Matching Items (52)
Description

Aminoglycosides contain a basic unit of an amino-modified glycoside (sugar) and have potent antibacterial properties used to treat a wide range of bacterial infections, including those that occur in the soft tissue, chest, urinary tract, and endocardial tissue.1, 2 With a broad spectrum of activity and convenient dosing schedule, Aminoglycoside

Aminoglycosides contain a basic unit of an amino-modified glycoside (sugar) and have potent antibacterial properties used to treat a wide range of bacterial infections, including those that occur in the soft tissue, chest, urinary tract, and endocardial tissue.1, 2 With a broad spectrum of activity and convenient dosing schedule, Aminoglycoside helps to effectively fight off Gram-negative bacteria.1, 3 In 1944 an aminoglycoside called streptomycin entered clinical trials to test its effectiveness as an antibiotic.4 After several years other classes of aminoglycosides were discovered such as neomycin, gentamicin, kanamycin, and netilmicin.4 When introduced these antibiotics presented major clinical advancements in the treatment of Tuberculosis and other bacterial infections.3, 4 However their use in modern medicine has diminished due to their toxicity, required parenteral delivery, and the availability of alternative antibiotics.3, 5 The dose-dependent toxicity of aminoglycosides limits their use due to a narrow range of safe aminoglycoside plasma concentrations.3, 5 Exceeding this range in non-target tissues can lead to negative effects on the audio-vestibular apparatus and kidneys.3, 5, 6 In the 1980’s, clinicians began treating infections with antibiotics that were perceived as less toxic and providing broader antibacterial activity.7 This resulted in aminoglycosides being prescribed for more persistent infections that were resistant to other antibiotics.3 With the amount of antibiotic resistant bacteria increasing, many scientists have begun looking into methods for minimizing aminoglycoside toxicity and maximizing its antibacterial activity.3, 8 These methods include encapsulation and polymer conjugation.9, 10 By encapsulating aminoglycosides in liposomes or other vesicles scientists aim to increase its concentration in infected tissues while decreasing nephro- and ototoxicity.9 With conjugated polymers scientists have created polymer complexes containing aminoglycosides and other compounds such as dopamine.11 The goal of these polymers is to limit toxicity by slowing antibiotic release and increasing efficacy of the antibiotic through targeted delivery and toxicity of other compounds.9, 10, 11 Other than its use in treating infections, aminoglycosides are gaining attention as an excellent vehicle for gene delivery.12 In this application aminoglycosides are used to correct a genetic defect by introducing a normal copy of the gene into affected cells.12,13 Currently scientists are assessing aminoglycosides for gene therapy in the treatment of cancer and various other diseases.12, 14 This review will focus on the diverse customizability of aminoglycosides in treating infections and as vehicles for gene therapy.

ContributorsWampler, Cole (Author) / Rege, Kaushal (Thesis director) / Schoepf, Jared (Committee member) / Barrett, The Honors College (Contributor) / Dean, W.P. Carey School of Business (Contributor) / Chemical Engineering Program (Contributor) / School of Life Sciences (Contributor)
Created2023-05
168478-Thumbnail Image.png
Description
Sutures, staples, and tissue glues remain the primary means of tissue approximation and vessel ligation. Laser-activated tissue sealing is an alternative approach that conventionally employs light-absorbing chromophores and nanoparticles for converting near-infrared (NIR) laser to heat. The local increase in temperature engenders interdigitation of sealant and tissue biomolecules, resulting in

Sutures, staples, and tissue glues remain the primary means of tissue approximation and vessel ligation. Laser-activated tissue sealing is an alternative approach that conventionally employs light-absorbing chromophores and nanoparticles for converting near-infrared (NIR) laser to heat. The local increase in temperature engenders interdigitation of sealant and tissue biomolecules, resulting in rapid tissue sealing. Light-activated sealants (LASE) were developed in which indocyanine green (ICG) dye is embedded within a biopolymer matrix (silk or chitosan) for incisional defect repair. Light-activated tissue-integrating sutures (LATIS) that synergize the benefits of conventional suturing and laser sealing were also fabricated and demonstrated higher efficacies for tissue biomechanical recovery and repair in a full-thickness, dorsal surgical incision model in mice compared to commercial sutures and cyanoacrylate skin glue. Localized delivery of modulators of tissue repair, including histamine and copper, from LASE and LATIS further improved healed skin strength. In addition to incisional wounds, histamine co-delivered with silk fibroin LASE films accelerated the closure of full thickness, splinted excisional wounds in immunocompetent BALB/c mice and genetically obese and diabetic db/db mice, resulting in faster closure than Tegaderm wound dressing. Immunohistochemistry analyses showed LASE-histamine treatment enhanced wound repair involving mechanisms of neoangiogenesis, myofibroblast activation, transient epidermal EMT, and also improve healed skin biomechanical strength which are hallmarks of improved healing outcomes. Benefit of temporal delivery was further investigated of a second therapeutic (growth factor nanoparticles) in modulating wound healing outcomes in both acute and diabetic wounds. The hypothesis of temporal delivery of second therapeutic around the ‘transition period’ in wounds further improved wound closure kinetics and biomechanical recovery of skin strength. Laser sealing and approximation, together with delivery of immunomodulatory mediators, can lead to faster healing and tissue repair, thus reducing wound dehiscence, preventing wounds moving towards chronicity and lowering incidence of surgical site infections, all of which can have significant impact in the clinic.
ContributorsGhosh, Deepanjan (Author) / Rege, Kaushal (Thesis advisor) / Acharya, Abhinav (Committee member) / Holloway, Julianne (Committee member) / DiCaudo, David (Committee member) / P. Leung, Kai (Committee member) / Arizona State University (Publisher)
Created2021
189267-Thumbnail Image.png
Description
Cellular models have been the backbone of models for drug therapeutics, discovery, or diagnostics, and for modeling a tumor microenvironment to understand the proliferation, migration, invasion, dormancy, angiogenesis, Conventional two-dimensional (2D) cell culture models are used because of the cost-effectiveness compared to animal models. But these models fail to mimic

Cellular models have been the backbone of models for drug therapeutics, discovery, or diagnostics, and for modeling a tumor microenvironment to understand the proliferation, migration, invasion, dormancy, angiogenesis, Conventional two-dimensional (2D) cell culture models are used because of the cost-effectiveness compared to animal models. But these models fail to mimic the cellular phenotype of a three-dimensional (3D) microenvironment. As a result, it is important to develop a 3D model that predicts cellular behaviors and their interaction with neighboring cells and extracellular matrix (ECM) in a more realistic setting. Various 3D cell culture methods have been employed to generate spheroids, in vitro, but most of these platforms face drawbacks such as spheroid size heterogeneity, low yield, use of specialized instruments etc. The hydrogel platform mentioned here was able to solve all the previous problems and can create a novel 3D tumor microenvironment. This thesis is focused on developing an in-vitro 3D model which can modulate the tumor microenvironment consisting of cancer cells and macrophages and how the Amikagel platform modulated the macrophage phenotype is discussed in detail here. This platform can be an ideal platform for macrophage phenotype modulation.
ContributorsChowdhury, Trishita (Author) / Rege, Kaushal (Thesis advisor) / Acharya, Abhinav (Committee member) / Khalifehzadeh, Layla (Committee member) / Arizona State University (Publisher)
Created2023
187385-Thumbnail Image.png
Description
The use of mRNA for therapeutic purposes has gained significant attention due to its potential to treat a wide range of diseases, including cancer, infectious diseases, and genetic disorders. However, the efficient delivery of mRNA to target cells remains a major challenge, and delivery of mRNA faces major issues such

The use of mRNA for therapeutic purposes has gained significant attention due to its potential to treat a wide range of diseases, including cancer, infectious diseases, and genetic disorders. However, the efficient delivery of mRNA to target cells remains a major challenge, and delivery of mRNA faces major issues such as rapid degradation and poor cellular uptake. Aminoglycoside-derived lipopolymer nanoparticles (LPNs) have been shown as a promising platform for plasmid DNA (pDNA) delivery due to their stability, biocompatibility, and ability to encapsulate mRNA. The current study aims to develop and optimize LPNs formulation for the delivery of mRNA in aggressive cancer cells, using a combination of chemical synthesis, physicochemical characterization, and in vitro biological assays. From a small library of aminoglycoside-derived lipopolymers, the lead lipopolymers were screened for the efficient delivery of mRNA. The complexes were synthesized with different ratios of lipopolymers to mRNA. The appropriate binding ratios of lipopolymers and mRNA were determined by gel electrophoresis. The complexes were characterized using dynamic light scattering (DLS) and zeta potential. The transgene expression efficacy of polymers was evaluated using in vitro bioluminescence assay. The toxicity of LPNs and LPNs-mRNA complexes was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The current study comprehensively investigates the optimization of the LPNs-mRNA formulation for enhanced efficacy in transgene expression in human advanced-stage melanoma cell lines.
ContributorsWubhayavedantapuram, Revanth (Author) / Rege, Kaushal (Thesis advisor) / Acharya, Abhinav (Committee member) / Yaron, Jordan (Committee member) / Arizona State University (Publisher)
Created2023
171360-Thumbnail Image.png
Description
Combining 3D bio-printing and drug delivery are promising techniques tofabricate scaffolds with well controlled and patient-specific structures for tissue engineering. In this study, silk derivatives of bioink were developed consisting of silk fibroin and gelatin then 3D printed into scaffolds. The scaffolds would be evaluated for small molecule release, cell growth, degradation, and

Combining 3D bio-printing and drug delivery are promising techniques tofabricate scaffolds with well controlled and patient-specific structures for tissue engineering. In this study, silk derivatives of bioink were developed consisting of silk fibroin and gelatin then 3D printed into scaffolds. The scaffolds would be evaluated for small molecule release, cell growth, degradation, and morphology. Preparations and design of the scaffolds are major parts of engineering and tissue engineering. Scaffolds are designed to mimic extracellular matrix by providing structural support as well as promoting cell attachment and proliferation with minimum inflammation while degrading at a controlled rate. Scaffolds offers new potentials in medicine by aiding in the preparation of personalized and controlled release therapeutic systems.
ContributorsNg, Johnny (Author) / Rege, Kaushal (Thesis advisor) / Holloway, Julianne (Committee member) / Jin, Kailong (Committee member) / Arizona State University (Publisher)
Created2022
171604-Thumbnail Image.png
Description
Skin wounds can be caused by traumatic lacerations or incisions which disrupt the structural and functional integrity of the skin. Wound closure and primary intention treatment of the wound as soon as possible is crucial to avoid or minimize the risk of infection that can result in a compromised healing

Skin wounds can be caused by traumatic lacerations or incisions which disrupt the structural and functional integrity of the skin. Wound closure and primary intention treatment of the wound as soon as possible is crucial to avoid or minimize the risk of infection that can result in a compromised healing rate or advanced functional intricacy. The gold standard treatment for skin wound healing is suturing. Light-activated tissue sealing is an appealing alternative to sutures as it seals the wound edges minimizing the risk of infection and scarring, especially when utilized along with biodegradable polymeric biomaterials in the wound bed. Silk fibroins can be used as a biodegradable biomaterial that possesses properties supporting cell migration and proliferation in the tissue it interacts with. In addition, histamine treatment is shown to have extensive effects on cellular functions promoting wound healing. Here, the evaluation of Laser-activated Sealants (LASE) consisting of silk fibroin films induced with Indocyanine Green dye in a wound sealed with laser in the presence of Histamine receptor agonists H1R, H2R and H4R take place. The results were evaluated using Trans-epidermal Water Loss (TEWL), histological and analytical techniques where immune cell biomarkers Arginase-1, Ly6G, iNOS, Alpha-SMA, Proliferating Cell Nuclear Antigen (PCNA), and E-Cadherin were used to study the activity of specific cells such as macrophages, neutrophils, and myofibroblasts that aid in wound healing. PBS was used as a control for histamine receptor agonists. It was found that TEWL increased when treated with H1 receptor agonists while decreasing significantly in H2R and H4R-treated wounds. Arginase-1 activity improved, while it displayed an inverse relationship compared to iNOS. H4R agonist escalated Alpha-SMA cells, while others did not have any significant difference. Ly6G activity depleted in all histamine agonists significantly, while PCNA and E-Cadherin failed to show a positive or negative effect.
ContributorsPatel, Dirghau Manishbhai (Author) / Rege, Kaushal (Thesis advisor) / Massia, Stephen (Committee member) / Brafman, David (Committee member) / Arizona State University (Publisher)
Created2022
171409-Thumbnail Image.png
Description
Drug delivery has made a significant contribution to cancer immunotherapy and can have a tremendous impact on modulating immunometabolism, thereby affecting cancer outcomes. Notably, the science of delivery of cancer vaccines and immunotherapeutics, modulating immune cell functions has inspired development of several successful companies and clinical products. For example, cancer

Drug delivery has made a significant contribution to cancer immunotherapy and can have a tremendous impact on modulating immunometabolism, thereby affecting cancer outcomes. Notably, the science of delivery of cancer vaccines and immunotherapeutics, modulating immune cell functions has inspired development of several successful companies and clinical products. For example, cancer vaccines require activation of dendritic cells (DCs) and tumour associated Mɸs (TAMs) through modulation of their energy metabolism (e.g., glycolysis, glutaminolysis, Krebs cycle). Similar to activated immune cells, cancer cells also upregulate glucose and glutamine transporters for proliferation and survival. Cancer cells having accelerated energy metabolism, which has been exploited as a target for various therapeutic studies. In the first strategy, an immunometabolism strategy based on sustained release of succinate from biomaterials, which incorporate succinate in the backbone of the polymer was developed. This study demonstrates that succinate-based polymeric microparticles act as alarmins by modulating the immunometabolism of DCs and Mɸs to generate robust pro-inflammatory responses for melanoma treatment in immunocompetent young as well as aging mice. In the second strategy, a biomaterial-based strategy was developed to deliver metabolites one-step downstream of the node where the glycolytic pathway is inhibited, to specifically rescue DCs from glycolysis inhibition. The study successfully demonstrated for the first time that the glycolysis of DCs can be rescued both in vitro and in vivo using a biomaterial strategy of delivering metabolites downstream of the inhibitory node. Overall, it is believed that advanced drug delivery strategies will play an important role in marrying the fields of immunometabolism and immunotherapy to generate translatable anti-cancer treatments.
ContributorsInamdar, Sahil (Author) / Acharya, Abhinav P (Thesis advisor) / Rege, Kaushal (Committee member) / Green, Matthew (Committee member) / Curtis, Marion (Committee member) / Seetharam, Mahesh (Committee member) / Arizona State University (Publisher)
Created2022
166211-Thumbnail Image.png
Description

The current clinical gold standards for tissue sealing include sutures, staples, and glues, however several adverse effects limit their use. Sutures and staples inherently cause additional trauma to tissue surrounding the wound, and glues can be lacking in adhesion and are potentially inflammatory. All three also introduce risk of infection.

The current clinical gold standards for tissue sealing include sutures, staples, and glues, however several adverse effects limit their use. Sutures and staples inherently cause additional trauma to tissue surrounding the wound, and glues can be lacking in adhesion and are potentially inflammatory. All three also introduce risk of infection. Light-activated tissue sealing, particularly the use of near-infrared light, is an attractive alternative, as it localizes heat, thereby preventing thermal damage to the surrounding healthy tissue. Previous work identified a glutaraldehyde-crosslinked chitosan film as a lead sealant for gastrointestinal incision sealing, but in vivo testing resulted in tissue degradation in and around the wound. The suggested causes for this degradation were excess acetic acid, endotoxins in the chitosan, and thermal damage. A basic buffer wash protocol was developed to remove excess acid from the films following fabrication. UV-Vis spectroscopy demonstrated that following the wash, films had the same concentration of Indocyanine green as unwashed films, allowing them to absorb light at the same wavelength, therefore showing the wash did not affect the film’s function. However subsequent washes led to degradation of film mass of nearly 20%. Standard chitosan films had significantly greater mass gain (p = 0.028) and significantly less subsequent loss (p= 0.012) than endotoxin free chitosan-films after soaking in phosphate buffered saline for varying durations , while soaking duration had no effect (p = 0.332). Leak pressure testing of films prepared with varying numbers of buffer washes, laser temperature, and lasering time revealed no significant interaction between any of the 3 variables. As such, it was confirmed that proceeding with in vivo testing with the buffer wash, various lasering temperatures, and laser times would not affect the sealing performance of the films. Future investigation will involve characterization of additional materials that may be effective for sealing of internal wounds, as well as drug loading of agents that may hasten the healing process.

ContributorsSira, Antara (Author) / Rege, Kaushal (Thesis director) / Weaver, Jessica (Committee member) / Barrett, The Honors College (Contributor) / Harrington Bioengineering Program (Contributor)
Created2022-05
154363-Thumbnail Image.png
Description
Relapse after tumor dormancy is one of the leading causes of cancer recurrence that ultimately leads to patient mortality. Upon relapse, cancer manifests as metastases that are linked to almost 90% cancer related deaths. Capture of the dormant and relapsed tumor phenotypes in high-throughput will allow for rapid targeted drug

Relapse after tumor dormancy is one of the leading causes of cancer recurrence that ultimately leads to patient mortality. Upon relapse, cancer manifests as metastases that are linked to almost 90% cancer related deaths. Capture of the dormant and relapsed tumor phenotypes in high-throughput will allow for rapid targeted drug discovery, development and validation. Ablation of dormant cancer will not only completely remove the cancer disease, but also will prevent any future recurrence. A novel hydrogel, Amikagel, was developed by crosslinking of aminoglycoside amikacin with a polyethylene glycol crosslinker. Aminoglycosides contain abundant amount of easily conjugable groups such as amino and hydroxyl moieties that were crosslinked to generate the hydrogel. Cancer cells formed 3D spheroidal structures that underwent near complete dormancy on Amikagel high-throughput drug discovery platform. Due to their dormant status, conventional anticancer drugs such as mitoxantrone and docetaxel that target the actively dividing tumor phenotype were found to be ineffective. Hypothesis driven rational drug discovery approaches were used to identify novel pathways that could sensitize dormant cancer cells to death. Strategies were used to further accelerate the dormant cancer cell death to save time required for the therapeutic outcome.

Amikagel’s properties were chemo-mechanically tunable and directly impacted the outcome of tumor dormancy or relapse. Exposure of dormant spheroids to weakly stiff and adhesive formulation of Amikagel resulted in significant relapse, mimicking the response to changes in extracellular matrix around dormant tumors. Relapsed cells showed significant differences in their metastatic potential compared to the cells that remained dormant after the induction of relapse. Further, the dissertation discusses the use of Amikagels as novel pDNA binding resins in microbead and monolithic formats for potential use in chromatographic purifications. High abundance of amino groups allowed their utilization as novel anion-exchange pDNA binding resins. This dissertation discusses Amikagel formulations for pDNA binding, metastatic cancer cell separation and novel drug discovery against tumor dormancy and relapse.
ContributorsGrandhi, Taraka Sai Pavan (Author) / Rege, Kaushal (Thesis advisor) / Meldrum, Deirdre R (Thesis advisor) / Stabenfeldt, Sarah (Committee member) / Caplan, Michael (Committee member) / Tian, Yanqing (Committee member) / Arizona State University (Publisher)
Created2016
157837-Thumbnail Image.png
Description
Tissue approximation and repair have been performed with sutures and staples for centuries, but these means are inherently traumatic. Tissue repair using laser-responsive nanomaterials can lead to rapid tissue sealing and repair and is an attractive alternative to existing clinical methods. Laser tissue welding is a sutureless technique for sealing

Tissue approximation and repair have been performed with sutures and staples for centuries, but these means are inherently traumatic. Tissue repair using laser-responsive nanomaterials can lead to rapid tissue sealing and repair and is an attractive alternative to existing clinical methods. Laser tissue welding is a sutureless technique for sealing incised or wounded tissue, where chromophores convert laser light to heat to induce in tissue sealing. Introducing chromophores that absorb near-infrared light creates differential laser absorption and allows for laser wavelengths that minimizes tissue damage.

In this work, plasmonic nanocomposites have been synthesized and used in laser tissue welding for ruptured porcine intestine ex vivo and incised murine skin in vivo. These laser-responsive nanocomposites improved tissue strength and healing, respectively. Additionally, a spatiotemporal model has been developed for laser tissue welding of porcine and mouse cadaver intestine sections using near-infrared laser irradiation. This mathematical model can be employed to identify optimal conditions for minimizing healthy cell death while still achieving a strong seal of the ruptured tissue using laser welding. Finally, in a model of surgical site infection, laser-responsive nanomaterials were shown to be efficacious in inhibiting bacterial growth. By incorporating an anti-microbial functionality to laser-responsive nanocomposites, these materials will serve as a treatment modality in sealing tissue, healing tissue, and protecting tissue in surgery.
ContributorsUrie, Russell Ricks (Author) / Rege, Kaushal (Thesis advisor) / Acharya, Abhinav (Committee member) / DeNardo, Dale (Committee member) / Holloway, Julianne (Committee member) / Thomas, Marylaura (Committee member) / Arizona State University (Publisher)
Created2019