Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(3): e0287733, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38427670

RESUMO

Immune checkpoint blockade (ICB) targeting the programmed cell death protein 1 (PD-1) and its ligand 1 (PD-L1) fails to provide clinical benefit for most cancer patients due to primary or acquired resistance. Drivers of ICB resistance include tumor antigen processing/presentation machinery (APM) and IFNγ signaling mutations. Thus, there is an unmet clinical need to develop alternative therapies for these patients. To this end, we have developed a CRISPR/Cas9 approach to generate murine tumor models refractory to PD-1/-L1 inhibition due to APM/IFNγ signaling mutations. Guide RNAs were employed to delete B2m, Jak1, or Psmb9 genes in ICB-responsive EMT6 murine tumor cells. B2m was deleted in ICB-responsive MC38 murine colon cancer cells. We report a detailed development and validation workflow including whole exome and Sanger sequencing, western blotting, and flow cytometry to assess target gene deletion. Tumor response to ICB and immune effects of gene deletion were assessed in syngeneic mice. This workflow can help accelerate the discovery and development of alternative therapies and a deeper understanding of the immune consequences of tumor mutations, with potential clinical implications.


Assuntos
Apresentação de Antígeno , Receptor de Morte Celular Programada 1 , Animais , Camundongos , Antígeno B7-H1 , Linhagem Celular Tumoral , Sistemas CRISPR-Cas/genética , Receptor de Morte Celular Programada 1/genética , RNA Guia de Sistemas CRISPR-Cas , Transdução de Sinais
2.
Cancer Immunol Immunother ; 72(8): 2783-2797, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37166485

RESUMO

There is strong evidence that chemotherapy can induce tumor necrosis which can be exploited for the targeted delivery of immuno-oncology agents into the tumor microenvironment (TME). We hypothesized that docetaxel, a chemotherapeutic agent that induces necrosis, in combination with the bifunctional molecule NHS-IL-12 (M9241), which delivers recombinant IL-12 through specific targeting of necrotic regions in the tumor, would provide a significant antitumor benefit in the poorly inflamed murine tumor model, EMT6 (breast), and in the moderately immune-infiltrated tumor model, MC38 (colorectal). Docetaxel, as monotherapy or in combination with NHS-IL-12, promoted tumor necrosis, leading to the improved accumulation and retention of NHS-IL-12 in the TME. Significant antitumor activity and prolonged survival were observed in cohorts receiving docetaxel and NHS-IL-12 combination therapy in both the MC38 and EMT6 murine models. The therapeutic effects were associated with increased tumor infiltrating lymphocytes and were dependent on CD8+ T cells. Transcriptomics of the TME of mice receiving the combination therapy revealed the upregulation of genes involving crosstalk between innate and adaptive immunity factors, as well as the downregulation of signatures of myeloid cells. In addition, docetaxel and NHS-IL-12 combination therapy effectively controlled tumor growth of PD-L1 wild-type and PD-L1 knockout MC38 in vivo, implying this combination could be applied in immune checkpoint refractory tumors, and/or tumors regardless of PD-L1 status. The data presented herein provide the rationale for the design of clinical studies employing this combination or similar combinations of agents.


Assuntos
Antígeno B7-H1 , Neoplasias , Camundongos , Animais , Docetaxel , Linfócitos T CD8-Positivos , Interleucina-12/farmacologia , Necrose , Microambiente Tumoral , Linhagem Celular Tumoral , Imunoterapia
3.
J Immunother Cancer ; 10(6)2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35764364

RESUMO

BACKGROUND: Immune checkpoint blockade (ICB) has achieved unprecedented success in treating multiple cancer types. However, clinical benefit remains modest for most patients with solid malignancies due to primary or acquired resistance. Tumor-intrinsic loss of major histocompatibility complex class I (MHC-I) and aberrations in antigen processing machinery (APM) and interferon gamma (IFN-γ) pathways have been shown to play an important role in ICB resistance. While a plethora of combination treatments are being investigated to overcome ICB resistance, there are few identified preclinical models of solid tumors harboring these deficiencies to explore therapeutic interventions that can bypass ICB resistance. Here, we investigated the combination of the epigenetic modulator entinostat and the tumor-targeted immunocytokine NHS-IL12 in three different murine tumor models resistant to αPD-1/αPD-L1 (anti-programmed cell death protein 1/anti-programmed death ligand 1) and harboring MHC-I, APM, and IFN-γ response deficiencies and differing tumor mutational burden (TMB). METHODS: Entinostat and NHS-IL12 were administered to mice bearing TC-1/a9 (lung, HPV16 E6/E7+), CMT.64 lung, or RVP3 sarcoma tumors. Antitumor efficacy and survival were monitored. Comprehensive tumor microenvironment (TME) and spleen analysis of immune cells, cytokines, and chemokines was performed. Additionally, whole transcriptomic analysis was carried out on TC-1/a9 tumors. Cancer Genome Atlas (TCGA) datasets were analyzed for translational relevance. RESULTS: We demonstrate that the combination of entinostat and NHS-IL12 therapy elicits potent antitumor activity and survival benefit through prolonged activation and tumor infiltration of cytotoxic CD8+ T cells, across αPD-1/αPD-L1 refractory tumors irrespective of TMB, including in the IFN-γ signaling-impaired RVP3 tumor model. The combination therapy promoted M1-like macrophages and activated antigen-presenting cells while decreasing M2-like macrophages and regulatory T cells in a tumor-dependent manner. This was associated with increased levels of IFN-γ, IL-12, chemokine (C-X-C motif) ligand 9 (CXCL9), and CXCL13 in the TME. Further, the combination therapy synergized to promote MHC-I and APM upregulation, and enrichment of JAK/STAT (janus kinase/signal transducers and activators of transcription), IFN-γ-response and antigen processing-associated pathways. A biomarker signature of the mechanism involved in these studies is associated with patients' overall survival across multiple tumor types. CONCLUSIONS: Our findings provide a rationale for combining the tumor-targeting NHS-IL12 with the histone deacetylase inhibitor entinostat in the clinical setting for patients unresponsive to αPD-1/αPD-L1 and/or with innate deficiencies in tumor MHC-I, APM expression, and IFN-γ signaling.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Animais , Apresentação de Antígeno , Antígeno B7-H1 , Benzamidas , Biomarcadores Tumorais , Linfócitos T CD8-Positivos , Antígenos HLA , Antígenos de Histocompatibilidade Classe I/genética , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Interferon gama , Interleucina-12/genética , Camundongos , Neoplasias/patologia , Receptor de Morte Celular Programada 1 , Piridinas , Microambiente Tumoral
4.
J Clin Invest ; 132(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35230974

RESUMO

Collagens in the extracellular matrix (ECM) provide a physical barrier to tumor immune infiltration, while also acting as a ligand for immune inhibitory receptors. Transforming growth factor-ß (TGF-ß) is a key contributor to shaping the ECM by stimulating the production and remodeling of collagens. TGF-ß activation signatures and collagen-rich environments have both been associated with T cell exclusion and lack of responses to immunotherapy. Here, we describe the effect of targeting collagens that signal through the inhibitory leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) in combination with blockade of TGF-ß and programmed cell death ligand 1 (PD-L1). This approach remodeled the tumor collagenous matrix, enhanced tumor infiltration and activation of CD8+ T cells, and repolarized suppressive macrophage populations, resulting in high cure rates and long-term tumor-specific protection across murine models of colon and mammary carcinoma. The results highlight the advantage of direct targeting of ECM components in combination with immune checkpoint blockade therapy.


Assuntos
Antígeno B7-H1 , Neoplasias , Receptores Imunológicos , Microambiente Tumoral , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Imunoterapia/métodos , Ligantes , Camundongos , Neoplasias/terapia , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Fator de Crescimento Transformador beta/metabolismo
5.
Mater Adv ; 2(5): 1644-1656, 2021 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-34368764

RESUMO

Cowpea mosaic virus (CPMV) is currently in the development pipeline for multiple biomedical applications, including cancer immunotherapy. In particular the application of CPMV as in situ vaccine has shown promise; here the plant viral nanoparticle is used as an adjuvant and is injected directly into a tumor to reverse immunosuppression and prime systemic anti-tumor immunity. Efficacy of this CPMV-based cancer immunotherapy has been demonstrated in multiple tumor mouse models and canine cancer patients. However, while CPMV is non-infectious to mammals, it is infectious to legumes and therefore, from a safety perspective, it is desired to develop non-infectious CPMV formulations. Non-infectious virus-like particles of CPMV devoid of nucleic acids have been produced; nevertheless, efficacy of such empty CPMV nanoparticles does not match efficacy of nucleic acid-laden CPMV. The multivalent capsid activates the innate immune system through pathogen pattern recognition receptors (PRRs) such as toll-like receptors (TLRs); the RNA cargo provides additional signaling through TLR-7/8, which boosts the efficacy of this adjuvant. Therefore, in this study, we set out to develop RNA-laden, but non-infectious CPMV. We report inactivation of CPMV using UV light and chemical inactivation using ß-propiolactone (ßPL) or formalin. 7.5 J cm-2 UV, 50 mM ßPL or 1 mM formalin was determined to be sufficient to inactivate CPMV and prevented plant infection. We compared the immunogenicity of native CPMV and inactivated CPMV formulations in vitro and in vivo using RAW-Blue™ reporter cells and a murine syngeneic, orthotropic melanoma model (using B16F10 cells and C57BL6 mice). While the in vitro assay indicated activation of the RAW-Blue™ reporter cells by formaldehyde and UV-inactivated CPMV at levels comparable to native CPMV; ßPL-inactivated CPMV appeared to have diminished activity. Tumor mouse model experiments indicate potent efficacy of the chemically-inactivated CPMV (UV-treated CPMV was not tested) leading to tumor regression and increased survival; efficacy was somewhat reduced when compared to CPMV, however these samples outperformed the empty CPMV nanoparticles. These results will facilitate the translational development of safe and potent CPMV-based cancer immunotherapies.

6.
Nat Commun ; 12(1): 5151, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34446712

RESUMO

Poorly inflamed carcinomas do not respond well to immune checkpoint blockade. Converting the tumour microenvironment into a functionally inflamed immune hub would extend the clinical benefit of immune therapy to a larger proportion of cancer patients. Here we show, by using comprehensive single-cell transcriptome, proteome, and immune cell analysis, that Entinostat, a class I histone deacetylase inhibitor, facilitates accumulation of the necrosis-targeted recombinant murine immune-cytokine, NHS-rmIL12, in experimental mouse colon carcinomas and poorly immunogenic breast tumours. This combination therapy reprograms the tumour innate and adaptive immune milieu to an inflamed landscape, where the concerted action of highly functional CD8+ T cells and activated neutrophils drive macrophage M1-like polarization, leading to complete tumour eradication in 41.7%-100% of cases. Biomarker signature of favourable overall survival in multiple human tumor types shows close resemblance to the immune pattern generated by Entinostat/NHS-rmIL12 combination therapy. Collectively, these findings provide a rationale for combining NHS-IL12 with Entinostat in the clinical setting.


Assuntos
Benzamidas/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/imunologia , Imunoglobulina G/administração & dosagem , Interleucina-12/administração & dosagem , Piridinas/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Imunidade Adaptativa/efeitos dos fármacos , Animais , Neoplasias da Mama/mortalidade , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/mortalidade , Quimioterapia Combinada , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Microambiente Tumoral/efeitos dos fármacos
7.
Annu Rev Virol ; 7(1): 559-587, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32991265

RESUMO

Viral nanotechnology exploits the prefabricated nanostructures of viruses, which are already abundant in nature. With well-defined molecular architectures, viral nanocarriers offer unprecedented opportunities for precise structural and functional manipulation using genetic engineering and/or bio-orthogonal chemistries. In this manner, they can be loaded with diverse molecular payloads for targeted delivery. Mammalian viruses are already established in the clinic for gene therapy and immunotherapy, and inactivated viruses or virus-like particles have long been used as vaccines. More recently, plant viruses and bacteriophages have been developed as nanocarriers for diagnostic imaging, vaccine and drug delivery, and combined diagnosis/therapy (theranostics). The first wave of these novel virus-based tools has completed clinical development and is poised to make an impact on clinical practice.


Assuntos
Bacteriófagos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Indicadores e Reagentes , Nanotecnologia/métodos , Vírus de Plantas/metabolismo , Terapia Genética/métodos , Humanos , Imunoterapia/métodos , Nanopartículas/administração & dosagem , Nanopartículas/uso terapêutico
8.
Bioconjug Chem ; 31(5): 1408-1416, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32281790

RESUMO

Amine/thiol-reactive chemistries are commonly used to conjugate antibodies to pharmaceuticals or nanoparticles. Yet, these conjugation strategies often result in unfavorable outcomes such as heterogeneous antibody display with hindered biological activity or aggregation due to multivalent interactions of the antibody and nanoparticles. Here, we report the application of a site-specific and enzymatically driven antibody conjugation strategy to functionalize virus-based nanoparticles (VNPs). Specifically, an azide-handle was introduced into the Fc region of a set of immunoglobulins using a two-step enzymatic reaction: (1) cleavage of N-linked glycan in the Fc region by a glycosidase and (2) conjugation of a chemically reactive linker (containing an azide functional handle) using a microbial transglutaminase. Conjugation of the azide-functional antibodies to several VNPs was achieved by making use of strain-promoted azide-alkyne cycloaddition. We report the conjugation of three immunoglobulin (IgG) isotypes (human IgG from sera, anti-CD47 Rat IgG2a, κ, and Trastuzumab recombinant humanized IgG1, κ) to the plant virus cowpea mosaic virus (CPMV) and the lysine mutant of tobacco mosaic virus (TMVlys) as well as bacteriophage Qß. Site-specific conjugation resulted in stable and functional antibody-VNP conjugates. In stark contrast, the use of heterobifunctional linkers targeting thiols and amines on the antibodies and VNPs, respectively, led to aggregation due to nonspecific and multivalent coupling between the antibodies and VNPs. We demonstrate that antibody-VNP conjugates were functional, and Trastuzumab-displaying VNPs targeted HER2-positive SKOV-3 human ovarian cancer cells. This bioconjugation strategy adds to the portfolio of methods that can be used for designing functional antibody-VNP conjugates.


Assuntos
Imunoglobulina G/química , Imunoglobulina G/metabolismo , Nanopartículas/química , Vírus/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Imunoconjugados/química , Imunoconjugados/metabolismo , Ratos
9.
ACS Nano ; 14(3): 2678-2701, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32125825

RESUMO

Nanocarrier-based delivery systems can be used to increase the safety and efficacy of active ingredients in medical, veterinary, or agricultural applications, particularly when such ingredients are unstable, sparingly soluble, or cause off-target effects. In this review, we highlight the diversity of nanocarrier materials and their key advantages compared to free active ingredients. We discuss current trends based on peer-reviewed research articles, patent applications, clinical trials, and the nanocarrier formulations already approved by regulatory bodies. Although most nanocarriers have been engineered to combat cancer, the number of formulations developed for other purposes is growing rapidly, especially those for the treatment of infectious diseases and parasites affecting humans, livestock, and companion animals. The regulation and prohibition of many pesticides have also fueled research to develop targeted pesticide delivery systems based on nanocarriers, which maximize efficacy while minimizing the environmental impact of agrochemicals.


Assuntos
Sistemas de Liberação de Medicamentos , Nanomedicina , Nanopartículas/química , Praguicidas/química , Animais , Portadores de Fármacos/química , Composição de Medicamentos , Humanos
10.
ACS Appl Nano Mater ; 3(8): 8037-8051, 2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-33969278

RESUMO

The solid tumor microenvironment (TME) poses a significant structural and biochemical barrier to immunotherapeutic agents. To address the limitations of tumor penetration and distribution, and to enhance antitumor efficacy of immunotherapeutics, we present here an autonomous active microneedle (MN) system for the direct intratumoral (IT) delivery of a potent immunoadjuvant, cowpea mosaic virus nanoparticles (CPMV) in vivo. In this active delivery system, magnesium (Mg) microparticles embedded into active MNs react with the interstitial fluid in the TME, generating a propulsive force to drive the nanoparticle payload into the tumor. Active delivery of CPMV payload into B16F10 melanomas in vivo demonstrated substantially more pronounced tumor regression and prolonged survival of tumor-bearing mice compared to that of passive MNs and conventional needle injection. Active MN administration of CPMV also enhanced local innate and systemic adaptive antitumor immunity. Our approach represents an elaboration of conventional CPMV in situ vaccination, highlighting substantial immune-mediated antitumor effects and improved therapeutic efficacy that can be achieved through an active and autonomous delivery system-mediated CPMV in situ vaccination.

11.
Nat Nanotechnol ; 14(7): 712-718, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31110265

RESUMO

Large doses of chemical pesticides are required to achieve effective concentrations in the rhizosphere, which results in the accumulation of harmful residues. Precision farming is needed to improve the efficacy of pesticides, but also to avoid environmental pollution, and slow-release formulations based on nanoparticles offer one solution. Here, we tested the mobility of synthetic and virus-based model nanopesticides by combining soil column experiments with computational modelling. We found that the tobacco mild green mosaic virus and cowpea mosaic virus penetrate soil to a depth of at least 30 cm, and could therefore deliver nematicides to the rhizosphere, whereas the Physalis mosaic virus remains in the first 4 cm of soil and would be more useful for the delivery of herbicides. Our experiments confirm that plant viruses are superior to synthetic mesoporous silica nanoparticles and poly(lactic-co-glycolic acid) for the delivery and controlled release of pesticides, and could be developed as the next generation of pesticide delivery systems.


Assuntos
Agricultura/métodos , Preparações de Ação Retardada/metabolismo , Vírus do Mosaico/metabolismo , Praguicidas/metabolismo , Microbiologia do Solo , Comovirus/metabolismo , Nanopartículas/metabolismo , Solo/química , Vírus do Mosaico do Tabaco/metabolismo , Tymovirus/metabolismo
12.
Macromol Biosci ; 19(5): e1800407, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30721575

RESUMO

The development of plant viral nanoparticles (VNP) loaded with different molecular versions of a photodynamic drug is described. Specifically, tobacco mosaic virus (TMV) and tobacco mild green mosaic virus (TMGMV) are developed as drug carriers that encapsulate the monocationic, dicationic, tricationic, and tetracationic versions of a porphyrin-based photosensitizer drug (Zn-Por). While TMV has been extensively explored for various nanotechnology applications, this is the first study investigating TMGMV for medical applications. Light-activated cancer cell killing of Zn-Por-loaded VNPs is studied in vitro using melanoma and cervical cancer models. Native and nucleolin-targeted VNP drug carriers are developed and their efficacy assessed. A fivefold increase in cancer cell killing is observed using nucleolin-targeted TMV loaded with tricationic Zn-Por and displaying the nucleolin-specific F3 peptide.


Assuntos
Melanoma Experimental/tratamento farmacológico , Metaloporfirinas , Nanopartículas , Fotoquimioterapia , Vírus do Mosaico do Tabaco/química , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Metaloporfirinas/química , Metaloporfirinas/farmacologia , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico
13.
Langmuir ; 35(1): 203-211, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30576145

RESUMO

The design of versatile tools to improve cell targeting and drug delivery in medicine has become increasingly pertinent to nanobiotechnology. Biological and inorganic nanocarrier drug delivery systems are being explored, showing advantages and disadvantages in terms of cell targeting and specificity, cell internalization, efficient payload delivery, and safety profiles. Combining the properties of a biological coating on top of an inorganic nanocarrier, we hypothesize that this hybrid system would improve nanoparticle-cell interactions, resulting in enhanced cell targeting and uptake properties compared to the bare inorganic nanocarrier. Toward this goal, we engineered a hierarchical assembly featuring the functionalization of cargo-loaded mesoporous silica nanoparticles (MSNPs) with tobacco mosaic virus (TMV) as a biological coating. The MSNP functions as a delivery system because the porous structure enables high therapeutic payload capacity, and TMV serves as a biocompatible coating to enhance cell interactions. The resulting MSNP@TMV nanohybrids have a wool-ball-like appearance and demonstrate enhanced cell uptake, hence cargo delivery properties. The MSNP@TMV have potential for medical applications such as drug delivery, contrast agent imaging, and immunotherapy.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Dióxido de Silício/química , Vírus do Mosaico do Tabaco/química , Carbocianinas/química , Liberação Controlada de Fármacos , Corantes Fluorescentes/química , Células HeLa , Humanos , Microscopia Confocal , Porosidade , Rodaminas/química
14.
Biomacromolecules ; 18(12): 4141-4153, 2017 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-29144726

RESUMO

Platform technologies based on plant virus nanoparticles (VNPs) and virus-like particles (VLPs) are attracting the attention of researchers and clinicians because the particles are biocompatible, biodegradable, noninfectious in mammals, and can readily be chemically and genetically engineered to carry imaging agents and drugs. When the Physalis mottle virus (PhMV) coat protein is expressed in Escherichia coli, the resulting VLPs are nearly identical to the viruses formed in vivo. Here, we isolated PhMV-derived VLPs from ClearColi cells and carried out external and internal surface modification with fluorophores using reactive lysine-N-hydroxysuccinimide ester and cysteine-maleimide chemistries, respectively. The uptake of dye-labeled particles was tested in a range of cancer cells and monitored by confocal microscopy and flow cytometry. VLPs labeled internally on cysteine residues were taken up with high efficiency by several cancer cell lines and were colocalized with the endolysosomal marker LAMP-1 within 6 h, whereas VLPs labeled externally on lysine residues were taken up with lower efficiency, probably reflecting differences in surface charge and the propensity to bind to the cell surface. The infusion of dye and drug molecules into the cavity of the VLPs revealed that the photosensitizer (PS), Zn-EpPor, and the drugs crystal violet, mitoxantrone (MTX), and doxorubicin (DOX) associated stably with the carrier via noncovalent interactions. We confirmed the cytotoxicity of the PS-PhMV and DOX-PhMV particles against prostate cancer, ovarian and breast cancer cell lines, respectively. Our results show that PhMV-derived VLPs provide a new platform technology for the delivery of imaging agents and drugs, with preferential uptake into cancer cells. These particles could therefore be developed as multifunctional tools for cancer diagnosis and therapy.


Assuntos
Portadores de Fármacos/química , Indicadores e Reagentes/química , Nanopartículas/química , Preparações Farmacêuticas/química , Tymovirus/química , Animais , Linhagem Celular , Linhagem Celular Tumoral , Doxorrubicina/química , Células HeLa , Humanos , Lisina/química , Maleimidas/química , Camundongos , Mitoxantrona/química , Neoplasias/diagnóstico , Neoplasias/diagnóstico por imagem , Fármacos Fotossensibilizantes/química , Células RAW 264.7
15.
ACS Nano ; 11(5): 4719-4730, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28345874

RESUMO

Plant parasitic nematodes are a major burden to the global agricultural industry, causing a $157 billion loss each year in crop production worldwide. Effective treatment requires large doses of nematicides to be applied, putting the environment and human health at risk. Challenges are to treat nematodes that are located deep within the soil, feeding on the roots of plants. To attack the problem at its roots, we propose the use of tobacco mild green mosaic virus (TMGMV), an EPA-approved herbicide as a carrier to deliver nematicides. TMGMV self-assembles into a 300 × 18 nm soft matter nanorod with a 4 nm-wide hollow channel. This plant virus is comprised of 2130 identical coat protein subunits, each of which displays solvent-exposed carboxylate groups from Glu/Asp as well as Tyr side chains, enabling the functionalization of the carrier with cargo. We report (1) the successful formulation and characterization of TMGMV loaded with ∼1500 copies of the anthelmintic drug crystal violet (CV), (2) the bioavailability and treatment efficacy of CVTMGMV vs CV to nematodes in liquid cultures, and (3) the superior soil mobility of CVTMGMV compared to free CV.


Assuntos
Agentes de Controle Biológico/administração & dosagem , Nematoides/efeitos dos fármacos , Agricultura , Animais , Disponibilidade Biológica , Sistemas de Liberação de Medicamentos , Praguicidas , RNA Viral , Vírus do Mosaico do Tabaco/genética , Vírus do Mosaico do Tabaco/patogenicidade
16.
J Phys Chem B ; 120(26): 6120-9, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27045770

RESUMO

Nanoparticle-based technologies, including platforms derived from plant viruses, hold great promise for targeting and delivering cancer therapeutics to solid tumors by overcoming dose-limiting toxicities associated with chemotherapies. A growing body of data indicates advantageous margination and penetration properties of high aspect-ratio nanoparticles, which enhance payload delivery, resulting in increased efficacy. Our lab has demonstrated that elongated rod-shaped and filamentous macromolecular nucleoprotein assemblies from plant viruses have higher tissue diffusion rates than spherical particles. In this study, we developed a mathematical model to quantify diffusion and uptake of tobacco mosaic virus (TMV) in a spheroid system approximating a capillary-free segment of a solid tumor. Model simulations predict TMV concentration distribution with time in a tumor spheroid for different sizes and cell densities. From simulations of TMV concentration distribution, we can quantify the effect of TMV aspect ratio (e.g., nanorod length-to-width) with and without cellular uptake by modulated surface chemistry. This theoretical analysis can be applied to other viral or nonviral delivery systems to complement the experimental development of the next generation of nanotherapeutics.


Assuntos
Transporte Biológico , Portadores de Fármacos/metabolismo , Nanotubos , Neoplasias/metabolismo , Neoplasias/terapia , Vírus do Mosaico do Tabaco/metabolismo , Contagem de Células , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Simulação por Computador , Difusão , Humanos , Modelos Biológicos , Propriedades de Superfície , Distribuição Tecidual
17.
Bioconjug Chem ; 26(2): 262-269, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25611133

RESUMO

Molecular imaging approaches and targeted drug delivery hold promise for earlier detection of diseases and treatment with higher efficacy while reducing side effects, therefore increasing survival rates and quality of life. Virus-based nanoparticles are a promising platform because their scaffold can be manipulated both genetically and chemically to simultaneously display targeting ligands while carrying payloads for diagnosis or therapeutic intervention. Here, we displayed a 12-amino-acid peptide ligand, GE11 (YHWYGYTPQNVI), on nanoscale filaments formed by the plant virus potato virus X (PVX). Bioconjugation was used to produce fluorescently labeled PVX-GE11 filaments targeted toward the epidermal growth factor receptor (EGFR). Cell detection and imaging was demonstrated using human skin epidermoid carcinoma, colorectal adenocarcinoma, and triple negative breast cancer cell lines (A-431, HT-29, MDA-MB-231), all of which upregulate EGFR to various degrees. Nonspecific uptake in ductal breast carcinoma (BT-474) cells was not observed. Furthermore, co-culture experiments with EGFR(+) cancer cells and macrophages indicate successful targeting and partitioning toward the cancer cells. This study lays a foundation for the development of EGFR-targeted filaments delivering contrast agents for imaging and diagnosis, and/or toxic payloads for targeted drug delivery.


Assuntos
Receptores ErbB/metabolismo , Nanopartículas , Neoplasias/diagnóstico , Peptídeos , Vírus de Plantas/química , Sequência de Aminoácidos , Linhagem Celular Tumoral , Diagnóstico por Imagem , Humanos , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Peptídeos/química , Peptídeos/metabolismo , Vírus de Plantas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA