Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kameron V. Kilchrist is active.

Publication


Featured researches published by Kameron V. Kilchrist.


Biomaterials | 2015

Tuning PEGylation of mixed micelles to overcome intracellular and systemic siRNA delivery barriers.

Martina Miteva; Kellye C. Kirkbride; Kameron V. Kilchrist; Thomas A. Werfel; Hongmei Li; Christopher E. Nelson; Mukesh K. Gupta; Todd D. Giorgio; Craig L. Duvall

A series of endosomolytic mixed micelles was synthesized from two diblock polymers, poly[ethylene glycol-b-(dimethylaminoethyl methacrylate-co-propylacrylic acid-co-butyl methacrylate)] (PEG-b-pDPB) and poly[dimethylaminoethyl methacrylate-b-(dimethylaminoethyl methacrylate-co-propylacrylic acid-co-butyl methacrylate)] (pD-b-pDPB), and used to determine the impact of both surface PEG density and PEG molecular weight on overcoming both intracellular and systemic siRNA delivery barriers. As expected, the percent PEG composition and PEG molecular weight in the corona had an inverse relationship with mixed micelle zeta potential and rate of cellular internalization. Although mixed micelles were internalized more slowly, they generally produced similar gene silencing bioactivity (∼ 80% or greater) in MDA-MB-231 breast cancer cells as the micelles containing no PEG (100 D/no PEG). The mechanistic explanation for the potent bioactivity of the promising 50 mol% PEG-b-DPB/50 mol% pD-b-pDPB (50 D) mixed micelle formulation, despite its relatively low rate of cellular internalization, was further investigated as a function of PEG molecular weight (5 k, 10 k, or 20 k PEG). Results indicated that, although larger molecular weight PEG decreased cellular internalization, it improved cytoplasmic bioavailability due to increased intracellular unpackaging (quantitatively measured via FRET) and endosomal release. When delivered intravenously in vivo, 50 D mixed micelles with a larger molecular weight PEG in the corona also demonstrated significantly improved blood circulation half-life (17.8 min for 20 k PEG micelles vs. 4.6 min for 5 kDa PEG micelles) and a 4-fold decrease in lung accumulation. These studies provide new mechanistic insights into the functional effects of mixed micelle-based approaches to nanocarrier surface PEGylation. Furthermore, the ideal mixed micelle formulation identified (50 D/20 k PEG) demonstrated desirable intracellular and systemic pharmacokinetics and thus has strong potential for in vivo therapeutic use.


Advanced Materials | 2016

Porous Silicon and Polymer Nanocomposites for Delivery of Peptide Nucleic Acids as Anti-MicroRNA Therapies

Kelsey R. Beavers; Thomas A. Werfel; Tianwei Shen; Taylor E. Kavanaugh; Kameron V. Kilchrist; Jeremy W. Mares; Joshua S. Fain; Carrie B. Wiese; Kasey C. Vickers; Sharon M. Weiss; Craig L. Duvall

Self-assembled polymer/porous silicon nanocomposites overcome intracellular and systemic barriers for in vivo application of peptide nucleic acid (PNA) anti-microRNA therapeutics. Porous silicon (PSi) is leveraged as a biodegradable scaffold with high drug-cargo-loading capacity. Functionalization with a diblock polymer improves PSi nanoparticle colloidal stability, in vivo pharmacokinetics, and intracellular bioavailability through endosomal escape, enabling PNA to inhibit miR-122 in vivo.


Science Translational Medicine | 2015

MK2 inhibitory peptide delivered in nanopolyplexes prevents vascular graft intimal hyperplasia

Brian C. Evans; Kyle M. Hocking; Igor Voskresensky; Julia Dmowska; Kameron V. Kilchrist; Colleen M. Brophy; Craig L. Duvall

Nanopolyplexes formulated from a pH-responsive, endosomolytic polymer with a peptide inhibitor of MAPKAP kinase 2 block inflammatory and migratory signaling in vascular smooth muscle cells and prevent intimal hyperplasia in human saphenous vein grafts. Nano keeps MK2 inhibitor intact, on-target A peptide, currently in clinical trials, that can penetrate cells and block the activity of MAPKAP kinase 2 (MK2) may be able to stop inflammation and fibrosis after vein grafting, but it has low bioavailability and is degraded easily once inside the cell. To more effectively translate this inhibitory peptide, called MK2i, Evans et al. formulated it in electrostatically complexed nanoparticles—nanopolyplexes—for delivery to vascular cells and tissues. The MK2i nanopolyplexes were taken up readily by vascular smooth muscle cells and endothelial cells in human saphenous veins and significantly inhibited neointima formation ex vivo. In rabbit vein grafts, treatment with the MK2 nanopolyplexes prevented intimal hyperplasia for 1 month after transplant; by contrast, free MK2i peptide had no effect. Thus, complexing the MK2 inhibitor peptide with an endosomolytic polymer could improve long-term graft patency. Both treatments were able to block macrophage recruitment and/or signaling in vivo, possibly leading to less inflammation. In human saphenous veins, the MK2i nanopolyplexes similarly reduced proinflammatory cytokines and were also shown to reduce vascular smooth muscle cell migration. Such new insights into the effects of MK2i on intimal hyperplasia could open doors to new therapeutic options in this multifactorial disease. Furthermore, this nanoencapsulation approach could be broadly applied to other therapeutic cell-penetrating peptides to prolong bioavailability and enhance stability in vivo. Autologous vein grafts are commonly used for coronary and peripheral artery bypass but have a high incidence of intimal hyperplasia (IH) and failure. We present a nanopolyplex (NP) approach that efficiently delivers a mitogen-activated protein kinase (MAPK)–activated protein (MAPKAP) kinase 2 inhibitory peptide (MK2i) to graft tissue to improve long-term patency by inhibiting pathways that initiate IH. In vitro testing in human vascular smooth muscle cells revealed that formulation into MK2i-NPs increased cell internalization, endosomal escape, and intracellular half-life of MK2i. This efficient delivery mechanism enabled MK2i-NPs to sustain potent inhibition of inflammatory cytokine production and migration in vascular cells. In intact human saphenous vein, MK2i-NPs blocked inflammatory and migratory signaling, as confirmed by reduced phosphorylation of the posttranscriptional gene regulator heterogeneous nuclear ribonucleoprotein A0, the transcription factor cAMP (adenosine 3′,5′-monophosphate) element–binding protein, and the chaperone heat shock protein 27. The molecular effects of MK2i-NPs caused functional inhibition of IH in human saphenous vein cultured ex vivo. In a rabbit vein transplant model, a 30-min intraoperative graft treatment with MK2i-NPs significantly reduced in vivo IH 28 days posttransplant compared with untreated or free MK2i–treated grafts. The decrease in IH in MK2i-NP–treated grafts in the rabbit model also corresponded with decreased cellular proliferation and maintenance of the vascular wall smooth muscle cells in a more contractile phenotype. These data indicate that nanoformulated MK2 inhibitors are a promising strategy for preventing graft failure.


ACS Nano | 2015

Endosomolytic Nano-Polyplex Platform Technology for Cytosolic Peptide Delivery To Inhibit Pathological Vasoconstriction

Brian C. Evans; Kyle M. Hocking; Kameron V. Kilchrist; Eric S. Wise; Colleen M. Brophy; Craig L. Duvall

A platform technology has been developed and tested for delivery of intracellular-acting peptides through electrostatically complexed nanoparticles, or nano-polyplexes, formulated from an anionic endosomolytic polymer and cationic therapeutic peptides. This delivery platform has been initially tested and optimized for delivery of two unique vasoactive peptides, a phosphomimetic of heat shock protein 20 and an inhibitor of MAPKAP kinase II, to prevent pathological vasoconstriction (i.e., vasospasm) in human vascular tissue. These peptides inhibit vasoconstriction and promote vasorelaxation by modulating actin dynamics in vascular smooth muscle cells. Formulating these peptides into nano-polyplexes significantly enhances peptide uptake and retention, facilitates cytosolic delivery through a pH-dependent endosomal escape mechanism, and enhances peptide bioactivity in vitro as measured by inhibition of F-actin stress fiber formation. In comparison to treatment with the free peptides, which were endowed with cell-penetrating sequences, the nano-polyplexes significantly increased vasorelaxation, inhibited vasoconstriction, and decreased F-actin formation in the human saphenous vein ex vivo. These results suggest that these formulations have significant potential for treatment of conditions such as cerebral vasospasm following subarachnoid hemorrhage. Furthermore, because many therapeutic peptides include cationic cell-penetrating segments, this simple and modular platform technology may have broad applicability as a cost-effective approach for enhancing the efficacy of cytosolically active peptides.


Journal of Biomedical Materials Research Part A | 2015

Conjugation of palmitic acid improves potency and longevity of siRNA delivered via endosomolytic polymer nanoparticles

Samantha M. Sarett; Kameron V. Kilchrist; Martina Miteva; Craig L. Duvall

Clinical translation of siRNA therapeutics has been limited by the inability to effectively overcome the rigorous delivery barriers associated with intracellular-acting biologics. Here, to address both potency and longevity of siRNA gene silencing, pH-responsive micellar nanoparticle (NP) carriers loaded with siRNA conjugated to palmitic acid (siRNA-PA) were investigated as a combined approach to improve siRNA endosomal escape and stability. Conjugation to hydrophobic PA improved NP loading efficiency relative to unmodified siRNA, enabling complete packaging of siRNA-PA at a lower polymer:siRNA ratio. PA conjugation also increased intracellular uptake of the nucleic acid cargo by 35-fold and produced a 3.1-fold increase in intracellular half-life. The higher uptake and improved retention of siRNA-PA NPs correlated to a 2- and 11-fold decrease in gene silencing IC50 in comparison to siRNA NPs in fibroblasts and mesenchymal stem cells, respectively, for both the model gene luciferase and the therapeutically relevant gene prolyl hydroxylase domain protein 2 (PHD2) . PA conjugation also significantly increased longevity of silencing activity following a single treatment in fibroblasts. Thus, conjugation of PA to siRNA paired with endosomolytic NPs is a promising approach to enhance the functional efficacy of siRNA in tissue regenerative and other applications.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Lipophilic siRNA targets albumin in situ and promotes bioavailability, tumor penetration, and carrier-free gene silencing

Samantha M. Sarett; Thomas A. Werfel; Linus Lee; Meredith A. Jackson; Kameron V. Kilchrist; Dana M. Brantley-Sieders; Craig L. Duvall

Significance Small interfering RNA (siRNA) has the capacity to silence traditionally undruggable targets, but in vivo delivery barriers limit clinical translation of siRNA, especially for nonhepatic targets such as solid tumors. Most delivery strategies for RNAi cancer therapies focus on synthetic nanocarriers, but their shortcomings include limited delivery to and variable distribution throughout the target site and low therapeutic indices due to nonspecific, carrier-associated toxicities. A diacyl lipid-modified siRNA can leverage albumin as an endogenous carrier, resulting in comprehensively enhanced pharmacokinetic properties that translate to greater quantity and homogeneity of tumor accumulation relative to nanocarriers. The albumin-binding siRNA conjugate strategy is synthetically simple and safe at high doses, and thus is a translatable and potentially transformative option for RNAi oncology therapies. Clinical translation of therapies based on small interfering RNA (siRNA) is hampered by siRNAs comprehensively poor pharmacokinetic properties, which necessitate molecule modifications and complex delivery strategies. We sought an alternative approach to commonly used nanoparticle carriers by leveraging the long-lived endogenous serum protein albumin as an siRNA carrier. We synthesized siRNA conjugated to a diacyl lipid moiety (siRNA-L2), which rapidly binds albumin in situ. siRNA-L2, in comparison with unmodified siRNA, exhibited a 5.7-fold increase in circulation half-life, an 8.6-fold increase in bioavailability, and reduced renal accumulation. Benchmarked against leading commercial siRNA nanocarrier in vivo jetPEI, siRNA-L2 achieved 19-fold greater tumor accumulation and 46-fold increase in per-tumor-cell uptake in a mouse orthotopic model of human triple-negative breast cancer. siRNA-L2 penetrated tumor tissue rapidly and homogeneously; 30 min after i.v. injection, siRNA-L2 achieved uptake in 99% of tumor cells, compared with 60% for jetPEI. Remarkably, siRNA-L2 achieved a tumor:liver accumulation ratio >40:1 vs. <3:1 for jetPEI. The improved pharmacokinetic properties of siRNA-L2 facilitated significant tumor gene silencing for 7 d after two i.v. doses. Proof-of-concept was extended to a patient-derived xenograft model, in which jetPEI tumor accumulation was reduced fourfold relative to the same formulation in the orthotopic model. The siRNA-L2 tumor accumulation diminished only twofold, suggesting that the superior tumor distribution of the conjugate over nanoparticles will be accentuated in clinical situations. These data reveal the immense promise of in situ albumin targeting for development of translational, carrier-free RNAi-based cancer therapies.


ACS Nano | 2017

Zwitterionic Nanocarrier Surface Chemistry Improves siRNA Tumor Delivery and Silencing Activity Relative to Polyethylene Glycol

Meredith A. Jackson; Thomas A. Werfel; Elizabeth J. Curvino; Fang Yu; Taylor E. Kavanaugh; Samantha M. Sarett; Mary D. Dockery; Kameron V. Kilchrist; Ayisha N. Jackson; Todd D. Giorgio; Craig L. Duvall

Although siRNA-based nanomedicines hold promise for cancer treatment, conventional siRNA-polymer complex (polyplex) nanocarrier systems have poor pharmacokinetics following intravenous delivery, hindering tumor accumulation. Here, we determined the impact of surface chemistry on the in vivo pharmacokinetics and tumor delivery of siRNA polyplexes. A library of diblock polymers was synthesized, all containing the same pH-responsive, endosomolytic polyplex core-forming block but different corona blocks: 5 kDa (benchmark) and 20 kDa linear polyethylene glycol (PEG), 10 kDa and 20 kDa brush-like poly(oligo ethylene glycol), and 10 kDa and 20 kDa zwitterionic phosphorylcholine-based polymers (PMPC). In vitro, it was found that 20 kDa PEG and 20 kDa PMPC had the highest stability in the presence of salt or heparin and were the most effective at blocking protein adsorption. Following intravenous delivery, 20 kDa PEG and PMPC coronas both extended circulation half-lives 5-fold compared to 5 kDa PEG. However, in mouse orthotopic xenograft tumors, zwitterionic PMPC-based polyplexes showed highest in vivo luciferase silencing (>75% knockdown for 10 days with single IV 1 mg/kg dose) and 3-fold higher average tumor cell uptake than 5 kDa PEG polyplexes (20 kDa PEG polyplexes were only 2-fold higher than 5 kDa PEG). These results show that high molecular weight zwitterionic polyplex coronas significantly enhance siRNA polyplex pharmacokinetics without sacrificing polyplex uptake and bioactivity within tumors when compared to traditional PEG architectures.


Cancer Research | 2018

Selective mTORC2 Inhibitor Therapeutically Blocks Breast Cancer Cell Growth and Survival

Thomas A. Werfel; Shan Wang; Meredith A. Jackson; Taylor E. Kavanaugh; Meghan Morrison Joly; Linus Lee; Donna Hicks; Violeta Sanchez; Paula I Gonzalez-Ericsson; Kameron V. Kilchrist; Somtochukwu C Dimobi; Samantha M. Sarett; Dana M. Brantley-Sieders; Rebecca S. Cook; Craig L. Duvall

Small-molecule inhibitors of the mTORC2 kinase (torkinibs) have shown efficacy in early clinical trials. However, the torkinibs under study also inhibit the other mTOR-containing complex mTORC1. While mTORC1/mTORC2 combined inhibition may be beneficial in cancer cells, recent reports describe compensatory cell survival upon mTORC1 inhibition due to loss of negative feedback on PI3K, increased autophagy, and increased macropinocytosis. Genetic models suggest that selective mTORC2 inhibition would be effective in breast cancers, but the lack of selective small-molecule inhibitors of mTORC2 have precluded testing of this hypothesis to date. Here we report the engineering of a nanoparticle-based RNAi therapeutic that can effectively silence the mTORC2 obligate cofactor Rictor. Nanoparticle-based Rictor ablation in HER2-amplified breast tumors was achieved following intratumoral and intravenous delivery, decreasing Akt phosphorylation and increasing tumor cell killing. Selective mTORC2 inhibition in vivo, combined with the HER2 inhibitor lapatinib, decreased the growth of HER2-amplified breast cancers to a greater extent than either agent alone, suggesting that mTORC2 promotes lapatinib resistance, but is overcome by mTORC2 inhibition. Importantly, selective mTORC2 inhibition was effective in a triple-negative breast cancer (TNBC) model, decreasing Akt phosphorylation and tumor growth, consistent with our findings that RICTOR mRNA correlates with worse outcome in patients with basal-like TNBC. Together, our results offer preclinical validation of a novel RNAi delivery platform for therapeutic gene ablation in breast cancer, and they show that mTORC2-selective targeting is feasible and efficacious in this disease setting.Significance: This study describes a nanomedicine to effectively inhibit the growth regulatory kinase mTORC2 in a preclinical model of breast cancer, targeting an important pathogenic enzyme in that setting that has been undruggable to date. Cancer Res; 78(7); 1845-58. ©2018 AACR.


Journal of Biomedical Materials Research Part A | 2016

Hydrolytic Charge‐reversal of PEGylated Polyplexes Enhances Intracellular Un‐packaging and Activity of siRNA

Thomas A. Werfel; Corban Swain; Christopher E. Nelson; Kameron V. Kilchrist; Brian C. Evans; Martina Miteva; Craig L. Duvall

Hydrolytically degrading nano-polyplexes (HDG-NPs) that reverse charge through conversion of tertiary amines to carboxylic acids were investigated to improve intracellular un-packaging of siRNA and target gene silencing compared to a non-degradable analog (non-HDG-NPs). Both NP types comprised reversible addition-fragmentation chain-transfer (RAFT) synthesized diblock copolymers of a poly(ethylene glycol) (PEG) corona-forming block and a cationic block for nucleic acid packaging that incorporated butyl methacrylate (BMA) and either dimethylaminoethyl methacrylate (DMAEMA, non-HDG-NPs) or dimethylaminoethyl acrylate (DMAEA, HDG-NPs). HDG-NPs decreased significantly in size and released significantly more siRNA (∼40%) than non-HDG-NPs after 24 h in aqueous solution. While both HDG-NPs and non-HDG-NPs had comparable uptake and cytotoxicity up to 150 nM siRNA doses, HDG-NPs achieved significantly higher target gene silencing of the model gene luciferase in vitro. High resolution FRET confocal microscopy was used to monitor the intracellular un-packaging of siRNA. Non-HDG-NPs had significantly higher FRET efficiency than HDG-NPs, indicating that siRNA delivered from HDG-NPs was more fully un-packaged and therefore had improved intracellular bioavailability.


Journal of Controlled Release | 2018

Excipients for the lyoprotection of MAPKAP kinase 2 inhibitory peptide nano-polyplexes

Alvin J. Mukalel; Brian C. Evans; Kameron V. Kilchrist; Eric A. Dailing; Benjamin Burdette; Joyce Cheung-Flynn; Colleen M. Brophy; Craig L. Duvall

ABSTRACT Herein, excipients are investigated to ameliorate the deleterious effects of lyophilization on peptide‐polymer nano‐polyplex (NP) morphology, cellular uptake, and bioactivity. The NPs are a previously‐described platform technology for intracellular peptide delivery and are formulated from a cationic therapeutic peptide and the anionic, pH‐responsive, endosomolytic polymer poly(propylacrylic acid) (PPAA). These NPs are effective when formulated and immediately used for delivery into cells and tissue, but they are not amenable to reconstitution following storage as a lyophilized powder due to aggregation. To develop a lyophilized NP format that facilitates longer‐term storage and ease of use, MAPKAP kinase 2 inhibitory peptide‐based NPs (MK2i‐NPs) were prepared in the presence of a range of concentrations of the excipients sucrose, trehalose, and lactosucrose prior to lyophilization and storage. All excipients improved particle morphology post‐lyophilization and significantly improved MK2i‐NP uptake in human coronary artery smooth muscle cells relative to lyophilized NPs without excipient. In particular, MK2i‐NPs lyophilized with 300 mM lactosucrose as an excipient demonstrated a 5.23 fold increase in cellular uptake (p < 0.001), a 2.52 fold increase in endosomal disruption (p < 0.05), and a 2.39 fold increase in ex vivo bioactivity (p < 0.01) compared to MK2i‐NPs lyophilized without excipients. In sum, these data suggest that addition of excipients, particularly lactosucrose, maintains and even improves the uptake and therapeutic efficacy of peptide‐polymer NPs post‐lyophilization relative to freshly‐made formulations. Thus, the use of excipients as lyoprotectants is a promising approach for the long‐term storage of biotherapeutic NPs and poises this NP platform for clinical translation. Graphical abstract Figure. No Caption available. HighlightsNano‐Polyplexes (NPs) aggregate and lose bioactivity post‐lyophilization.Excipients preserve NP size and morphology post‐lyophilization.NPs lyophilized with lactosucrose have higher cellular uptake than even fresh NPs.NPs lyophilized with lactosucrose retain effective endosomal disruption ability.NPs lyophilized with lactosucrose have potent bioactivity in intact human vein.

Collaboration


Dive into the Kameron V. Kilchrist's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Colleen M. Brophy

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge