Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Sci (Weinh) ; 10(7): e2205389, 2023 03.
Article in English | MEDLINE | ID: mdl-36642846

ABSTRACT

Proteins are among the most common therapeutics for the treatment of diabetes, autoimmune diseases, cancer, and metabolic diseases, among others. Despite their common use, current protein therapies, most of which are injectables, have several limitations. Large proteins such as monoclonal antibodies (mAbs) suffer from poor absorption after subcutaneous injections, thus forcing their administration by intravenous injections. Even small proteins such as insulin suffer from slow pharmacokinetics which poses limitations in effective management of diabetes. Here, a deep eutectic-based delivery strategy is used to offer a generalized approach for improving protein absorption after subcutaneous injections. The lead formulation enhances absorption of mAbs after subcutaneous injections by ≈200%. The same composition also improves systemic absorption of subcutaneously injected insulin faster than Humalog, the current gold-standard of rapid acting insulin. Mechanistic studies reveal that the beneficial effect of deep eutectics on subcutaneous absorption is mediated by their ability to reduce the interactions of proteins with the subcutaneous matrix, especially collagen. Studies also confirm that these deep eutectics are safe for subcutaneous injections. Deep eutectic-based formulations described here open new possibilities for subcutaneous injections of therapeutic proteins.


Subject(s)
Biological Products , Deep Eutectic Solvents , Humans , Antibodies, Monoclonal/pharmacokinetics , Deep Eutectic Solvents/pharmacology , Deep Eutectic Solvents/therapeutic use , Diabetes Mellitus/drug therapy , Diabetes Mellitus/therapy , Injections, Subcutaneous/methods , Insulin , Biological Products/administration & dosage , Biological Products/therapeutic use
2.
Bioeng Transl Med ; 7(2): e10283, 2022 May.
Article in English | MEDLINE | ID: mdl-35600639

ABSTRACT

Induced neural stem cells (iNSCs) have emerged as a promising therapeutic platform for glioblastoma (GBM). iNSCs have the innate ability to home to tumor foci, making them ideal carriers for antitumor payloads. However, the in vivo persistence of iNSCs limits their therapeutic potential. We hypothesized that by encapsulating iNSCs in the FDA-approved, hemostatic matrix FLOSEAL®, we could increase their persistence and, as a result, therapeutic durability. Encapsulated iNSCs persisted for 95 days, whereas iNSCs injected into the brain parenchyma persisted only 2 weeks in mice. Two orthotopic GBM tumor models were used to test the efficacy of encapsulated iNSCs. In the GBM8 tumor model, mice that received therapeutic iNSCs encapsulated in FLOSEAL® survived 30 to 60 days longer than mice that received nonencapsulated cells. However, the U87 tumor model showed no significant differences in survival between these two groups, likely due to the more solid and dense nature of the tumor. Interestingly, the interaction of iNSCs with FLOSEAL® appears to downregulate some markers of proliferation, anti-apoptosis, migration, and therapy which could also play a role in treatment efficacy and durability. Our results demonstrate that while FLOSEAL® significantly improves iNSC persistence, this alone is insufficient to enhance therapeutic durability.

3.
Adv Healthc Mater ; 10(13): e2002192, 2021 07.
Article in English | MEDLINE | ID: mdl-34050617

ABSTRACT

The mucus barrier lining the gastrointestinal tract poses a significant barrier to the oral delivery of macromolecular drugs. Successful approaches to overcoming this barrier have primarily focused on reducing drug and carrier interactions with mucus or disrupting the mucus layer directly. Choline-based ionic liquids (ILs) such as choline geranate and choline glycolate (CGLY) have recently been shown to be effective in enhancing the intestinal absorption of macromolecules such as insulin and immunoglobulin (IgG), respectively. Herein, the use of choline-based ILs as mucus-modulating agents for safely improving drug penetration through mucus is described. Choline-based ILs significantly increase the diffusion rates of cationic dextrans through mucin solution. Choline-maleic acid (CMLC 2:1) enhances the diffusion of 4 kDa cationic dextran in mucin solution by more than fourfold when compared to phosphate-buffered saline control. Choline-based ILs also reduce mucus viscosity without significantly impacting the native mucus gel structure. In vitro studies in a mucus-secreting coculture model with Caco-2 and HT29MTX-E12 cells further demonstrate the effectiveness of ILs in improving transport of cationic molecules in the presence of secreted mucus. This work demonstrates the potential for choline-based ionic liquids to be used as nondestructive mucus-modulating agents for enabling enhanced oral delivery of macromolecular drugs.


Subject(s)
Ionic Liquids , Pharmaceutical Preparations , Caco-2 Cells , Drug Delivery Systems , Humans , Mucus
4.
Tissue Eng Part A ; 27(13-14): 857-866, 2021 07.
Article in English | MEDLINE | ID: mdl-32907497

ABSTRACT

Engineered neural stem cells (NSCs) have recently emerged as a promising therapy. Acting as a tumor-homing drug-delivery system, NSCs migrate through brain tissue to seek out primary and invasive tumor foci. NSCs can deliver therapeutic agents, such as TNFα-related apoptosis-inducing ligand, directly to the tumor and suppress glioblastoma (GBM) in murine models. While the mainstays for evaluating NSC migration and efficacy have been two-dimensional chemotaxis assays and mouse models, these low-throughput and small-scale systems limit our ability to implant and track these cells for human translation. To circumvent these challenges, we developed a three-dimensional culture system using a matrix of poly-l-lactic acid 6100 microfibers suspended in agar. These bioinspired brain matrices were used to model tumor growth, NSC migration, and efficacy of NSC therapy at small and human scale. Kinetic fluorescent imaging confirmed growth of tumors in both small and human-sized bioinspired brain matrix. Tumors proliferated 50-fold and 3-fold for GBM and human metastatic breast cancer, respectively, over 7 days. We next explored the impact of tumor location on NSC migration. When NSCs were implanted 2 mm lateral from the tumor foci, NSCs colocalized with the GBM within 7 days. In models of multifocal disease, NSCs were found to colocalize with multiple tumors, preferentially migrating to tumor foci closest to the site of NSC implantation. Lastly, therapeutic NSCs were implanted at increasing distances (0, 2, 5, or 10 mm) laterally from GBM foci to investigate the effects of distance on NSC efficacy. Serial imaging showed reduced fluorescence at tumor sites, implicating GBM apoptosis across all distances. NSCs coinjected with tumor induced a near-complete response in <10 days, while NSCs implanted 10 mm laterally from the tumor induced a near-complete response by day 30. Lastly, GBM foci were established in each hemisphere of the model and control or therapeutic NSCs were implanted adjacent to tumor cells in the right hemisphere. Kinetic imaging showed that NSC therapy attenuated progression of GBM foci, while GBM cells treated with control NSC expanded rapidly over 21 days. In conclusion, we developed a new bioinspired model that supports growth of human brain cancer cells and enables rapid tracking of NSC therapy. Impact statement Tumor-homing and tumor-killing-engineered neural stem cell (NSC) therapies have shown immense promise in both preclinical and clinical trials. However, as cell therapies continue to evolve, cost-effective and high-throughput screening assays are needed to assess the proliferation, migration, and efficacy of these cells. In this study, we developed a bioinspired brain matrix for the evaluation of engineered NSCs. Importantly, this matrix is easy to fabricate, scalable, and allows for sterile real-time, noninvasive imaging using our custom bioreactor. We then utilized the bioinspired brain matrix system to answer key questions around the tumor-homing migration and efficacy of engineered NSC therapies that are challenging to address with traditional models.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Stem Cells , Animals , Apoptosis , Brain/diagnostic imaging , Brain Neoplasms/therapy , Glioblastoma/therapy , Mice
5.
Sci Adv ; 6(48)2020 11.
Article in English | MEDLINE | ID: mdl-33239302

ABSTRACT

The rapid clearance of intravenously administered nanoparticles (NPs) from the bloodstream is a major unsolved problem in nanomedicine. Here, we describe the first use of biocompatible protein-avoidant ionic liquids (PAILs) as NP surface modifiers to reduce opsonization. An ionic liquid choline hexenoate, selected for its aversion to serum proteins, was used to stably coat the surface of poly(lactic-co-glycolic acid) (PLGA) NPs. Compared with bare PLGA and poly(ethylene glycol)-coated PLGA particles, the PAIL-PLGA NPs showed resistance to protein adsorption in vitro and greater retention in blood of mice at 24 hours. Choline hexenoate redirected biodistribution of NPs, with preferential accumulation in the lungs with 50% of the administered dose accumulating in the lungs and <5% in the liver. Lung accumulation was attributed to spontaneous attachment of the PAIL-coated NPs on red blood cells in vivo. Overall, ionic liquids are a promising class of materials for NP modification for biomedical applications.


Subject(s)
Ionic Liquids , Nanoparticles , Animals , Choline , Drug Carriers , Lactic Acid , Mice , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Distribution
6.
Tissue Eng Part A ; 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33085922

ABSTRACT

Engineered neural stem cells (NSCs) have recently emerged as a promising therapy. Acting as a tumor-homing drug-delivery system, NSCs migrate through brain tissue to seek out primary and invasive tumor foci. NSCs can deliver therapeutic agents, such as TNFα-related apoptosis-inducing ligand, directly to the tumor and suppress glioblastoma (GBM) in murine models. While the mainstays for evaluating NSC migration and efficacy have been two-dimensional chemotaxis assays and mouse models, these low-throughput and small-scale systems limit our ability to implant and track these cells for human translation. To circumvent these challenges, we developed a three-dimensional culture system using a matrix of poly-l-lactic acid 6100 microfibers suspended in agar. These bioinspired brain matrices were used to model tumor growth, NSC migration, and efficacy of NSC therapy at small and human scale. Kinetic fluorescent imaging confirmed growth of tumors in both small and human-sized bioinspired brain matrix. Tumors proliferated 50-fold and 3-fold for GBM and human metastatic breast cancer, respectively, over 7 days. We next explored the impact of tumor location on NSC migration. When NSCs were implanted 2 mm lateral from the tumor foci, NSCs colocalized with the GBM within 7 days. In models of multifocal disease, NSCs were found to colocalize with multiple tumors, preferentially migrating to tumor foci closest to the site of NSC implantation. Lastly, therapeutic NSCs were implanted at increasing distances (0, 2, 5, or 10 mm) laterally from GBM foci to investigate the effects of distance on NSC efficacy. Serial imaging showed reduced fluorescence at tumor sites, implicating GBM apoptosis across all distances. NSCs coinjected with tumor induced a near-complete response in <10 days, while NSCs implanted 10 mm laterally from the tumor induced a near-complete response by day 30. Lastly, GBM foci were established in each hemisphere of the model and control or therapeutic NSCs were implanted adjacent to tumor cells in the right hemisphere. Kinetic imaging showed that NSC therapy attenuated progression of GBM foci, while GBM cells treated with control NSC expanded rapidly over 21 days. In conclusion, we developed a new bioinspired model that supports growth of human brain cancer cells and enables rapid tracking of NSC therapy. Impact statement Tumor-homing and tumor-killing-engineered neural stem cell (NSC) therapies have shown immense promise in both preclinical and clinical trials. However, as cell therapies continue to evolve, cost-effective and high-throughput screening assays are needed to assess the proliferation, migration, and efficacy of these cells. In this study, we developed a bioinspired brain matrix for the evaluation of engineered NSCs. Importantly, this matrix is easy to fabricate, scalable, and allows for sterile real-time, noninvasive imaging using our custom bioreactor. We then utilized the bioinspired brain matrix system to answer key questions around the tumor-homing migration and efficacy of engineered NSC therapies that are challenging to address with traditional models.

7.
Adv Mater ; 32(46): e2002990, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33058352

ABSTRACT

Adjuvants play a critical role in the design and development of novel vaccines. Despite extensive research, only a handful of vaccine adjuvants have been approved for human use. Currently used adjuvants are mostly composed of components that are non-native to the human body, such as aluminum salt, bacterial lipids, or foreign genomic material. Here, a new ionic-liquid-based adjuvant is explored, synthesized using two metabolites of the body, choline and lactic acid (ChoLa). ChoLa distributes the antigen efficiently upon injection, maintains antigen integrity, enhances immune infiltration at the injection site, and leads to a potent immune response against the antigen. Thus, it can serve as a promising safe adjuvant platform that can help to protect against pandemics and future infectious threats.


Subject(s)
Adjuvants, Immunologic/chemistry , Ionic Liquids/chemistry , Safety , Adjuvants, Immunologic/chemical synthesis , Animals , Glycerylphosphorylcholine/chemistry , Lactic Acid/chemistry
8.
Nat Med ; 25(6): 1022, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30996326

ABSTRACT

In the version of this article originally published, the graph in Extended Data Fig. 2c was a duplication of Extended Data Fig. 2b. The correct version of Extended Data Fig. 2c is now available online.

9.
Nat Med ; 25(3): 462-469, 2019 03.
Article in English | MEDLINE | ID: mdl-30742119

ABSTRACT

Immune checkpoint inhibitors have been successful across several tumor types; however, their efficacy has been uncommon and unpredictable in glioblastomas (GBM), where <10% of patients show long-term responses. To understand the molecular determinants of immunotherapeutic response in GBM, we longitudinally profiled 66 patients, including 17 long-term responders, during standard therapy and after treatment with PD-1 inhibitors (nivolumab or pembrolizumab). Genomic and transcriptomic analysis revealed a significant enrichment of PTEN mutations associated with immunosuppressive expression signatures in non-responders, and an enrichment of MAPK pathway alterations (PTPN11, BRAF) in responders. Responsive tumors were also associated with branched patterns of evolution from the elimination of neoepitopes as well as with differences in T cell clonal diversity and tumor microenvironment profiles. Our study shows that clinical response to anti-PD-1 immunotherapy in GBM is associated with specific molecular alterations, immune expression signatures, and immune infiltration that reflect the tumor's clonal evolution during treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Female , Gene Expression Profiling , Genomics , Glioblastoma/genetics , Glioblastoma/immunology , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Longitudinal Studies , Male , Middle Aged , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/immunology , T-Lymphocytes/immunology , Treatment Outcome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...