGA-based NPs of drugs with similar properties and solubility profiles.
Collapse
|
5
|
Richard AS, Verma RS. Bioactive nano yarns as surgical sutures for wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112334. [PMID: 34474885 DOI: 10.1016/j.msec.2021.112334] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 11/19/2022]
Abstract
Surgical sutures are the most widely used medical device in any surgical procedure worldwide. In this study, modified electrospinning technique has been used as manufacturing technique to produce nanofiber bundles twisted simultaneously to obtain nanofiber yarns. Taking the advantage of nanofiber yarns in terms of biomimetic structure, mechanical strength and handling properties, the material is chosen. Curcumin, a natural compound is incorporated to the nanofiber yarns by blend electrospinning technique for its anti-inflammatory, antibiotic and wound healing properties. The synthesized nanofiber yarns were characterized by various characterization techniques such as XRD, FTIR, SEM, Tensile testing, stem cell interaction, hemocompatibility, bacterial response, drug release profiling and in vivo studies. Curcumin loaded nanofiber yarns demonstrated sustained release with improved antibacterial, antiplatelet, cell migration and stem cell interaction in vitro. The results from skin inflammation animal model revealed that curcumin laden nanofiber yarn suture manifested reduced inflammation and cellularity. The three dimensional structure, adequate mechanical strength and biological properties of the nanofiber yarn provide naive environment for wound healing with the balanced degradation of suture material in rat model.
Collapse
Affiliation(s)
- Arthi Sunil Richard
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Rama Shankar Verma
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
6
|
B. S S, Gopalakrishnan-Prema V, Raju G, Mathew SE, Katiyar N, Menon D, Shankarappa SA. Anisotropic microparticles for differential drug release in nerve block anesthesia. RSC Adv 2021; 11:4623-4630. [PMID: 35424395 PMCID: PMC8694510 DOI: 10.1039/d0ra08386k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/18/2021] [Indexed: 12/17/2022] Open
Abstract
Microparticle shape, as a tunable design parameter, holds much promise for controlling drug-release kinetics from polymeric microparticulate systems. In this study we hypothesized that the intensity and duration of a local nerve block can be controlled by administration of bupivacaine-loaded stretch-induced anisotropic poly(lactic-co-glycolic acid) microparticles (MPs). MPs of size 27.3 ± 8.5 μm were synthesized by single emulsion method and subjected to controlled stretching force. The aspect ratio of the anisotropic–bupivacaine MPs was quantified, and bupivacaine release was measured in vitro. The anisotropic MPs were administered as local nerve block injections in rats, and the intensity and duration of local anesthesia was measured. Bupivacaine-loaded anisotropic MPs used in this study were ellipsoid in shape and exhibited increased surface pores in comparison to spherical MPs. Anisotropic MPs exhibited a higher rate of bupivacaine release in vitro, and showed significantly (P < 0.05) stronger sensory nerve blocking as compared to spherical bupivacaine MPs, even though the duration of the nerve block remained similar. This study demonstrates the utility of stretch-induced anisotropic MPs in controlling drug release profiles from polymeric MPs, under both in vitro and in vivo conditions. We show that shape, as a tunable design parameter, could play an important role in engineering drug-delivery systems. Stretch-induced anisotropy in bupivacaine-loaded PLGA micro particles (BMPs) induced stronger nerve blocks compared to spherical particles.![]()
Collapse
Affiliation(s)
- Shivakumar B. S
- Center for Nanosciences & Molecular Medicine
- Amrita Vishwa Vidyapeetham
- Kochi 682041
- India
| | | | - Gayathri Raju
- Center for Nanosciences & Molecular Medicine
- Amrita Vishwa Vidyapeetham
- Kochi 682041
- India
| | - Sumi E. Mathew
- Center for Nanosciences & Molecular Medicine
- Amrita Vishwa Vidyapeetham
- Kochi 682041
- India
| | - Neeraj Katiyar
- Center for Nanosciences & Molecular Medicine
- Amrita Vishwa Vidyapeetham
- Kochi 682041
- India
| | - Deepthy Menon
- Center for Nanosciences & Molecular Medicine
- Amrita Vishwa Vidyapeetham
- Kochi 682041
- India
| | | |
Collapse
|
7
|
Farto-Vaamonde X, Auriemma G, Aquino RP, Concheiro A, Alvarez-Lorenzo C. Post-manufacture loading of filaments and 3D printed PLA scaffolds with prednisolone and dexamethasone for tissue regeneration applications. Eur J Pharm Biopharm 2019; 141:100-110. [PMID: 31112767 DOI: 10.1016/j.ejpb.2019.05.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
Abstract
Strategies to load prednisolone or dexamethasone in preformed poly(L-lactic acid) (PLA) filaments and 3D printed scaffolds were explored as a way of personalizing the drug, the dose and the release profile for regenerative medicine purposes. Instead of starting from a PLA filament preloaded with a given content of drug, we explored two more versatile strategies. The first one involved the soaking of PLA filaments into a drug solution prepared in a solvent that reversibly swelled PLA; during 3D printing the melting of PLA contributed to the efficient integration (encapsulation) of the drug inside the printed strand. The second strategy consisted in first printing the 3D PLA scaffolds followed by soaking in a suitable drug solution in order to exploit the higher specific surface of the printed strands compared to the filament. Sustained release profiles were recorded when either prednisolone or dexamethasone were loaded in preformed PLA filaments, while rapid release was recorded for 3D PLA scaffolds loaded after printing. The combination of the two proposed methods reported here opened the possibility of creating concentration gradients of different drugs in the same scaffold exhibiting distinct release patterns. Namely, the strand core contained an active ingredient to be slowly released, while the surface was covered with other active ingredient that could be rapidly delivered. The feasibility of this approach was confirmed through dual loading of dexamethasone in the filament and of prednisolone on the preformed scaffold. Drug-loaded scaffolds were characterized in terms of printability, structural characteristics (DSC, XRD), mechanical properties, biodegradation, and ability to promote cell attachment and proliferation. Finally, anti-inflammatory response and osteoinductive properties were verified in cell cultures.
Collapse
Affiliation(s)
- Xián Farto-Vaamonde
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano (SA), Italy
| | - Rita Patrizia Aquino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano (SA), Italy
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
8
|
Ruan S, Gu Y, Liu B, Gao H, Hu X, Hao H, Jin L, Cai T. Long-Acting Release Microspheres Containing Novel GLP-1 Analog as an Antidiabetic System. Mol Pharm 2018; 15:2857-2869. [PMID: 29763559 DOI: 10.1021/acs.molpharmaceut.8b00344] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) has recently received significant attention as an efficacious way to treat diabetes mellitus. However, the short half-life of the peptide limits its clinical application in diabetes. In our previous study, a novel GLP-1 analog (PGLP-1) with a longer half-life was synthesized and evaluated. Herein, we prepared the PGLP-1-loaded poly(d,l-lactide- co-glycolide) microspheres to achieve long-term effects on blood glucose control. The incorporation of zinc ion into the formulation can effectively decrease the initial burst release, and a uniform drug distribution was obtained, in contrast to native PGLP-1 encapsulated microspheres. We demonstrated that the solubility of the drug encapsulated in microspheres played an important role in in vitro release behavior and drug distribution inside the microspheres. The Zn-PGLP-1 microspheres had a prominent acute glucose reduction effect in the healthy mice. A hypoglycemic effect was observed in the streptozotocin (STZ) induced diabetic mice through a 6-week treatment of Zn-PGLP-1-loaded microspheres. Meanwhile, the administration of Zn-PGLP-1 microspheres led to the β-cell protection and stimulation of insulin secretion. The novel GLP-1 analog-loaded sustained microspheres may greatly improve patient compliance along with a desirable safety feature.
Collapse
|
9
|
Tatara AM, Kontoyiannis DP, Mikos AG. Drug delivery and tissue engineering to promote wound healing in the immunocompromised host: Current challenges and future directions. Adv Drug Deliv Rev 2018; 129:319-329. [PMID: 29221962 PMCID: PMC5988908 DOI: 10.1016/j.addr.2017.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/23/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022]
Abstract
As regenerative medicine matures as a field, more promising technologies are being translated from the benchtop to the clinic. However, many of these strategies are designed with otherwise healthy hosts in mind and validated in animal models without other co-morbidities. In reality, many of the patient populations benefiting from drug delivery and tissue engineering-based devices to enhance wound healing also have significant underlying immunodeficiency. Specifically, patients suffering from diabetes, malignancy, human immunodeficiency virus, post-organ transplantation, and other compromised states have significant pleotropic immune defects that affect wound healing. In this work, we review the role of different immune cells in the regenerative process, highlight the effect of several common immunocompromised states on wound healing, and discuss different drug delivery strategies for overcoming immunodeficiencies.
Collapse
Affiliation(s)
- Alexander M Tatara
- Medical Scientist Training Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, United States; Department of Bioengineering, Rice University, Houston, TX, United States.
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States.
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, United States.
| |
Collapse
|
10
|
Galla TJ, Vedecnik SV, Halbgewachs J, Steinmann S, Friedrich C, Stark GB. Fibrin/Schwann Cell Matrix in Poly-Epsilon-Caprolactone Conduits Enhances Guided Nerve Regeneration. Int J Artif Organs 2018; 27:127-36. [PMID: 15068007 DOI: 10.1177/039139880402700208] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The goal of this study was to investigate if a three dimensional matrix, loaded homogeneously with Schwann cells and the neurotrophic factor LIF (leukemia inhibitory factor), enhances regeneration in a biodegradable nerve guidance channel as compared to non-structured cell suspensions. Therefore a 10 mm nerve gap in the buccal branch of the rat's facial nerve was bridged with tubular PCL (poly-epsilon-caprolactone) conduits filled with no matrix, Schwann cells, the three dimensional fibrin/Schwann cell matrix or the fibrin/Schwann cell matrix added with LIF. Four weeks after the nerve defects were bridged histological and morphometric analyses of the implants were performed. In conclusion, the three dimensional fibrin/Schwann cells matrix enhanced the quantity and the quality of peripheral nerve regeneration through PCL conduits. The application of LIF prevented hyperneurotization. Therefore, tissue engineered fibrin/Schwann cells matrices are new invented biocompatible and biodegradable devices for enhancing peripheral nerve regeneration as compared to non-structured cell suspensions without neurotrophic factors.
Collapse
Affiliation(s)
- T J Galla
- Department of Hand and Plastic Surgery, ValleyTEC, Albert-Ludwigs-University, Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
11
|
Doty AC, Weinstein DG, Hirota K, Olsen KF, Ackermann R, Wang Y, Choi S, Schwendeman SP. Mechanisms of in vivo release of triamcinolone acetonide from PLGA microspheres. J Control Release 2017; 256:19-25. [DOI: 10.1016/j.jconrel.2017.03.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/03/2017] [Accepted: 03/18/2017] [Indexed: 02/05/2023]
|
12
|
Hirota K, Doty AC, Ackermann R, Zhou J, Olsen KF, Feng MR, Wang Y, Choi S, Qu W, Schwendeman AS, Schwendeman SP. Characterizing release mechanisms of leuprolide acetate-loaded PLGA microspheres for IVIVC development I: In vitro evaluation. J Control Release 2016; 244:302-313. [DOI: 10.1016/j.jconrel.2016.08.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/07/2016] [Accepted: 08/21/2016] [Indexed: 11/28/2022]
|
13
|
Validation of a cage implant system for assessing in vivo performance of long-acting release microspheres. Biomaterials 2016; 109:88-96. [DOI: 10.1016/j.biomaterials.2016.07.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 07/28/2016] [Accepted: 07/31/2016] [Indexed: 12/11/2022]
|
14
|
Doty AC, Zhang Y, Weinstein DG, Wang Y, Choi S, Qu W, Mittal S, Schwendeman SP. Mechanistic analysis of triamcinolone acetonide release from PLGA microspheres as a function of varying in vitro release conditions. Eur J Pharm Biopharm 2016; 113:24-33. [PMID: 27865933 DOI: 10.1016/j.ejpb.2016.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 10/29/2016] [Accepted: 11/04/2016] [Indexed: 10/20/2022]
Abstract
In vitro tests for controlled release PLGA microspheres in their current state often do not accurately predict in vivo performance of these products during formulation development. Here, we introduce a new mechanistic and multi-phase approach to more clearly understand in vitro-in vivo relationships, and describe the first "in vitro phase" with the model drug, triamcinolone acetonide (Tr-A). Two microsphere formulations encapsulating Tr-A were prepared from PLGAs of different molecular weights and end-capping (18kDa acid-capped and 54kDa ester-capped). In vitro release kinetics and the evidence for controlling mechanisms (i.e., erosion, diffusion, and water-mediated processes) were studied in four release media: PBST pH 7.4 (standard condition), PBST pH 6.5, PBS+1.0% triethyl citrate (TC), and HBST pH 7.4. The release mechanism in PBST was primarily polymer erosion-controlled as indicated by the similarity of release and mass loss kinetics. Release from the low MW PLGA was accelerated at low pH due to increased rate of hydrolysis and in the presence of the plasticizer TC due to slightly increased hydrolysis and much higher diffusion in the polymer matrix. TC also increased release from the high MW PLGA due to increased hydrolysis, erosion, and diffusion. This work demonstrates how in vitro conditions can be manipulated to change not only rates of drug release from PLGA microspheres but also the mechanism(s) by which release occurs. Follow-on studies in the next phases of this approach will utilize these results to compare the mechanistic data of the Tr-A/PLGA microsphere formulations developed here after recovery of microspheres in vivo. This new approach based on measuring mechanistic indicators of release in vitro and in vivo has the potential to design better, more predictive in vitro release tests for these formulations and potentially lead to mechanism-based in vitro-in vivo correlations.
Collapse
Affiliation(s)
- Amy C Doty
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, United States; Discovery Pharmaceutical Sciences, Pharmaceutical Sciences & Clinical Supply, Merck Sharp and Dohme Corp, 33 Avenue Louis Pasteur, Boston, MA 02115, United States(1)
| | - Ying Zhang
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, United States; 3M Critical & Chronic Care Solutions Division, 3M Center, St. Paul, MN 55144, United States(1)
| | - David G Weinstein
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, United States
| | - Yan Wang
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Generic Drugs, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Stephanie Choi
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Generic Drugs, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Wen Qu
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Generic Drugs, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Sachin Mittal
- Sterile Formulation Sciences, Pharmaceutical Sciences & Clinical Supply, Merck Sharp and Dohme Corp, 2000 Galloping Hill Road, Kenilworth, NJ 07033, United States
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
15
|
Ranne T, Tirri T, Yli-Urpo A, Närhi T, Laine V, Rich J, Seppälä J, Aho A. In Vivo Behavior of Poly(∈-Caprolactone-co-DL-Lactide)/Bioactive Glass Composites in Rat Subcutaneous Tissue. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911507078270] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this study, tissue reactions and possible toxicological responses of two different bioactive and degradable composite materials consisting of poly( ∈-caprolactone-co-DL-lactide) and bioactive glass (S53P4) granules were evaluated. The chosen materials were implanted subcutaneously in the back of rats for six months. The glass granules retained their bioactivity within the polymer matrices. A fibrous capsule formed around all tested materials and around the materials containing bioactive glass the fibrous capsules appeared to be thicker. Tissue growth into these materials was observed during the healing period while no growth was noticed into the plain polymer matrices. No adverse reactions were seen with any of the evaluated materials.
Collapse
Affiliation(s)
- T. Ranne
- Department of Prosthetic Dentistry and Biomaterials Science
| | - T. Tirri
- Department of Prosthetic Dentistry and Biomaterials Science,
| | - A. Yli-Urpo
- Department of Prosthetic Dentistry and Biomaterials Science
| | - T.O. Närhi
- Department of Prosthetic Dentistry and Biomaterials Science
| | - V.J.O. Laine
- Department of Pathology, University of Turku, Turku 20520, Finland
| | - J. Rich
- Department of Chemical Technology, Helsinki University of Technology, Espoo 02150, Finland
| | - J. Seppälä
- Department of Chemical Technology, Helsinki University of Technology, Espoo 02150, Finland
| | - A. Aho
- Department of Surgery, University of Turku, Turku 20520, Finland
| |
Collapse
|
16
|
Xu Y, Kim CS, Saylor DM, Koo D. Polymer degradation and drug delivery in PLGA-based drug-polymer applications: A review of experiments and theories. J Biomed Mater Res B Appl Biomater 2016; 105:1692-1716. [PMID: 27098357 DOI: 10.1002/jbm.b.33648] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/25/2016] [Accepted: 02/12/2016] [Indexed: 01/03/2023]
Abstract
Poly (lactic-co-glycolic acid) (PLGA) copolymers have been broadly used in controlled drug release applications. Because these polymers are biodegradable, they provide an attractive option for drug delivery vehicles. There are a variety of material, processing, and physiological factors that impact the degradation rates of PLGA polymers and concurrent drug release kinetics. This work is intended to provide a comprehensive and collective review of the physicochemical and physiological factors that dictate the degradation behavior of PLGA polymers and drug release from contemporary PLGA-based drug-polymer products. In conjunction with the existing experimental results, analytical and numerical theories developed to predict drug release from PLGA-based polymers are summarized and correlated with the experimental observations. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1692-1716, 2017.
Collapse
Affiliation(s)
- Yihan Xu
- Materials Science and Engineering Department, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53211
| | - Chang-Soo Kim
- Materials Science and Engineering Department, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53211
| | - David M Saylor
- Division of Biology, Chemistry, and Materials Science, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, 20993
| | - Donghun Koo
- Materials Science R&D, MilliporeSigma, Milwaukee, Wisconsin, 53209
| |
Collapse
|
17
|
Lapointe S, Zhim F, Sidéris L, Drolet P, Célestin-Noël S, Dubé P. Effect of chemotherapy and heat on biomechanical properties of absorbable sutures. J Surg Res 2015; 200:59-65. [PMID: 26505660 DOI: 10.1016/j.jss.2015.06.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 05/26/2015] [Accepted: 06/11/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND The quality of tissue repairs depends on tissue integrity, surgical technique, and material properties of the sutures used. Currently, there is no clear consensus on which is the best suture to use during cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy. The aim of this study was to evaluate the impact of heat and chemotherapy on sutures' biomechanical properties. METHODS Six different 3.0 absorbable sutures (Biosyn, Dexon II, Maxon, Monocryl, PDS II, and Vicryl Plus) were tested. All suture strands were incubated for a 24-h period in saline, mitomycin-c, and oxaliplatin at 37 and 45°C. Suture loops were then loaded to failure using a servohydraulic testing machine. Data for tensile breaking force (TBF) and elongation rate were collected for all samples. RESULTS Under basal condition, Maxon was the strongest of all sutures with a TBF of 59.6 ± 4.3 N (P < 0.01), and no significant difference in TBF was observed between other sutures. Heat alone had no impact on sutures' biomechanical parameters. Exposition to mitomycin-c at 45°C did not significantly affect sutures' basal tensile properties, with Maxon remaining the strongest suture. When incubated in oxaliplatin at 45°C, the six suture types had a similar TBF. In all experimental conditions, multifilament sutures had a significantly lower elongation rate than monofilament sutures, and no correlations were demonstrated between elongation rate and the TBF of sutures. CONCLUSIONS This study showed that exposition to heated chemotherapy did not significantly affect absorbable sutures biomechanical properties.
Collapse
Affiliation(s)
- Stéphanie Lapointe
- Division of General Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Fouad Zhim
- Department of Biomedical Engineering, École Polytechnique de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Lucas Sidéris
- Division of General Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Drolet
- Department of Anesthesiology, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Syndie Célestin-Noël
- Division of General Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Dubé
- Division of General Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
18
|
Wittchow E, Adden N, Riedmüller J, Savard C, Waksman R, Braune M. Bioresorbable drug-eluting magnesium-alloy scaffold: design and feasibility in a porcine coronary model. EUROINTERVENTION 2013; 8:1441-50. [PMID: 23680959 DOI: 10.4244/eijv8i12a218] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIMS Among three versions of bioresorbable magnesium scaffolds featuring different paclitaxel-elution kinetics, we determined the best-performing scaffold and compared it with established, paclitaxel-eluting, permanent stents TAXUS Liberté and eucaTAX. METHODS AND RESULTS Drug-elution kinetics in magnesium scaffolds were modulated by varying the composition of their bioresorbable poly(lactide-co-glycolide) coating loaded with paclitaxel. A 50:50 ratio of lactide to glycolide, or an 85:15 ratio and either high- or low-molecular-weight polymer was applied in the "50/50", "85/15H", and "85/15L" scaffolds, respectively. Seventy-three magnesium scaffolds (25 50/50, 24 85/15H, 24 85/15L) and 36 control stents (18 TAXUS Liberté, 18 eucaTAX) were implanted in coronary arteries of 50 Yucatan mini-pigs. Angiography, histomorphometry, and histopathology data were acquired at 28, 90 and 180 days. The best-performing magnesium scaffold, 85/15H, was equivalent to TAXUS Liberté and superior to eucaTAX regarding late luminal loss, intimal area, fibrin score, and endothelialisation. Intimal inflammation score was higher in 85/15H than in the control stents at 28 days, but this effect disappeared at later time points. CONCLUSIONS By selecting suitable paclitaxel-elution kinetics, it was feasible to develop a bioresorbable magnesium scaffold whose efficacy and healing characteristics in a porcine coronary model are comparable with those of established paclitaxel-eluting permanent metallic stents.
Collapse
|
19
|
Zhu C, Kustra SR, Bettinger CJ. Photocrosslinkable biodegradable elastomers based on cinnamate-functionalized polyesters. Acta Biomater 2013; 9:7362-70. [PMID: 23567941 DOI: 10.1016/j.actbio.2013.03.041] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/18/2013] [Accepted: 03/29/2013] [Indexed: 10/27/2022]
Abstract
Synthetic biodegradable elastomers are an emerging class of materials that play a critical role in supporting innovations in bioabsorbable medical implants. This paper describes the synthesis and characterization of poly(glycerol-co-sebacate)-cinnamate (PGS-CinA), a biodegradable elastomer based on hyperbranched polyesters derivatized with pendant cinnamate groups. PGS-CinA can be prepared via photodimerization in the absence of photoinitiators using monomers that are found in common foods. The resulting network exhibits a Young's modulus of 50.5-152.1kPa and a projected in vitro degradation half-life time between 90 and 140days. PGS-CinA elastomers are intrinsically cell-adherent and support rapid proliferation of fibroblasts. Spreading and proliferation of fibroblasts are loosely governed by the substrate stiffness within the range of Young's moduli in PGS-CinA networks that were prepared. The thermo-mechanical properties, biodegradability and intrinsic support of cell attachment and proliferation suggest that PGS-CinA networks are broadly applicable for use in next generation bioabsorable materials including temporary medical devices and scaffolds for soft tissue engineering.
Collapse
|
20
|
Hu L, Zhang H, Song W. An overview of preparation and evaluation sustained-release injectable microspheres. J Microencapsul 2012; 30:369-82. [DOI: 10.3109/02652048.2012.742158] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
21
|
Ma X, Oyamada S, Wu T, Robich MP, Wu H, Wang X, Buchholz B, McCarthy S, Bianchi CF, Sellke FW, Laham R. In vitro and in vivo degradation of poly(D, L-lactide-co-glycolide)/amorphous calcium phosphate copolymer coated on metal stents. J Biomed Mater Res A 2011; 96:632-8. [PMID: 21268237 PMCID: PMC3291328 DOI: 10.1002/jbm.a.33016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 10/14/2010] [Accepted: 11/29/2010] [Indexed: 12/20/2022]
Abstract
The purpose of this study was to optimize a novel biodegradable polymer for drug eluting stent (DES) applications. Degradation profiles of different poly(D,L-lactide-co-glycolide)/amorphous calcium phosphate (PLGA/ACP) composites coated on stents were studied both in vitro and in vivo for three months. For the in vitro study, stents were immersed into the phosphate buffered saline (37 °C, pH 7.4) with constant shaking. The polymer weight loss was measured weekly and morphological changes were analyzed. The results demonstrated that approximately 60% of polymer was degraded within the three-month period and there was no significant difference between the different PLGA/ACP composites. However, the composite of 50% PLGA (65/35) with 50% ACP showed a slightly faster degradation rate than other composites. Morphologically, all stent surfaces changed from a micro-porous before degradation to a corrugated solid micro-net-like structure at two months post degradation. Based on in vitro results, 65% PLGA (65/35) with 35% ACP) coated stents were selected and implanted into rat aortas (n = 12) for the in vivo study. Microscopic observation showed that no composite was found on any of the implanted stents at 12 weeks post implantation, which indicated the selected PLGA/ACP composite is desired for DES applications.
Collapse
Affiliation(s)
- Xiaodong Ma
- Biomedical Engineering and Biotechnology Doctoral Program, University of Massachusetts, Lowell, Massachusetts 01854, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Engineer C, Parikh J, Raval A. Effect of copolymer ratio on hydrolytic degradation of poly(lactide-co-glycolide) from drug eluting coronary stents. Chem Eng Res Des 2011. [DOI: 10.1016/j.cherd.2010.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
23
|
Preparation, characterization and in vivo pharmacodynamic evaluation of thymopentin loaded poly(lactide acid)/poly(lactide-co-glycolide acid) implants. Int J Pharm 2010; 398:123-9. [DOI: 10.1016/j.ijpharm.2010.07.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 07/09/2010] [Accepted: 07/22/2010] [Indexed: 11/20/2022]
|
24
|
Xi T, Gao R, Xu B, Chen L, Luo T, Liu J, Wei Y, Zhong S. In vitro and in vivo changes to PLGA/sirolimus coating on drug eluting stents. Biomaterials 2010; 31:5151-8. [PMID: 20382420 DOI: 10.1016/j.biomaterials.2010.02.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 02/02/2010] [Indexed: 10/19/2022]
|
25
|
Wischke C, Schwendeman SP. Principles of encapsulating hydrophobic drugs in PLA/PLGA microparticles. Int J Pharm 2008; 364:298-327. [PMID: 18621492 DOI: 10.1016/j.ijpharm.2008.04.042] [Citation(s) in RCA: 566] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 04/29/2008] [Accepted: 04/29/2008] [Indexed: 10/22/2022]
Abstract
Injectable biodegradable and biocompatible copolymers of lactic and glycolic acid (PLGA) are an important advanced delivery system for week-to-month controlled release of hydrophobic drugs (e.g., from biopharmaceutical classification system class IV), which often display poor oral bioavailability. The basic principles and considerations to develop such microparticle formulations is reviewed here based on a comprehensive study of papers and patents from the beginnings of hydrophobic drug encapsulation in polylactic acid and PLGA up through the very recent literature. Challenges with the diversity of drug properties, microencapsulation methods, and organic solvents are evaluated in light of the precedence of commercialized formulations and with a focus on decreasing the time to lab-scale encapsulation of water-insoluble drug candidates in the early stage of drug development. The influence of key formulation variables on final microparticle characteristics, and how best to avoid undesired microparticle properties, is analyzed mechanistically. Finally, concepts are developed to manage the common issues of maintaining sink conditions for in vitro drug release assays of hydrophobic compounds. Overall, against the backdrop of an increasing number of new, poorly orally available drug entities entering development, microparticle delivery systems may be a viable strategy to rescue an otherwise undeliverable substance.
Collapse
Affiliation(s)
- Christian Wischke
- Department of Pharmaceutical Sciences, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA
| | | |
Collapse
|
26
|
Abstract
BACKGROUND Biodegradable elastomers have been used in many different manners for controlled drug delivery. The development of new biodegradable elastomers has recently increased, driven mainly by tissue engineering research. OBJECTIVE This review outlines the different uses of biodegradable elastomers in controlled release. METHODS This review was limited to those papers wherein the polymer chosen as the delivery vehicle was demonstrably elastomeric. CONCLUSION Biodegradable elastomers have an established role in controlled release and an expanding role in combination scaffolds providing controlled release and mechanical stimulation capability for tissue regeneration/engineering.
Collapse
Affiliation(s)
- Brian G Amsden
- Queen's University, Department of Chemical Engineering, Kingston, Ontario, Canada.
| |
Collapse
|
27
|
Scaffolds from electrospun polyhydroxyalkanoate copolymers: Fabrication, characterization, bioabsorption and tissue response. Biomaterials 2008; 29:1307-17. [DOI: 10.1016/j.biomaterials.2007.11.031] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Accepted: 11/24/2007] [Indexed: 11/18/2022]
|
28
|
Amsden B. Curable, biodegradable elastomers: emerging biomaterials for drug delivery and tissue engineering. SOFT MATTER 2007; 3:1335-1348. [PMID: 32900112 DOI: 10.1039/b707472g] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Biodegradable elastomers have a number of potential applications in the biomedical area, especially in the emerging field of soft-tissue engineering where the mechanical properties of the polymer scaffold should match those of the tissue to be grown. An increasing number of synthesis strategies have been employed in order to prepare such elastomers. In this review, these synthesis strategies and the properties of these elastomers are outlined. The factors that influence the characteristics of these elastomers including mechanical properties, degradation rate, and mechanical property change during degradation, are discussed in terms of the design of the elastomer and their advantages and disadvantages for the biomedical applications considered.
Collapse
Affiliation(s)
- Brian Amsden
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
29
|
Kim MS, Ahn HH, Shin YN, Cho MH, Khang G, Lee HB. An in vivo study of the host tissue response to subcutaneous implantation of PLGA- and/or porcine small intestinal submucosa-based scaffolds. Biomaterials 2007; 28:5137-43. [PMID: 17764737 DOI: 10.1016/j.biomaterials.2007.08.014] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 08/02/2007] [Indexed: 10/22/2022]
Abstract
An innate immune response is often found at the site of biomaterial implantation. Since the effective use of biomaterials in vivo requires good biocompatibility and biofunctionality, it is vital that we assess and compare the inflammatory reactions provoked by various implanted biomaterials in vivo. In the present study, we assessed the host tissue response to poly(lactic-co-glycolic acid) (PLGA)- and small intestinal submucosa (SIS)-based scaffolds subcutaneously implanted in Fischer rats. Our results revealed that the PLGA-based scaffolds resulted in severe post-implantation inflammation, whereas the SIS-based scaffolds induced only a slight post-implantation inflammation and a PLGA/SIS-based copolymer yielded intermediate results.
Collapse
Affiliation(s)
- Moon Suk Kim
- Fusion Biotechnology Research Center, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong, Daejeon 305-600, Republic of Korea
| | | | | | | | | | | |
Collapse
|
30
|
Luong-Van E, Grøndahl L, Song S, Nurcombe V, Cool S. The in vivo assessment of a novel scaffold containing heparan sulfate for tissue engineering with human mesenchymal stem cells. J Mol Histol 2007; 38:459-68. [PMID: 17694276 DOI: 10.1007/s10735-007-9129-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 07/26/2007] [Indexed: 10/23/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive tissue engineering avenue for the repair and regeneration of bone. In this study we detail the in vivo performance of a novel electrospun polycaprolactone scaffold incorporating the glycosaminoglycan heparan sulfate (HS) as a carrier for hMSC. HS is a multifunctional regulator of many key growth factors expressed endogenously during bone wound repair, and we have found it to be a potent stimulator of proliferation in hMSCs. To assess the potential of the scaffolds to support hMSC function in vivo, hMSCs pre-committed to the osteogenic lineage (human osteoprogenitor cells) were seeded onto the scaffolds and implanted subcutaneously into the dorsum of nude rats. After 6 weeks the scaffolds were retrieved and examined by histological methods. Implanted human cells were identified using a human nuclei-specific antibody. The host response to the implants was characterized by ED1 and ED2 antibody staining for monocytes/macrophages and mature tissue macrophages, respectively. It was found that the survival of the implanted human cells was affected by the host response to the implant regardless of the presence of HS, highlighting the importance of controlling the host response to tissue engineering devices.
Collapse
Affiliation(s)
- Emma Luong-Van
- Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore
| | | | | | | | | |
Collapse
|
31
|
Fulzele SV, Satturwar PM, Dorle AK. Novel Biopolymers as Implant Matrix for the Deliveryof Ciprofloxacin: Biocompatibility, Degradation, and In Vitro Antibiotic Release. J Pharm Sci 2007; 96:132-44. [PMID: 16960824 DOI: 10.1002/jps.20730] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The purpose of this study was to investigate the in vitro-in vivo degradation and tissue compatibility of three novel biopolymers viz. polymerized rosin (PR), glycerol ester of polymerized rosin (GPR) and pentaerythritol ester of polymerized rosin (PPR) and study their potential as implant matrix for the delivery of ciprofloxacin hydrochloride. Free films of polymers were used for in vitro degradation in PBS (pH 7.4) and in vivo in rat subcutaneous model. Sample weight loss, molecular weight decline, and morphological changes were analyzed after periodic intervals (30, 60, and 90 days) to monitor the degradation profile. Biocompatibility was evaluated by examination of the inflammatory tissue response to the implanted films on postoperative days 7, 14, 21, and 28. Furthermore, direct compression of dry blends of various polymer matrices with 20%, 30%, and 40% w/w drug loading was performed to investigate their potential for implant systems. The implants were characterized in terms of porosity and ciprofloxacin release. Biopolymer films showed slow rate of degradation, in vivo rate being faster on comparative basis. Heterogeneous bulk degradation was evident with the esterified products showing faster rates than PR. Morphologically all the films were stiff and intact with no significant difference in their appearance. The percent weight remaining in vivo was 90.70 +/- 6.2, 85.59 +/- 5.8, and 75.56 +/- 4.8 for PR, GPR, and PPR films respectively. Initial rapid drop in Mw was demonstrated with nearly 20.0% and 30.0% decline within 30 days followed by a steady decline to nearly 40.0% and 50.0% within 90 days following in vitro and in vivo degradation respectively. Biocompatibility demonstrated by acute and subacute tissue reactions showed minimal inflammatory reactions with prominent fibrous encapsulation and absence of necrosis demonstrating good tissue compatibility to the extent evaluated. All implants showed erosion and increase in porosity that affected the drug release. Increase in drug loading significantly altered the ciprofloxacin release in extended dissolution studies. PPR produced drug release >90% over a period of 90 days promising its utility in implant systems. The results demonstrated the utility of novel film forming biopolymers as implant matrix for controlled/sustained drug delivery with excellent biocompatibility characteristics.
Collapse
Affiliation(s)
- Suniket V Fulzele
- Department of Pharmaceutical Sciences, Nagpur University Campus, Amravati Road, Nagpur-440 033, India
| | | | | |
Collapse
|
32
|
van Dijkhuizen-Radersma R, Roosma JR, Sohier J, Péters FLAMA, van den Doel M, van Blitterswijk CA, de Groot K, Bezemer JM. Biodegradable poly(ether-ester) multiblock copolymers for controlled release applications: Anin vivoevaluation. J Biomed Mater Res A 2004; 71:118-27. [PMID: 15368261 DOI: 10.1002/jbm.a.30136] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiblock poly(ether-ester)s based on poly(ethylene glycol), butylene terephthalate, and butylene succinate segments were evaluated for their in vivo degradation and biocompatibility in order to establish a correlation with previously reported in vitro results. Porous polymer sheets were implanted subcutaneously for 32 weeks in rats. The degradation was monitored visually (histology), by molecular weight (GPC), and by copolymer composition (NMR). Substitution of the aromatic terephthalate units by aliphatic succinate units was shown to accelerate the degradation rate of the copolymers. Direct correlation of the in vivo and in vitro degradation of the porous implants showed a slightly faster initial molecular weight decrease in vivo. Besides hydrolysis, oxidation occurs in vivo due to the presence of radicals produced by inflammatory cells. In addition, the higher molecular weight plateau of the residue found in vivo indicated a higher solubility of the oligomers in the extracellular fluid compared to a phosphate buffer. Minor changes in the poly(ether-ester) compositions were noted due to degradation. Microscopically, fragmentation of the porous implants was observed in time. At later stages of degradation, macrophages were observed phagocytozing small polymer particles. Both in vitro cytotoxicity studies and histology on in vivo samples proved the biocompatibility of the poly(ether-ester)s.
Collapse
|
33
|
Aliyev E, Sakallioğlu U, Eren Z, Açikgöz G. The effect of polylactide membranes on the levels of reactive oxygen species in periodontal flaps during wound healing. Biomaterials 2004; 25:4633-7. [PMID: 15120509 DOI: 10.1016/j.biomaterials.2003.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Accepted: 12/02/2003] [Indexed: 10/26/2022]
Abstract
It is consented that reactive oxygen species (ROS) are deleterious to wound healing process due to the harmful effects on cells and tissues. Absorbable synthetic biomaterials are considered to be degraded via ROS. Free-radical-scavenging enzymes (FRSE) are a cytoprotective enzymal group that has an essential role in the reduction, de-activation and removal of ROS as well as regulating wound healing process. In the present study, synthetic and absorbable polylactide (PLA) barrier membranes were evaluated by means of ROS activity levels during degradation in the healing periodontal flaps measuring the activity of FRSE superoxide dismutase (SOD) and catalase (CAT). Gingival biopsies taken from 10 patients allowing both guided tissue regeneration (test) and conventional flap surgery (control) before and 1 month after the operations were processed and the supernatants were studied by Mc Cord and Fridovich, Flohe and Otting, and Luck methods to measure total SOD and CAT levels respectively. A significantly increased enzyme activity of SOD and CAT was observed in both groups (p<0.05). SOD activity change was 62.92% in the test and 3.97% in the control group, and, CAT activity change was 48.04% in the test and 11.58% in the control group. Our results suggest that ROS, particularly superoxide anions, may contribute to the degradation phase of PLA membranes and this may affect the wound healing of periodontium at least for one-month period.
Collapse
Affiliation(s)
- Eldar Aliyev
- Department of Molecular Biology, Faculty of Science, University of Ondokuz Mayis, Samsun 55139, Turkey
| | | | | | | |
Collapse
|
34
|
Xu P, Sefton MV. Expression of matrix metalloproteinase-2 and -9 in exudates associated with polydimethyl siloxane and gelatin tubes implanted in mice. ACTA ACUST UNITED AC 2004; 71:226-32. [PMID: 15386490 DOI: 10.1002/jbm.a.30139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The expression of matrix metalloproteinases (MMPs), MMP-2 and -9, during the inflammatory response to two implanted biomaterials was determined. To illustrate this phenomenon, polydimethyl siloxane (PDMS), weakly crosslinked gelatin, and highly crosslinked gelatin tubes were implanted subcutaneously in mice for up to 3 weeks. Latent MMP-2 (pro-MMP-2) was seen in exudates throughout the experimental period and did not show any apparent changes in time or among the three biomaterials. The expression of MMP-9, however, showed a very different pattern. At days 4 and 8, the predominant form of MMP-9 is pro-MMP-9 (the latent form) and its level is much higher for the PDMS tube than either of the gelatin tubes. The active form of MMP-9, initially low, rose progressively over the 3-week period and reached its peak at week 2 for PDMS and highly crosslinked gelatin and at week 3 for the more degradable, weakly crosslinked gelatin. Data from the histological and cytological analysis also showed that the PDMS tube induced a much stronger cellular response than gelatin tubes, although endotoxin contamination precludes a conclusion related to biomaterial effects. We concluded that MMP-9 is a useful marker of the host inflammatory response to the implanted biomaterials and it may be useful in the assessment of biomaterial biocompatibility. Perhaps more importantly, the consequences of MMPs on matrix remodeling may become of concern in biomaterials used for tissue engineering.
Collapse
Affiliation(s)
- Ping Xu
- Department of Chemical Engineering and Applied Chemistry, and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9
| | | |
Collapse
|
35
|
Satturwar PM, Fulzele SV, Dorle AK. Biodegradation and in vivo biocompatibility of rosin: a natural film-forming polymer. AAPS PharmSciTech 2003; 4:E55. [PMID: 15198550 PMCID: PMC2750648 DOI: 10.1208/pt040455] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The specific aim of the present study was to investigate the biodegradation and biocompatibility characteristics of rosin, a natural film-forming polymer. Both in vitro as well as in vivo methods were used for assessment of the same. The in vitro degradation of rosin films was followed in pH 7.4 phosphate buffered saline at 37 degrees C and in vivo by subdermal implantation in rats for up to 90 days. Initial biocompatibility was followed on postoperative days 7, 14, 21, and 28 by histological observations of the surrounding tissues around the implanted films. Poly (DL-lactic-co-glycolic acid) (PLGA) (50:50) was used as reference material for biocompatibility. Rate and extent of degradation were followed in terms of dry film weight loss, molecular weight (MW) decline, and surface morphological changes. Although the rate of in vitro degradation was slow, rosin-free films showed complete degradation between 60 and 90 days following subdermal implantation in rats. The films degraded following different rates, in vitro and in vivo, but the mechanism followed was primarily bulk degradation. Rosin films demonstrated inflammatory reactions similar to PLGA, indicative of good biocompatibility. Good biocompatibility comparable to PLGA is demonstrated by the absence of necrosis or abscess formation in the surrounding tissues. The study provides valuable insight, which may lead to new applications of rosin in the field of drug delivery.
Collapse
Affiliation(s)
- Prashant M Satturwar
- Department of Pharmaceutical Sciences, Nagpur University Campus, Amravati Road, Nagpur-440010, India.
| | | | | |
Collapse
|
36
|
Biggs DL, Lengsfeld CS, Hybertson BM, Ng KY, Manning MC, Randolph TW. In vitro and in vivo evaluation of the effects of PLA microparticle crystallinity on cellular response. J Control Release 2003; 92:147-61. [PMID: 14499193 DOI: 10.1016/s0168-3659(03)00325-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous research suggests that crystallinity of poly(L-lactide) P(L)LA microparticles can influence surface free energy, which in turn might influence biocompatibility. This work studies the cellular response to P(L)LA microparticles of different crystallinity both in vitro and in vivo. Following incubation with P(L)LA microparticles, the in vitro production of reactive oxygen intermediates (ROI) was measured as a marker of cellular response. In both fluorescence and chemiluminescence experiments to measure ROI, a small effect of microparticle crystallinity on NR8383 AM response was observed. Microparticles of higher crystallinity elicited a smaller inflammatory response compared to lower crystallinity particles. Compared to the elevated inflammatory response induced by zymosan, the response to all P(L)LA microparticles tested was practically negligible. Results from in vivo experiments further supported conclusions that P(L)LA microparticles elicit minimal inflammatory response. Following acute exposure to P(L)LA microparticles in guinea-pig lungs, the inflammatory response was not significantly different from the response observed when sterile saline was administered. In contrast to the in vitro experiments, there were not apparent differences in cellular responses to microparticles of different crystallinity.
Collapse
Affiliation(s)
- Danielle L Biggs
- Department of Chemical Engineering, Center for Pharmaceutical Biotechnology, University of Colorado, Engineering Center, Room ECCH 111,, Boulder, CO 80309-0424, USA
| | | | | | | | | | | |
Collapse
|
37
|
Fulzele SV, Satturwar PM, Dorle AK. Study of the biodegradation and in vivo biocompatibility of novel biomaterials. Eur J Pharm Sci 2003; 20:53-61. [PMID: 13678793 DOI: 10.1016/s0928-0987(03)00168-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The degradation of two rosin-based biomaterials, the glycerol ester of maleic rosin (GMR) and the pentaerythritol ester of maleic rosin (PMR), was examined in vitro in phosphate-buffered saline at pH 7.4 and in vivo in a subcutaneous rat model. Free films of the two biomaterials with mean thickness 0.4+/-0.02 mm were used for the study. The initial biocompatibility was followed by microscopic examination of the inflammatory tissue response to the implanted films. Sample weight loss and molecular weight decline of the free films was used to monitor the degradation quantitatively, while surface morphological changes were analysed for qualitative estimation. Biocompatibility response was followed on post-operative days 7, 14, 21 and 28 and compared with those of poly(DL-lactic-co-glycolic acid) (PLGA) (50:50) films. Both biomaterials showed slow in vitro degradation when compared with the in vivo rate. The mechanism followed was, however, bulk degradation of the films. The penta-esterified form of maleic rosin was observed to degrade more rapidly than glycerol esterified maleic rosin. The acute and subacute inflammatory reactions were characterized by fibrosis at the end of 28 days. The biomaterials showed reasonable tissue tolerance to the extent evaluated. There was a total absence of tissue necrosis or abscess formation for all implanted films. The response, although not identical to that of PLGA, is reasonable, promising new drug delivery applications for rosin biomaterials.
Collapse
Affiliation(s)
- S V Fulzele
- Department of Pharmaceutical Sciences, Nagpur University Campus, Amravati Road, 440010, Nagpur, India.
| | | | | |
Collapse
|
38
|
Satturwar PM, Mandaogade PM, Darwhekar GN, Fulzele SV, Joshi SB, Dorle AK. Biodegradation studies of rosin-based polymers. Drug Dev Ind Pharm 2003; 29:669-77. [PMID: 12889785 DOI: 10.1081/ddc-120021316] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This study was designed to investigate two rosin-based polymers (R-1 and R-2) for their in vitro and in vivo biodegradation behavior. The in vitro hydrolytic degradation was carried out in buffer solutions of pH 4.4, 7.4, and 10.4 at 37 degrees C. Enzymatic degradation was studied using enzymes lipase, pancreatine, and pectinase. Free films of the two polymers were subcutaneously implanted in rabbits for the in vivo biodegradation. The extent of degradation was determined quantitatively by weight loss and was followed qualitatively by scanning electron microscopy. The extent and the rate of degradation was better in vivo than in vitro. The polymers showed poor enzymatic degradation and a highly pH-dependent hydrolytic degradation.
Collapse
Affiliation(s)
- P M Satturwar
- Department of Pharmaceutical Sciences, Nagpur University, Nagpur, India
| | | | | | | | | | | |
Collapse
|
39
|
Sandor M, Harris J, Mathiowitz E. A novel polyethylene depot device for the study of PLGA and P(FASA) microspheres in vitro and in vivo. Biomaterials 2002; 23:4413-23. [PMID: 12219832 DOI: 10.1016/s0142-9612(02)00183-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Polymer microspheres (0.5-5.0 microm) are difficult to characterize in vivo because they degrade, migrate, and are endocytosed. A novel polyethylene mesh pouch containing microspheres allowed for retrieval of degraded polymeric products from rats without affecting the rate of degradation. Pouches containing poly(lactic-co-glycolic acid) (PLGA) or poly(fumaric-co-sebacic acid) (P(FASA)) microspheres were implanted intramuscularly, subcutaneously, and intraperitoneally and analyzed after 3, 7, 14, and 28 days. In vivo, subcutaneous or intraperitoneal implants experienced an immediate mass loss and a delayed decrease in molecular weight (Mw). Intramuscular implants behaved similarly to in vitro samples, decreasing in Mw immediately and lagging in mass loss. These results suggest that mass loss, which is usually dependent on Mw loss in vitro, may be directly due to enzymatic, rather than hydrolytic, degradation subcutaneously and intraperitoneally, while intramuscular implants appear to be mostly dependent on hydrolytic cleavage. This observation is further supported by histology. Additional experiments on pouches loaded with PLGA microspheres encapsulating osteoprotegerin, a protein drug used to prevent bone resorption, revealed that use of the device prevented the artifactual polymer compression inherent to microsphere centrifugation during release studies and allowed for the extraction of active protein from microspheres implanted for 3 days in vivo.
Collapse
Affiliation(s)
- Maryellen Sandor
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | | | | |
Collapse
|
40
|
Zippel R, Wilhelm L, Marusch F, Koch A, Urban G, Schlosser M. Antigenicity of Polyester (Dacron) Vascular Prostheses in an Animal Model. Eur J Vasc Endovasc Surg 2001; 21:202-7. [PMID: 11352677 DOI: 10.1053/ejvs.2000.1298] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Objectives to investigate the specific humoral immune response to three different polyester (Dacron) prostheses in pigs. Design, materials and methods twenty-four growing pigs were randomly divided into three groups. The infrarenal aorta was replaced by a segment of collagen-impregnated woven polyester prosthesis (low, medium and high porosity). Serum antibodies were detected by modified enzyme immunoassay using non-impregnated prosthesis as the target for the blood samples taken on experimental days 1, 10, 17, 24, 62 and 116 of the 22 pigs followed over the whole observation period. Results significantly enhanced (p <0.05) mean IgG antibody binding against polyester was detected on experimental days 10, 17, 24 and 62 with antibody prevalences of 41%, 41%, 32% and 37%, respectively. Antibody positive pigs were divided into early responders (n =9) and late responders ( n =5) with antibody detection on day 10 and/or 17 vs day 62 and/or 116. No significant differences between the three different prostheses were found. The formation of specific IgG antibodies against polyester in the animals investigated demonstrates a broad individual variability. Conclusions polyester is an antigenic polymer. Specific antibodies, reflecting the inflammatory response, might be not only a parameter for testing biomaterials but also for determining individual bio(in)compatibility for long-term biomaterial function.
Collapse
Affiliation(s)
- R Zippel
- Department of Surgery, Carl Thiem Hospital, Thiem Street 111, Cottbus, D-03048, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Kamalesh S, Tan P, Wang J, Lee T, Kang ET, Wang CH. Biocompatibility of electroactive polymers in tissues. JOURNAL OF BIOMEDICAL MATERIALS RESEARCH 2000; 52:467-78. [PMID: 11007614 DOI: 10.1002/1097-4636(20001205)52:3<467::aid-jbm4>3.0.co;2-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The biocompatibility of ethylene-vinyl acetate copolymer (EVAc), polyethylene (PE), and polyaniline (PANi) films in the emeraldine (EM), nigraniline (NA) and leucoemeraldine (LM) intrinsic oxidation states were assessed through subcutaneous implantation into male Sprague-Dawley rats beneath the dorsal skin, for a period ranging from 19 to 90 weeks. Histological examination, interstitial pressure measurement, and X-ray photoelectron spectroscopy (XPS) were employed to determine the biocompatibility of the polymers. The polymers did not provoke inflammatory responses in the subcutaneous tissues over the entire implantation period. Characteristics features associated with tissue-implant incompatibility were not evident near the implantation. Interstitial pressure was measured to evaluate the development of tissue. Low interstitial pressure readings on the region of implantation confirmed the biocompatibility of these polymer types. The surface composition of the electroactive aniline polymers before and after the implantation was characterized by XPS.
Collapse
Affiliation(s)
- S Kamalesh
- Department of Chemical and Environmental Engineering, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
In the last 3 decades, much progress has been made in the development of biodegradable osteosyntheses. Despite this progress, these materials are still only used in small numbers, and the scope of their application has been limited. The limitations of biodegradable osteosyntheses mainly are related to problems with their mechanical properties and, in particular, biocompatibility. These problems need to be solved so that biodegradable osteosyntheses can perform up to their full potential and thus, eventually, make their general clinical application routine. This paper presents a historical perspective on the development of biodegradable osteosyntheses, discusses the successful developmental achievements and the still-existing problems, and gives a perspective on their future development.
Collapse
Affiliation(s)
- F W Cordewener
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | |
Collapse
|
43
|
Schmitz JP, Lemke RR, Zardeneta G, Hollinger JO, Milam SB. Isolation of particulate degradation debris 1 year after implantation of a Guidor membrane for guided bone regeneration: case report. J Oral Maxillofac Surg 2000; 58:888-93. [PMID: 10935589 DOI: 10.1053/joms.2000.8216] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- J P Schmitz
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at San Antonio, 78284-7823, USA.
| | | | | | | | | |
Collapse
|
44
|
Blanco MD, Gómez C, Olmo R, Muñiz E, Teijón JM. Chitosan microspheres in PLG films as devices for cytarabine release. Int J Pharm 2000; 202:29-39. [PMID: 10915924 DOI: 10.1016/s0378-5173(00)00408-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cytarabine was included in chitosan microspheres and several of these microspheres were embedded in a poly(lactide-co-glycolide) (PLG) film to constitute a comatrix system, to develop a prolonged release form. Chitosan microspheres, in the range of 92+/-65 microm, having good spherical geometry and a smooth surface incorporating cytarabine, were prepared. The cytarabine amount included in chitosan microspheres was 43.7 microg of ara-C per milligram microsphere. The incorporation efficiency of the cytarabine in microspheres was 70.6%. Total cytarabine release from microspheres in vitro was detected at 48 h. Inclusion of cytarabine-loaded microspheres in poly(lactide-co-glycolide) film initiated a slower release of the drug and, in this way, the maximum of cytarabine released (80%) took place in vitro at 94.5 h. Comatrices, with 8.7 mg of cytarabine, signifying a dose of 34.5 microg/kg, were subcutaneously implanted in the back of rats. Maximum plasma cytarabine concentration was 18.5+/-1.5 microg/ml, 48 h after the device implantation and the drug was detected in plasma for 13 days. The histological studies show a slow degradative process. After 6 months of implantation, most of the microspheres of the matrix seemed to be intact, the comatrix appeared surrounded by conjunctive tissue and small blood vessels and nerve packets were detected in the periphery of the implant.
Collapse
Affiliation(s)
- M D Blanco
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | | | | | | |
Collapse
|
45
|
Tjia JS, Moghe PV. Analysis of 3-D microstructure of porous poly(lactide-glycolide) matrices using confocal microscopy. JOURNAL OF BIOMEDICAL MATERIALS RESEARCH 2000; 43:291-9. [PMID: 9730067 DOI: 10.1002/(sici)1097-4636(199823)43:3<291::aid-jbm10>3.0.co;2-j] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Porous matrices of biodegradable polymers are extensively used as scaffolds in tissue engineering and as drug delivery devices. A critical component of the design, processing, and utility of such polymeric systems concerns the local void microarchitecture. In this study, a novel approach based on confocal fluorescence imaging was employed to visualize and quantify in 3 dimensions (3-D) the individual and population-level void morphology within porous polymeric matrices. Poly(lactic acid-glycolic acid) copolymer matrices were cast to yield void configurations of variable void sizes but constant cumulative voidage. Using confocal microscopy, fluorescently saturated polymer matrices were optically sectioned into serial 2-D images, and 3-D void contours were reconstructed via object discrimination and connectivity analysis. The resultant data was used to quantitate the matrix microstructure and map its evolution following polymer degradation. Under conditions of accelerated degradation, matrix erosion was found to cause a significant change in the disposition of voids; this involves two processes (void formation and void enlargement), the extent of which was influenced by the initial void size and the duration of erosion. By virtue of providing both static and dynamic descriptions of the void morphology in poly(lactic acid-glycolic acid) matrices, this is the first spatiotemporal study of the 3-D microarchitecture of porous, bioerodible tissue analog matrices.
Collapse
Affiliation(s)
- J S Tjia
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
46
|
Wooley PH, Fitzgerald RH, Song Z, Davis P, Whalen JD, Trumble S, Nasser S. Proteins bound to polyethylene components in patients who have aseptic loosening after total joint arthroplasty. A preliminary report. J Bone Joint Surg Am 1999; 81:616-23. [PMID: 10360690 DOI: 10.2106/00004623-199905000-00003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Immunological responses to proteins that adhere to ultra-high molecular weight polyethylene have not, to our knowledge, been examined previously in patients who have aseptic loosening. In the current study, polyethylene components from forty-nine failed prostheses recovered during revision procedures were examined for the presence of antibodies that were bound to the polyethylene surface or that were reactive with other proteins that were bound to the polyethylene surface. METHODS The polyethylene components consisted of thirty acetabular cups recovered during revision total hip arthroplasties and nineteen tibial components recovered during revision total knee arthroplasties. After extensive washing, bound proteins were extracted from the polyethylene components with use of 0.1-molar glycine-hydrogen chloride solution followed by four-molar guanidine hydrochloride solution. RESULTS Sufficient protein for analysis was recovered from forty-two polyethylene components. Polyacrylamide gel electrophoresis demonstrated a minimum of one and a maximum of twelve protein bands, with molecular weights ranging from thirteen to 231 kilodaltons. Immunoblotting revealed the presence of type-I collagen in most (thirty-four) of the forty-two explants, whereas aggrecan proteoglycans were detected in eight samples. Immunoglobulin also was detected in most (thirty-three) extracts, whereas type-II collagen was consistently absent. The presence of autologous antibodies directed against polyethylene-bound proteins in sera drawn at the time of the revision was investigated. Antibodies that were reactive against the ultra-high molecular weight polyethylene-bound proteins were detected in twenty-six of the forty-two patients with use of the Western blot technique. The number of reactive bands ranged from one to six, and the strongest binding was directed against a 103-kilodalton protein. Assays for specificity revealed that these sera autologous antibodies were reactive against the type-I collagen that was present in the explant solutions. CONCLUSIONS We hypothesize that immunoglobulin complexed with polyethylene may fix complement and that the complement cascade may in turn attract inflammatory cells to the polyethylene surface. Our data support the hypothesis that an immunological response to antigens bound to the polyethylene surface may contribute to aseptic loosening. CLINICAL RELEVANCE Despite improvements in materials and designs of prostheses, aseptic loosening is the most common complication of total joint replacement, frequently leading to revision operations. We examined the immunological response to proteins that bind to ultra-high molecular weight polyethylene in patients who had aseptic loosening and discovered a high prevalence of antibodies to polyethylene-bound proteins. This immunological response may contribute to an inflammatory reaction in the periprosthetic tissue, ultimately leading to increased bone resorption around the prosthesis.
Collapse
Affiliation(s)
- P H Wooley
- Department of Orthopaedic Surgery, Wayne State University School of Medicine, Hutzel Hospital, and the Veterans Administration Medical Center, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Smetana K, Lukás J, Palecková V, Bartůnkovä J, Liu FT, Vacík J, Gabius HJ. Effect of chemical structure of hydrogels on the adhesion and phenotypic characteristics of human monocytes such as expression of galectins and other carbohydrate-binding sites. Biomaterials 1997; 18:1009-14. [PMID: 9212197 DOI: 10.1016/s0142-9612(97)00037-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The reactivity of diverse immune aspects to the presence of synthetic polymers represents one of the most important aspects of implantable device biocompatibility. In this report, we show the effect of the chemical structure of a synthetic polymer support on monocyte adhesion and selected phenotypic characteristics in vitro as a model for the initial steps of non-self-recognition of an implant. The extent of monocyte adhesion was significantly influenced by the support chemistry. The highest level of monocyte adhesion was observed on a surface copolymer of 2-hydroxyethyl methacrylate with dimethyl aminoethyl methacrylate relative to results of experiments in which poly(2-hydroxyethyl methacrylate) or the copolymer of 2-hydroxyethyl methacrylate with the sodium salt of methacrylic acid was used. Cell adhesion to the polymers tested and to glass was accompanied by enhanced expression of the carbohydrate-binding sites tested for asialoglycoprotein beta-galactosides such as galectins, beta-N-acetylgalactosamine, alpha-mannoside, specific lectin for heparin as well as the lymphokine-macrophage migration inhibitory factor in the monocytes tested. These results suggest the importance of monocyte adhesion to the biomaterial surface for their development into macrophages and further non-self-recognition of the implanted device.
Collapse
Affiliation(s)
- K Smetana
- Institute of Anatomy, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
48
|
Smetana K, Jelínková M, Vacík J, Fischer J, Gabius HJ. Influence of intraperitoneal injection of three types of hydrogel beads on expression of carbohydrate-binding sites in spleen macrophages. Biomaterials 1996; 17:2335-41. [PMID: 8982473 DOI: 10.1016/s0142-9612(96)00083-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The influence of the chemical structure of polymer implants on selected characteristics of macrophages was studied to improve our understanding of the mechanisms of non-self recognition of synthetic materials. Three types of polymers differing in net charge were prepared to compare in vivo responses. Beads from preparation of poly(2-hydroxyethyl methacrylate), a copolymer of 2-hydroxyethyl methacrylate with sodium methacrylate, and a copolymer of 2-hydroxyethyl methacrylate and dimethylaminoethyl methacrylate were injected intraperitoneally into rats and harvested 48 h later. The effects of these polymers on the presence of inflammatory cells in the peritoneal exudate, on the adhesion of macrophages to individual batches of the different types of beads and on distinct molecular aspects of macrophages in the red pulp of spleen were evaluated. Beads from both types of copolymer caused an elevation in the number of macrophages in the exudate, in contrast to the situation in rats treated with poly(2-hydroxyethyl methacrylate) beads and physiological saline solution as control. The molecular design of the implant had no significant influence on the extent of macrophage adhesion to beads or on the expression of selected carbohydrate-binding sites. Since important cellular functions such as cell adhesion and glycoprotein routing depend on the sugar part of glycoconjugates, labelled neoglycoproteins were employed to analyse this aspect of macrophages in the tested animals. The beads of the copolymer of 2-hydroxyethyl methacrylate with dimethylaminoethyl methacrylate clearly led to an elevation of the expression of specific binding sites for beta-galactoside-terminating structures which are presented by asialofetuin, for mannose, fucose, sialic acid and N-acetylgalactosamine, which had been used as the ligand parts of biotinylated neoglycoproteins, in spleen macrophages whereas the levels of sites which recognize mannose-6-phosphate were unaffected. Expression of sites with specificity to N-acetylglucosamine was lessened. The effect of beads from the copolymer of 2-hydroxyethyl methacrylate with sodium methacrylate on the measured glycobiological features in the splenic macrophages was only negligible. These results suggest the possibility of systemic effects of implanted polymers on the distinct recognitive functions of macrophages.
Collapse
Affiliation(s)
- K Smetana
- Institute of Anatomy 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
49
|
Pouton CW, Akhtar S. Biosynthetic polyhydroxyalkanoates and their potential in drug delivery. Adv Drug Deliv Rev 1996. [DOI: 10.1016/0169-409x(95)00092-l] [Citation(s) in RCA: 187] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|