1
|
Moody CT, Durham PG, Dayton PA, Brudno Y. Loading Intracranial Drug-Eluting Reservoirs Across the Blood-Brain Barrier With Focused Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1679-1685. [PMID: 37120330 PMCID: PMC10192093 DOI: 10.1016/j.ultrasmedbio.2023.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 05/10/2023]
Abstract
OBJECTIVE Efficient, sustained and long-term delivery of therapeutics to the brain remains an important challenge to treatment of diseases such as brain cancer, stroke and neurodegenerative disease. Focused ultrasound can assist movement of drugs into the brain, but frequent and long-term use has remained impractical. Single-use intracranial drug-eluting depots show promise but are limited for the treatment of chronic diseases as they cannot be refilled non-invasively. Refillable drug-eluting depots could serve as a long-term solution, but refilling is hindered by the blood-brain barrier (BBB), which prevents drug refills from accessing the brain. In this article, we describe how focused ultrasound enables non-invasive loading of intracranial drug depots in mice. METHODS Female CD-1 mice (n = 6) were intracranially injected with click-reactive and fluorescent molecules that are capable of anchoring in the brain. After healing, animals were treated with high-intensity focused ultrasound and microbubbles to temporarily increase the permeability of the blood-brain barrier and deliver dibenzocyclooctyne (DBCO)-Cy7. The mice were perfused, and the brains were imaged via ex vivo fluorescence imaging. RESULTS Fluorescence imaging indicated small molecule refills are captured by intracranial depots as long as 4 wk after administration and are retained for up to 4 wk based on fluorescence imaging. Efficient loading was dependent on both focused ultrasound and the presence of refillable depots in the brain as absence of either prevented intracranial loading. CONCLUSION The ability to target and retain small molecules at predetermined intracranial sites with pinpoint accuracy provides opportunities to continuously deliver drugs to the brain over weeks and months without excessive BBB opening and with minimal off-target side effects.
Collapse
Affiliation(s)
- Christopher T. Moody
- Joint Department of Biomedical Engineering. University of North Carolina – Chapel Hill and North Carolina State University – Raleigh. 1840 Entrepreneur Drive. Raleigh, NC 27695, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC USA
| | - Phillip G Durham
- Joint Department of Biomedical Engineering. University of North Carolina – Chapel Hill and North Carolina State University – Raleigh. 1840 Entrepreneur Drive. Raleigh, NC 27695, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering. University of North Carolina – Chapel Hill and North Carolina State University – Raleigh. 1840 Entrepreneur Drive. Raleigh, NC 27695, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering. University of North Carolina – Chapel Hill and North Carolina State University – Raleigh. 1840 Entrepreneur Drive. Raleigh, NC 27695, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC USA
| |
Collapse
|
2
|
Agarwalla P, Ogunnaike EA, Ahn S, Froehlich KA, Jansson A, Ligler FS, Dotti G, Brudno Y. Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells. Nat Biotechnol 2022; 40:1250-1258. [PMID: 35332339 PMCID: PMC9376243 DOI: 10.1038/s41587-022-01245-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/02/2022] [Indexed: 12/29/2022]
Abstract
Despite their clinical success, chimeric antigen receptor (CAR)-T cell therapies for B cell malignancies are limited by lengthy, costly and labor-intensive ex vivo manufacturing procedures that might lead to cell products with heterogeneous composition. Here we describe an implantable Multifunctional Alginate Scaffold for T Cell Engineering and Release (MASTER) that streamlines in vivo CAR-T cell manufacturing and reduces processing time to a single day. When seeded with human peripheral blood mononuclear cells and CD19-encoding retroviral particles, MASTER provides the appropriate interface for viral vector-mediated gene transfer and, after subcutaneous implantation, mediates the release of functional CAR-T cells in mice. We further demonstrate that in vivo-generated CAR-T cells enter the bloodstream and control distal tumor growth in a mouse xenograft model of lymphoma, showing greater persistence than conventional CAR-T cells. MASTER promises to transform CAR-T cell therapy by fast-tracking manufacture and potentially reducing the complexity and resources needed for provision of this type of therapy.
Collapse
Affiliation(s)
- Pritha Agarwalla
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Edikan A Ogunnaike
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah Ahn
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristen A Froehlich
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anton Jansson
- Analytical Instrumentation Facility, North Carolina State University, Raleigh, NC, USA
| | - Frances S Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA.
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Moody CT, Brown AE, Massaro NP, Patel AS, Agarwalla PA, Simpson AM, Brown AC, Zheng H, Pierce JG, Brudno Y. Restoring Carboxylates on Highly Modified Alginates Improves Gelation, Tissue Retention and Systemic Capture. Acta Biomater 2022; 138:208-217. [PMID: 34728426 PMCID: PMC8738153 DOI: 10.1016/j.actbio.2021.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/05/2021] [Accepted: 10/26/2021] [Indexed: 01/17/2023]
Abstract
Alginate hydrogels are gaining traction for use in drug delivery, regenerative medicine, and as tissue engineered scaffolds due to their physiological gelation conditions, high tissue biocompatibility, and wide chemical versatility. Traditionally, alginate is decorated at the carboxyl group to carry drug payloads, peptides, or proteins. While low degrees of substitution do not cause noticeable mechanical changes, high degrees of substitution can cause significant losses to alginate properties including complete loss of calcium cross-linking. While most modifications used to decorate alginate deplete the carboxyl groups, we propose that alginate modifications that replenish the carboxyl groups could overcome the loss in gel integrity and mechanics. In this report, we demonstrate that restoring carboxyl groups during functionalization maintains calcium cross-links as well as hydrogel shear-thinning and self-healing properties. In addition, we demonstrate that alginate hydrogels modified to a high degree with azide modifications that restore the carboxyl groups have improved tissue retention at intramuscular injection sites and capture blood-circulating cyclooctynes better than alginate hydrogels modified with azide modifications that deplete the carboxyl groups. Taken together, alginate modifications that restore carboxyl groups could significantly improve alginate hydrogel mechanics for clinical applications. STATEMENT OF SIGNIFICANCE: Chemical modification of hydrogels provides a powerful tool to regulate cellular adhesion, immune response, and biocompatibility with local tissues. Alginate, due to its biocompatibility and easy chemical modification, is being explored for tissue engineering and drug delivery. Unfortunately, modifying alginate to a high degree of substitution consumes carboxyl group, which are necessary for ionic gelation, leading to poor hydrogel crosslinking. We introduce alginate modifications that restore the alginate's carboxyl groups. We demonstrate that modifications that reintroduce carboxyl groups restore gelation and improve gel mechanics and tissue retention. In addition to contributing to a basic science understanding of hydrogel properties, we anticipate our approach will be useful to create tissue engineered scaffolds and drug delivery platforms.
Collapse
Affiliation(s)
- C T Moody
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University at Raleigh, NC United States of America; Comparative Medicine Institute, North Carolina State University, Raleigh, NC United States of America
| | - A E Brown
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University at Raleigh, NC United States of America
| | - N P Massaro
- Department of Chemistry, North Carolina State University, Raleigh, NC United States of America; Comparative Medicine Institute, North Carolina State University, Raleigh, NC United States of America
| | - A S Patel
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC United States of America
| | - P A Agarwalla
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University at Raleigh, NC United States of America; Comparative Medicine Institute, North Carolina State University, Raleigh, NC United States of America
| | - A M Simpson
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University at Raleigh, NC United States of America
| | - A C Brown
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University at Raleigh, NC United States of America; Comparative Medicine Institute, North Carolina State University, Raleigh, NC United States of America
| | - H Zheng
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC United States of America
| | - J G Pierce
- Department of Chemistry, North Carolina State University, Raleigh, NC United States of America; Comparative Medicine Institute, North Carolina State University, Raleigh, NC United States of America
| | - Y Brudno
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University at Raleigh, NC United States of America; Department of Chemistry, North Carolina State University, Raleigh, NC United States of America; Comparative Medicine Institute, North Carolina State University, Raleigh, NC United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC United States.
| |
Collapse
|
4
|
Abstract
Stimuli-responsive, on-demand release of drugs from drug-eluting depots could transform the treatment of many local diseases, providing intricate control over local dosing. However, conventional on-demand drug release approaches rely on locally implanted drug depots, which become spent over time and cannot be refilled or reused without invasive procedures. New strategies to noninvasively refill drug-eluting depots followed by on-demand release could transform clinical therapy. Here we report an on-demand drug delivery paradigm that combines bioorthogonal click chemistry to locally enrich protodrugs at a prelabeled site and light-triggered drug release at the target tissue. This approach begins with introduction of the targetable depot through local injection of chemically reactive azide groups that anchor to the extracellular matrix. The anchored azide groups then capture blood-circulating protodrugs through bioorthogonal click chemistry. After local capture and retention, active drugs can be released through external light irradiation. In this report, a photoresponsive protodrug was constructed consisting of the chemotherapeutic doxorubicin (Dox), conjugated to dibenzocyclooctyne (DBCO) through a photocleavable ortho-nitrobenzyl linker. The protodrug exhibited excellent on-demand light-triggered Dox release properties and light-mediated in vitro cytotoxicity in U87 glioblastoma cell lines. Furthermore, in a live animal setting, azide depots formed in mice through intradermal injection of activated azide-NHS esters. After i.v. administration, the protodrug was captured by the azide depots with intricate local specificity, which could be increased with multiple refills. Finally, doxorubicin could be released from the depot upon light irradiation. Multiple rounds of depot refilling and light-mediated release of active drug were accomplished, indicating that this system has the potential for multiple rounds of treatment. Taken together, these in vitro and in vivo proof of concept studies establish a novel method for in vivo targeting and on-demand delivery of cytotoxic drugs at target tissues.
Collapse
Affiliation(s)
- Sandeep Palvai
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Christopher T Moody
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Sharda Pandit
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, North Carolina 27607, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
5
|
Pharmacokinetics of Single Domain Antibodies and Conjugated Nanoparticles Using a Hybrid near Infrared Method. Int J Mol Sci 2021; 22:ijms22168695. [PMID: 34445399 PMCID: PMC8395466 DOI: 10.3390/ijms22168695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
Iron oxide nanoparticles and single domain antibodies from camelids (VHHs) have been increasingly recognized for their potential uses for medical diagnosis and treatment. However, there have been relatively few detailed characterizations of their pharmacokinetics (PK). The aim of this study was to develop imaging methods and pharmacokinetic models to aid the future development of a novel family of brain MRI molecular contrast agents. An efficient near-infrared (NIR) imaging method was established to monitor VHH and VHH conjugated nanoparticle kinetics in mice using a hybrid approach: kinetics in blood were assessed by direct sampling, and kinetics in kidney, liver, and brain were assessed by serial in vivo NIR imaging. These studies were performed under "basal" circumstances in which the VHH constructs and VHH-conjugated nanoparticles do not substantially interact with targets nor cross the blood brain barrier. Using this approach, we constructed a five-compartment PK model that fits the data well for single VHHs, engineered VHH trimers, and iron oxide nanoparticles conjugated to VHH trimers. The establishment of the feasibility of these methods lays a foundation for future PK studies of candidate brain MRI molecular contrast agents.
Collapse
|
6
|
Welling MM, Duszenko N, van Willigen DM, Hensbergen AW, Buckle T, Rietbergen DDD, Roestenberg M, van Leeuwen FWB. Interventional nuclear medicine: "click" chemistry as an in vivo targeting strategy for imaging microspheres and bacteria. Biomater Sci 2021; 9:1683-1690. [PMID: 33410436 DOI: 10.1039/d0bm01823f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM Pre-targeting is a proven strategy for in vivo delivery of a diagnostic or therapeutic payload. The pre-targeting concept can be realized through various conjugation strategies, one of which is based on copper-free "click" chemistry. Copper-free click reactions have shown in vivo potential for imaging and radionuclide therapy, but this conjugation strategy has not yet been explored in combination with microspheres or unicellular organisms. This study aims to evaluate the in vivo efficacy of strain-promoted azide-alkyne cycloaddition (SPAAC) reactions to achieve imaging and targeting of azide-functionalized macro-aggregated albumin (MAA) microspheres and Staphylococcus aureus bacteria. METHODS MAA microspheres (diameter 10-90 μm) were functionalized with a biorthogonal Cy5 fluorophore, bearing an azide functionality (N3), to generate MAA-Cy5-N3. S. aureus (diameter ∼1 μm) were functionalized with 99mTc-UBI29-41-Cy5-N3, generating S. aureus-99mTc-UBI29-41-Cy5-N3. In situ and in vitro click conjugation on the -N3 moieties was studied for 20 h using a radioactivity-based assay and fluorescence microscopy. For in vivo validation, both primary entities, radiolabeled with 99mTc, were deposited into the microvasculature of the liver via intrasplenic injections. Secondary targeting was realized following the intravenous administration of indium-111-radiolabeled diethylenetriaminepentaacetic acid-dibenzocyclooctyne (111In-DTPA-DBCO). To assess click reaction efficiency in vivo, 99mTc and 111In-biodistributions were measured (SPECT and %ID g-1). Use of 111In-DTPA-DBCO in mice without MAA deposits or mice infected with non-functionalized S. aureus served as controls. Ex vivo confocal fluorescence imaging was carried out in excised tissues to confirm the presence of functionalized MAA and bacteria. RESULTS In vitro data confirmed effective click reactions on both the MAA particles and the bacterial membrane. SPECT imaging and biodistribution studies revealed significantly (p < 0.05) increased accumulation of 111In-DTPA-DBCO at the sites where MAA-Cy5-N3 (7.5 ± 1.5%ID g-1vs. 3.5 ± 0.5%ID g-1 in control mice) and S. aureus-99mTc-UBI29-41-Cy5-N3 (9.3 ± 1.3%ID g-1vs. 6.0 ± 0.5%ID g-1 in control mice) resided. Ex vivo fluorescence imaging confirmed the presence of either functionalized MAA or S. aureus in excised spleens and livers of mice. CONCLUSION Copper-free click chemistry between a DBCO moiety and Cy5-N3-functionalized microspheres or bacterial entities in the liver can be used to realize in vivo imaging and targeting.
Collapse
Affiliation(s)
- M M Welling
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands.
| | - N Duszenko
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands. and Departments of Parasitology and Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - D M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands.
| | - A W Hensbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands.
| | - T Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands.
| | - D D D Rietbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands. and Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - M Roestenberg
- Departments of Parasitology and Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - F W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, Netherlands.
| |
Collapse
|
7
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
8
|
Porte K, Riberaud M, Châtre R, Audisio D, Papot S, Taran F. Bioorthogonal Reactions in Animals. Chembiochem 2020; 22:100-113. [PMID: 32935888 DOI: 10.1002/cbic.202000525] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/15/2020] [Indexed: 01/04/2023]
Abstract
The advent of bioorthogonal chemistry has led to the development of powerful chemical tools that enable increasingly ambitious applications. In particular, these tools have made it possible to achieve what is considered to be the holy grail of many researchers involved in chemical biology: to perform unnatural chemical reactions within living organisms. In this minireview, we present an update of bioorthogonal reactions that have been carried out in animals for various applications. We outline the advances made in the understanding of fundamental biological processes, and the development of innovative imaging and therapeutic strategies using bioorthogonal chemistry.
Collapse
Affiliation(s)
- Karine Porte
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| | - Maxime Riberaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| | - Rémi Châtre
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), 86022, Poitiers, France) E-mail
| | - Davide Audisio
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| | - Sébastien Papot
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), 86022, Poitiers, France) E-mail
| | - Frédéric Taran
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| |
Collapse
|
9
|
Palvai S, Bhangu J, Akgun B, Moody CT, Hall DG, Brudno Y. In Vivo Targeting Using Arylboronate/Nopoldiol Click Conjugation. Bioconjug Chem 2020; 31:2288-2292. [PMID: 32960584 DOI: 10.1021/acs.bioconjchem.0c00453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bioorthogonal click reactions yielding stable and irreversible adducts are in high demand for in vivo applications, including in biomolecular labeling, diagnostic imaging, and drug delivery. Previously, we reported a novel bioorthogonal "click" reaction based on the coupling of ortho-acetyl arylboronates and thiosemicarbazide-functionalized nopoldiol. We now report that a detailed structural analysis of the arylboronate/nopoldiol adduct by X-ray crystallography and 11B NMR reveals that the bioorthogonal reactants form, unexpectedly, a tetracyclic adduct through the cyclization of the distal nitrogen into the semithiocarbazone leading to a strong B-N dative bond and two new 5-membered rings. The cyclization adduct, which protects the boronate unit against hydrolytic breakdown, sheds light on the irreversible nature of this polycondensation. The potential of this reaction to work in a live animal setting was studied through in vivo capture of fluorescently labeled molecules in vivo. Arylboronates were introduced into tissues through intradermal injection of their activated NHS esters, which react with amines in the extracellular matrix. Fluorescently labeled nopoldiol molecules were administered systemically and were efficiently captured by the arylboronic acids in a location-specific manner. Taken together, these in vivo proof-of-concept studies establish arylboronate/nopoldiol bioorthogonal chemistry as a candidate for wide array of applications in chemical biology and drug delivery.
Collapse
Affiliation(s)
- Sandeep Palvai
- Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill and North Carolina State University Raleigh, 1840 Entrepreneur Drive, Raleigh, North Carolina 27695, United States
| | - Jasmine Bhangu
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Burcin Akgun
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Christopher T Moody
- Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill and North Carolina State University Raleigh, 1840 Entrepreneur Drive, Raleigh, North Carolina 27695, United States
| | - Dennis G Hall
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill and North Carolina State University Raleigh, 1840 Entrepreneur Drive, Raleigh, North Carolina 27695, United States
| |
Collapse
|
10
|
Moody CT, Palvai S, Brudno Y. Click cross-linking improves retention and targeting of refillable alginate depots. Acta Biomater 2020; 112:112-121. [PMID: 32497743 PMCID: PMC7365769 DOI: 10.1016/j.actbio.2020.05.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Injectable alginate hydrogels have demonstrated utility in tissue engineering and drug delivery applications due in part to their mild gelation conditions, low host responses and chemical versatility. Recently, the potential of these gels has expanded with the introduction of refillable hydrogel depots - alginate gels chemically decorated with click chemistry groups to efficiently capture prodrug refills from the blood. Unfortunately, high degrees of click group substitution on alginate lead to poor viscoelastic properties and loss of ionic cross-linking. In this work, we introduce tetrabicyclononyne (tBCN) agents that covalently cross-link azide-modified alginate hydrogels for tissue engineering and drug delivery application in vivo. Adjusting cross-linker concentration allowed tuning the hydrogel mechanical properties for tissue-specific mechanical strength. The bioorthogonal and specific click reaction creates stable hydrogels with improved in vivo properties, including improved retention at injected sites. Azide-alginate hydrogels cross-linked with tBCN elicited minimal inflammation and maintained structural integrity over several months and efficiently captured therapeutics drug surrogates from the circulation. Taken together, azide-alginate hydrogels cross-linked with tBCN convey the benefits of alginate hydrogels for use in tissue engineering and drug delivery applications of refillable drug delivery depots. STATEMENT OF SIGNIFICANCE: Ionically cross-linked, injectable alginate biomaterials hold promise in many different clinical settings. However, adding new chemical functionality to alginate can disrupt their ionic cross-linking, limiting their utility. We have developed a "click" cross-linking strategy to improve the mechanical properties and tissue function of modified alginate biomaterials and enable them to capture small molecule drugs from the blood. We show that click cross-linked materials remain in place better than ionically cross-linked materials and efficiently capture payloads from the blood. Development of click cross-linking for refillable depots represents a crucial step toward clinical application of this promising drug delivery platform.
Collapse
Affiliation(s)
- Christopher T Moody
- Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill and North Carolina State University - Raleigh, 1840 Entrepreneur Drive, Raleigh, NC 27695, USA
| | - Sandeep Palvai
- Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill and North Carolina State University - Raleigh, 1840 Entrepreneur Drive, Raleigh, NC 27695, USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina - Chapel Hill and North Carolina State University - Raleigh, 1840 Entrepreneur Drive, Raleigh, NC 27695, USA.
| |
Collapse
|
11
|
He W, Reaume M, Hennenfent M, Lee BP, Rajachar R. Biomimetic hydrogels with spatial- and temporal-controlled chemical cues for tissue engineering. Biomater Sci 2020; 8:3248-3269. [PMID: 32490441 PMCID: PMC7323904 DOI: 10.1039/d0bm00263a] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biomimetic hydrogels have emerged as the most useful tissue engineering scaffold materials. Their versatile chemistry can recapitulate multiple physical and chemical features to integrate cells, scaffolds, and signaling molecules for tissue regeneration. Due to their highly hydrophilic nature hydrogels can recreate nutrient-rich aqueous environments for cells. Soluble regulatory molecules can be incorporated to guide cell proliferation and differentiation. Importantly, the controlled dynamic parameters and spatial distribution of chemical cues in hydrogel scaffolds are critical for cell-cell communication, cell-scaffold interaction, and morphogenesis. Herein, we review biomimetic hydrogels that provide cells with spatiotemporally controlled chemical cues as tissue engineering scaffolds. Specifically, hydrogels with temporally controlled growth factor-release abilities, spatially controlled conjugated bioactive molecules/motifs, and targeting delivery and reload properties for tissue engineering applications are discussed in detail. Examples of hydrogels that possess clinically favorable properties, such as injectability, self-healing ability, stimulus-responsiveness, and pro-remodeling features, are also covered.
Collapse
Affiliation(s)
- Weilue He
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
- FM Wound Care, LLC, Hancock, MI 49930, USA
| | - Max Reaume
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Maureen Hennenfent
- Department of Civil and Environmental Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Bruce P. Lee
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Rupak Rajachar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
12
|
Shankaraiah N, Sakla AP, Laxmikeshav K, Tokala R. Reliability of Click Chemistry on Drug Discovery: A Personal Account. CHEM REC 2020; 20:253-272. [DOI: 10.1002/tcr.201900027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/08/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Nagula Shankaraiah
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Akash P. Sakla
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Kritika Laxmikeshav
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Ramya Tokala
- Department of Medicinal ChemistryNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| |
Collapse
|
13
|
Brambila CJ, Lux J, Mattrey RF, Boyd D, Borden MA, de Gracia Lux C. Bubble Inflation Using Phase-Change Perfluorocarbon Nanodroplets as a Strategy for Enhanced Ultrasound Imaging and Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:2954-2965. [PMID: 32090572 DOI: 10.1021/acs.langmuir.9b03647] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Phase-change perfluorocarbon microdroplets were introduced over 2 decades ago to occlude downstream vessels in vivo. Interest in perfluorocarbon nanodroplets has recently increased to enable extravascular targeting, to rescue the weak ultrasound signal of perfluorocarbon droplets by converting them to microbubbles and to improve ultrasound-based therapy. Despite great scientific interest and advances, applications of phase-change perfluorocarbon agents have not reached clinical testing because of efficacy and safety concerns, some of which remain unexplained. Here, we report that the coexistence of perfluorocarbon droplets and microbubbles in blood, which is inevitable when droplets spontaneously or intentionally vaporize to form microbubbles, is a major contributor to the observed side effects. We develop the theory to explain why the coexistence of droplets and microbubbles results in microbubble inflation induced by perfluorocarbon transfer from droplets to adjacent microbubbles. We also present the experimental data showing up to 6 orders of magnitude microbubble volume expansion, which occludes a 200 μm tubing in the presence of perfluorocarbon nanodroplets. More importantly, we demonstrate that the rate of microbubble inflation and ultimate size can be controlled by manipulating formulation parameters to tailor the agent's design for the potential theranostic application while minimizing the risk to benefit ratio.
Collapse
Affiliation(s)
- Carlos J Brambila
- Translational Research in Ultrasound Theranostics (TRUST) Program, Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Jacques Lux
- Translational Research in Ultrasound Theranostics (TRUST) Program, Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Organic Chemistry Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Robert F Mattrey
- Translational Research in Ultrasound Theranostics (TRUST) Program, Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Dustin Boyd
- Translational Research in Ultrasound Theranostics (TRUST) Program, Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Mark A Borden
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado 80309, United States
| | - Caroline de Gracia Lux
- Translational Research in Ultrasound Theranostics (TRUST) Program, Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
14
|
McNamara SL, Brudno Y, Miller AB, Ham HO, Aizenberg M, Chaikof EL, Mooney DJ. Regenerating Antithrombotic Surfaces through Nucleic Acid Displacement. ACS Biomater Sci Eng 2020; 6:2159-2166. [PMID: 33455325 DOI: 10.1021/acsbiomaterials.0c00038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Blood-contacting devices are commonly coated with antithrombotic agents to prevent clot formation and to extend the lifespan of the device. However, in vivo degradation of these bioactive surface agents ultimately limits device efficacy and longevity. Here, a regenerative antithrombotic catheter surface treatment is developed using oligodeoxynucleotide (ODN) toehold exchange. ODN strands modified to carry antithrombotic payloads can inhibit the thrombin enzyme when bound to a surface and exchange with rapid kinetics over multiple cycles, even while carrying large payloads. The surface-bound ODNs inhibit thrombin activity to significantly reduce fibrinogen cleavage and fibrin formation, and this effect is sustained after ODN exchange of the surface-bound strands with a fresh antithrombotic payload. This study presents a unique strategy for achieving a continuous antithrombotic state for blood-contacting devices using an ODN-based regeneration method.
Collapse
Affiliation(s)
- Stephanie L McNamara
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02138, United States.,Harvard-MIT Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02138, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, 911 Oval Drive, Raleigh, North Carolina 27695, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| | - Alex B Miller
- Harvard-MIT Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02138, United States
| | - Hyun Oki Ham
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States.,Department of Surgery, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Michael Aizenberg
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| | - Elliot L Chaikof
- Harvard-MIT Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02138, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States.,Department of Surgery, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02138, United States.,Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, Massachusetts 02215, United States
| |
Collapse
|
15
|
Adams MR, Moody CT, Sollinger JL, Brudno Y. Extracellular-Matrix-Anchored Click Motifs for Specific Tissue Targeting. Mol Pharm 2020; 17:392-403. [PMID: 31829613 DOI: 10.1021/acs.molpharmaceut.9b00589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Local presentation of cancer drugs by injectable drug-eluting depots reduces systemic side effects and improves efficacy. However, local depots deplete their drug stores and are difficult to introduce into stiff tissues, or organs, such as the brain, that cannot accommodate increased pressure. We present a method for introducing targetable depots through injection of activated ester molecules into target tissues that react with and anchor themselves to the local extracellular matrix (ECM) and subsequently capture systemically administered small molecules through bioorthogonal click chemistry. A computational model of tissue-anchoring depot formation and distribution was verified by histological analysis and confocal imaging of cleared tissues. ECM-anchored click groups do not elicit any noticeable local or systemic toxicity or immune response and specifically capture systemically circulating molecules at intradermal, intratumoral, and intracranial sites for multiple months. Taken together, ECM anchoring of click chemistry motifs is a promising approach to specific targeting of both small and large therapeutics, enabling repeated local presentation for cancer therapy and other diseases.
Collapse
Affiliation(s)
- Mary R Adams
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Christopher T Moody
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Jennifer L Sollinger
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States.,Lineberger Comprehensive Cancer Center , University of North Carolina, Chapel Hill , 450 West Dr. , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
16
|
El Malah T, Abdel Mageid RE, Awad HM, Nour HF. Copper( i)-catalysed azide–alkyne cycloaddition and antiproliferative activity of mono- and bis-1,2,3-triazole derivatives. NEW J CHEM 2020. [DOI: 10.1039/d0nj04308g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A series of mono- and bis-1,2,3-triazole derivatives were prepared via the copper(i)-catalysed azide–alkyne cycloaddition between substituted aromatic derivatives, comprising one or two terminal alkyne groups and a selection of aromatic azides.
Collapse
Affiliation(s)
- Tamer El Malah
- Photochemistry Department
- Chemical Industries Research Division
- National Research Centre
- Cairo
- Egypt
| | - Randa E. Abdel Mageid
- Photochemistry Department
- Chemical Industries Research Division
- National Research Centre
- Cairo
- Egypt
| | - Hanem M. Awad
- Department of Tanning Materials and Leather Technology
- National Research Centre
- Cairo
- Egypt
| | - Hany F. Nour
- Photochemistry Department
- Chemical Industries Research Division
- National Research Centre
- Cairo
- Egypt
| |
Collapse
|
17
|
Wang H, Sobral MC, Snyder T, Brudno Y, Gorantla VS, Mooney DJ. Clickable, acid labile immunosuppressive prodrugs forin vivotargeting. Biomater Sci 2020; 8:266-277. [DOI: 10.1039/c9bm01487j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clickable immunosuppressive prodrugs enablein vivoreplenishment of drugs in biomaterial depots to maintain long-term immunosuppression in tissue/organ transplantation.
Collapse
Affiliation(s)
- Hua Wang
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| | - Miguel C. Sobral
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| | - Tracy Snyder
- Wyss Institute for Biologically Inspired Engineering
- Cambridge
- USA
| | - Yevgeny Brudno
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| | - Vijay S. Gorantla
- Surgery
- Ophthalmology and Bioengineering
- Wake Forest School of Medicine
- Winston-Salem
- USA
| | - David J. Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences
- Harvard University
- Cambridge
- Massachusetts 02138
- USA
| |
Collapse
|
18
|
Xiao Y, Liu Q, Clulow AJ, Li T, Manohar M, Gilbert EP, de Campo L, Hawley A, Boyd BJ. PEGylation and surface functionalization of liposomes containing drug nanocrystals for cell-targeted delivery. Colloids Surf B Biointerfaces 2019; 182:110362. [PMID: 31351271 DOI: 10.1016/j.colsurfb.2019.110362] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 07/04/2019] [Accepted: 07/13/2019] [Indexed: 01/09/2023]
Abstract
Liposomal formulations have important therapeutic applications in anti-cancer treatments but current formulations suffer from serious side effects, high dosage requirements and prolonged treatment. In this study, PEGylated azide-functionalized liposomes containing drug nanocrystals were investigated with the aim of increasing the drug payload and achieving functionalization for targeted delivery. Liposomes were characterized using cryogenic transmission electron microscopy (cryo-TEM), dynamic light scattering (DLS), small and ultra-small angle neutron scattering (SANS/USANS) and small and wide angle X-ray scattering (SAXS/WAXS). Cryo-TEM experiments revealed the dimensions of the nanocrystal-loaded liposomes and the change of shape from spherical to elongated after the formation of nanocrystals. Results from SANS/USANS experiments confirmed the asymmetric particle shape. SAXS/WAXS experiments confirmed that the crystalline drug only occurred in freeze-thawed samples and correlated with a new unidentified polymorphic form of ciprofloxacin. Using a small molecule dye, dibenzocyclooctyne (DBCO)-cy5, specific conjugation between DBCO groups and surface azide groups on the liposomes was confirmed; this indicates the promise of this system for tumour-targeted delivery.
Collapse
Affiliation(s)
- Yunxin Xiao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University Parkville Campus, Australia
| | - Qingtao Liu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University Parkville Campus, Australia
| | - Andrew J Clulow
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Tang Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University Parkville Campus, Australia
| | - Madhura Manohar
- National Deuteration Facility (NDF), Australian Nuclear Science and Technology Organisation (ANSTO), Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Elliot P Gilbert
- Australian Centre for Neutron Scattering (ACNS), Australian Nuclear Science and Technology Organisation (ANSTO), Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Liliana de Campo
- Australian Centre for Neutron Scattering (ACNS), Australian Nuclear Science and Technology Organisation (ANSTO), Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Adrian Hawley
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation (ANSTO), 800 Blackburn Rd, Clayton, VIC, 3168, Australia
| | - Ben J Boyd
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University Parkville Campus, Australia.
| |
Collapse
|
19
|
Brudno Y, Pezone MJ, Snyder TK, Uzun O, Moody CT, Aizenberg M, Mooney DJ. Replenishable drug depot to combat post-resection cancer recurrence. Biomaterials 2018; 178:373-382. [PMID: 29779862 PMCID: PMC6075722 DOI: 10.1016/j.biomaterials.2018.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 01/05/2023]
Abstract
Local drug presentation made possible by drug-eluting depots has demonstrated benefits in a vast array of diseases, including in cancer, microbial infection and in wound healing. However, locally-eluting depots are single-use systems that cannot be refilled or reused after implantation at inaccessible sites, limiting their clinical utility. New strategies to noninvasively refill drug-eluting depots could dramatically enhance their clinical use. In this report we present a refillable hydrogel depot system based on bioorthogonal click chemistry. The click-modified hydrogel depots capture prodrug refills from the blood and subsequently release active drugs locally in a sustained manner. Capture of the systemically-administered refills serves as an efficient and non-toxic method to repeatedly refill depots. Refillable depots in combination with prodrug refills achieve sustained release at precancerous tumor sites to improve cancer therapy while eliminating systemic side effects. The ability to target tissues without enhanced permeability could allow the use of refillable depots in cancer and many other medical applications.
Collapse
Affiliation(s)
- Yevgeny Brudno
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA; School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138, USA; Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, 911 Oval Drive, Raleigh, NC 27695, USA; Lineberger Comprehensive Cancer Center, University of North Carolina - Chapel Hill, 450 West Dr, Chapel Hill, NC 27599, USA
| | - Matthew J Pezone
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Tracy K Snyder
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Oktay Uzun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Christopher T Moody
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, 911 Oval Drive, Raleigh, NC 27695, USA
| | - Michael Aizenberg
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115, USA; School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138, USA.
| |
Collapse
|
20
|
Madl CM, Heilshorn SC. Bioorthogonal Strategies for Engineering Extracellular Matrices. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1706046. [PMID: 31558890 PMCID: PMC6761700 DOI: 10.1002/adfm.201706046] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Hydrogels are commonly used as engineered extracellular matrix (ECM) mimics in applications ranging from tissue engineering to in vitro disease models. Ideal mechanisms used to crosslink ECM-mimicking hydrogels do not interfere with the biology of the system. However, most common hydrogel crosslinking chemistries exhibit some form of cross-reactivity. The field of bio-orthogonal chemistry has arisen to address the need for highly specific and robust reactions in biological contexts. Accordingly, bio-orthogonal crosslinking strategies have been incorporated into hydrogel design, allowing for gentle and efficient encapsulation of cells in various hydrogel materials. Furthermore, the selective nature of bio-orthogonal chemistries can permit dynamic modification of hydrogel materials in the presence of live cells and other biomolecules to alter matrix mechanical properties and biochemistry on demand. In this review, we provide an overview of bio-orthogonal strategies used to prepare cell-encapsulating hydrogels and highlight the potential applications of bio-orthogonal chemistries in the design of dynamic engineered ECMs.
Collapse
Affiliation(s)
- Christopher M Madl
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA,
| |
Collapse
|
21
|
Abstract
Hydrogel delivery systems can leverage therapeutically beneficial outcomes of drug delivery and have found clinical use. Hydrogels can provide spatial and temporal control over the release of various therapeutic agents, including small-molecule drugs, macromolecular drugs and cells. Owing to their tunable physical properties, controllable degradability and capability to protect labile drugs from degradation, hydrogels serve as a platform in which various physiochemical interactions with the encapsulated drugs control their release. In this Review, we cover multiscale mechanisms underlying the design of hydrogel drug delivery systems, focusing on physical and chemical properties of the hydrogel network and the hydrogel-drug interactions across the network, mesh, and molecular (or atomistic) scales. We discuss how different mechanisms interact and can be integrated to exert fine control in time and space over the drug presentation. We also collect experimental release data from the literature, review clinical translation to date of these systems, and present quantitative comparisons between different systems to provide guidelines for the rational design of hydrogel delivery systems.
Collapse
Affiliation(s)
- Jianyu Li
- John A. Paulson School of Engineering and Applied Sciences, and the Wyss Institute for biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, and the Wyss Institute for biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
22
|
Mejia Oneto JM, Khan I, Seebald L, Royzen M. In Vivo Bioorthogonal Chemistry Enables Local Hydrogel and Systemic Pro-Drug To Treat Soft Tissue Sarcoma. ACS CENTRAL SCIENCE 2016; 2:476-82. [PMID: 27504494 PMCID: PMC4965853 DOI: 10.1021/acscentsci.6b00150] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Indexed: 05/18/2023]
Abstract
The ability to activate drugs only at desired locations avoiding systemic immunosuppression and other dose limiting toxicities is highly desirable. Here we present a new approach, named local drug activation, that uses bioorthogonal chemistry to concentrate and activate systemic small molecules at a location of choice. This method is independent of endogenous cellular or environmental markers and only depends on the presence of a preimplanted biomaterial near a desired site (e.g., tumor). We demonstrate the clear therapeutic benefit with minimal side effects of this approach in mice over systemic therapy using a doxorubicin pro-drug against xenograft tumors of a type of soft tissue sarcoma (HT1080).
Collapse
Affiliation(s)
- Jose M. Mejia Oneto
- Shasqi Inc., 665 Third Street, Suite 250, San Francisco, California 94107, United States
- E-mail:
| | - Irfan Khan
- University
at Albany, Department of Chemistry, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Leah Seebald
- University
at Albany, Department of Chemistry, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Maksim Royzen
- University
at Albany, Department of Chemistry, 1400 Washington Avenue, Albany, New York 12222, United States
| |
Collapse
|
23
|
Madl CM, Katz LM, Heilshorn SC. Bio-Orthogonally Crosslinked, Engineered Protein Hydrogels with Tunable Mechanics and Biochemistry for Cell Encapsulation. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3612-3620. [PMID: 27642274 PMCID: PMC5019573 DOI: 10.1002/adfm.201505329] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Covalently-crosslinked hydrogels are commonly used as 3D matrices for cell culture and transplantation. However, the crosslinking chemistries used to prepare these gels generally cross-react with functional groups present on the cell surface, potentially leading to cytotoxicity and other undesired effects. Bio-orthogonal chemistries have been developed that do not react with biologically relevant functional groups, thereby preventing these undesirable side reactions. However, previously developed biomaterials using these chemistries still possess less than ideal properties for cell encapsulation, such as slow gelation kinetics and limited tuning of matrix mechanics and biochemistry. Here, engineered elastin-like proteins (ELPs) are developed that cross-link via strain-promoted azide-alkyne cycloaddition (SPAAC) or Staudinger ligation. The SPAAC-crosslinked materials form gels within seconds and complete gelation within minutes. These hydrogels support the encapsulation and phenotypic maintenance of human mesenchymal stem cells, human umbilical vein endothelial cells, and murine neural progenitor cells. SPAAC-ELP gels exhibit independent tuning of stiffness and cell adhesion, with significantly improved cell viability and spreading observed in materials containing a fibronectin-derived arginine-glycine-aspartic acid (RGD) domain. The crosslinking chemistry used permits further material functionalization, even in the presence of cells and serum. These hydrogels are anticipated to be useful in a wide range of applications, including therapeutic cell delivery and bioprinting.
Collapse
Affiliation(s)
| | - Lily M. Katz
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Stejskalová A, Kiani MT, Almquist BD. Programmable biomaterials for dynamic and responsive drug delivery. Exp Biol Med (Maywood) 2016; 241:1127-37. [PMID: 27190245 PMCID: PMC4950367 DOI: 10.1177/1535370216649445] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Biomaterials are continually being designed that enable new methods for interacting dynamically with cell and tissues, in turn unlocking new capabilities in areas ranging from drug delivery to regenerative medicine. In this review, we explore some of the recent advances being made in regards to programming biomaterials for improved drug delivery, with a focus on cancer and infection. We begin by explaining several of the underlying concepts that are being used to design this new wave of drug delivery vehicles, followed by examining recent materials systems that are able to coordinate the temporal delivery of multiple therapeutics, dynamically respond to changing tissue environments, and reprogram their bioactivity over time.
Collapse
Affiliation(s)
- Anna Stejskalová
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Mehrdad T Kiani
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | | |
Collapse
|
25
|
On-demand drug delivery from local depots. J Control Release 2015; 219:8-17. [PMID: 26374941 DOI: 10.1016/j.jconrel.2015.09.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/08/2015] [Accepted: 09/08/2015] [Indexed: 11/22/2022]
Abstract
Stimuli-responsive polymeric depots capable of on-demand release of therapeutics promise a substantial improvement in the treatment of many local diseases. These systems have the advantage of controlling local dosing so that payload is released at a time and with a dose chosen by a physician or patient, and the dose can be varied as disease progresses or healing occurs. Macroscale drug depot can be induced to release therapeutics through the action of physical stimuli such as ultrasound, electric and magnetic fields and light as well as through the addition of pharmacological stimuli such as nucleic acids and small molecules. In this review, we highlight recent advances in the development of polymeric systems engineered for releasing therapeutic molecules through physical and pharmacological stimulation.
Collapse
|