Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 35(51): e2305255, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37541432

RESUMO

Stimuli-responsive nanomaterials have the potential to improve the performance and overcome existing barriers of conventional nanotherapeutics. Molecular cooperativity design in stimuli-responsive nanomedicine can amplify physiological signals, enabling a cooperative response for improved diagnostic and therapeutic precision. Previously, this work reported an ultra-pH-sensitive polymer, PEG-b-PC7A, that possesses innate immune activating properties by binding to the stimulator of interferon genes (STING) through polyvalent phase condensation. This interaction enhances STING activation and synergizes with the endogenous STING ligand for robust cancer immunotherapy. Despite its successes in innate immune activation, the fundamental physicochemical and pH-responsive properties of PC7A require further investigation. Here, this study elucidates the protonation cooperativity driven by the phase transition of PC7A copolymer. The highly cooperative system displays an "all-or-nothing" proton distribution between highly charged unimer (all) and neutral micelle (nothing) states without gradually protonated intermediates. The binary protonation behavior is further illustrated in pH-precision-controlled release of a representative anticancer drug, ß-lapachone, by PC7A micelles over a noncooperative PE5A polymer. Furthermore, the bimodal distribution of protons is represented by a high Hill coefficient (nH  > 9), featuring strong positive cooperativity. This study highlights the nanoscale pH cooperativity of an immune activating polymer, providing insights into the physicochemical characterization and design parameters for future nanotherapeutics development.


Assuntos
Antineoplásicos , Proteínas de Membrana , Nanoestruturas , Concentração de Íons de Hidrogênio , Micelas , Transição de Fase , Polímeros/química , Proteínas de Membrana/agonistas , Proteínas de Membrana/metabolismo
2.
Nat Commun ; 13(1): 4981, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36068198

RESUMO

Lactate is a key metabolite produced from glycolytic metabolism of glucose molecules, yet it also serves as a primary carbon fuel source for many cell types. In the tumor-immune microenvironment, effect of lactate on cancer and immune cells can be highly complex and hard to decipher, which is further confounded by acidic protons, a co-product of glycolysis. Here we show that lactate is able to increase stemness of CD8+ T cells and augments anti-tumor immunity. Subcutaneous administration of sodium lactate but not glucose to mice bearing transplanted MC38 tumors results in CD8+ T cell-dependent tumor growth inhibition. Single cell transcriptomics analysis reveals increased proportion of stem-like TCF-1-expressing CD8+ T cells among intra-tumoral CD3+ cells, a phenotype validated by in vitro lactate treatment of T cells. Mechanistically, lactate inhibits histone deacetylase activity, which results in increased acetylation at H3K27 of the Tcf7 super enhancer locus, leading to increased Tcf7 gene expression. CD8+ T cells in vitro pre-treated with lactate efficiently inhibit tumor growth upon adoptive transfer to tumor-bearing mice. Our results provide evidence for an intrinsic role of lactate in anti-tumor immunity independent of the pH-dependent effect of lactic acid, and might advance cancer immune therapy.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Animais , Linhagem Celular Tumoral , Glicólise , Ácido Láctico/metabolismo , Camundongos , Neoplasias/patologia , Microambiente Tumoral
3.
J Immunother Cancer ; 10(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35623658

RESUMO

BACKGROUND: Cancer vaccines are able to achieve tumor-specific immune editing in early-phase clinical trials. However, the infiltration of cytotoxic T cells into immune-deserted tumors is still a major limiting factor. An optimized vaccine approach to induce antigen-specific T cells that can perform robust tumor infiltration is important to accelerate their clinical translation. We previously developed a STING-activating PC7A nanovaccine that produces a strong anti-tumor T cell response on subcutaneous injection. This study systematically investigated the impact of administration methods on the performance of nanovaccines. METHODS: Tumor growth inhibition by intratumoral delivery and subcutaneous delivery of nanovaccine was investigated in TC-1 human papillomavirus-induced cancer model and B16-OVA melanoma model. Nanovaccine distribution in vivo was detected by clinical camera imaging, systemic T cell activation and tumor infiltration were tested by in vivo cytotoxicity killing assay and flow cytometry. For mechanism analysis, T cell recruitment was investigated by in vivo migration blocking assay, multiplex chemokine array, flow cytometry, RT-qPCR, chemotaxis assay and gene knockout mice. RESULTS: Nanovaccine administration was found to alter T cell production and infiltration in tumors. Intratumoral delivery of nanovaccines displayed superior antitumor effects in multiple tumor models compared with subcutaneous delivery. Mechanistic investigation revealed that intratumoral administration of the nanovaccine significantly increased the infiltration of antigen-specific T cells in TC-1 tumors, despite the lower systemic levels of T cells compared with subcutaneous injection. The inhibition of tumor growth by nanovaccines is primarily dependent on CD8+ cytotoxic T cells. Nanovaccine accumulation in tumors upregulates CXCL9 expression in myeloid cells in a STING dependent manner, leading to increased recruitment of IFNγ-expressing CD8+ T cells from the periphery, and IFNγ reciprocally stimulates CXCL9 expression in myeloid cells, resulting in positive feedback between myeloid-CXCL9 and T cell-IFNγ to promote T cell recruitment. However, the STING agonist alone could not sustain this effect in the presence of a systemic deficiency in antigen-specific T cells. CONCLUSIONS: Our results demonstrate that intratumoral administration of PC7A nanovaccine achieved stronger antitumor immunity and efficacy over subcutaneous injection. These data suggest intratumoral administration should be included in the therapeutic design in the clinical use of nanovaccine.


Assuntos
Vacinas Anticâncer , Melanoma Experimental , Nanopartículas , Animais , Linfócitos T CD8-Positivos , Humanos , Fatores Imunológicos/farmacologia , Imunoterapia/métodos , Melanoma Experimental/terapia , Camundongos
4.
Am J Ind Med ; 65(5): 357-370, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35235683

RESUMO

BACKGROUND: Although racial and ethnic identities are associated with a multitude of disparate medical outcomes, surveillance of these subpopulations in the occupational clinic setting could benefit enormously from a more detailed and nuanced recognition of racial and ethnic identity. METHODS: The research group designed a brief questionnaire to capture several dimensions of this identity and collected data from patients seen for work-related conditions in four occupational medicine clinics from May 2019 through March 2020. Responses were used to calculate the sensitivity and specificity of extant racial/ethnic identity data within our electronic health records system, and were compared to participants' self-reported industry and occupation, coded according to North American Industry Classification System and Standard Occupational Classification System listings. RESULTS: Our questionnaire permitted collection of data that defined our patients' specific racial/ethnic identity with far greater detail, identified patients with multiple ethnic identities, and elicited their preferred language. Response rate was excellent (94.2%, n = 773). Non-White participants frequently selected a racial/ethnic subcategory (78.1%-92.2%). Using our race/ethnicity data as a referent, the electronic health record (EHR) had a high specificity (>87.1%), widely variable sensitivity (11.8%-82.2%), and poorer response rates (75.1% for race, 82.5% for ethnicity, as compared to 93.8% with our questionnaire). Additional analyses revealed some industries and occupations disproportionately populated by patients of particular racial/ethnic identities. CONCLUSIONS: Our project demonstrates the usefulness of a questionnaire which more effectively identifies racial/ethnic subpopulations in an occupational medicine clinic, permitting far more detailed characterization of their occupations, industries, and diagnoses.


Assuntos
Etnicidade , Ocupações , Humanos , Estados Unidos
6.
Nat Biomed Eng ; 5(5): 455-466, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33558734

RESUMO

The stimulator of interferon genes (STING) is an endoplasmic reticulum transmembrane protein that is a target of therapeutics for infectious diseases and cancer. However, early-phase clinical trials of small-molecule STING agonists have shown limited antitumour efficacy and dose-limiting toxicity. Here, we show that a polyvalent STING agonist-a pH-sensitive polymer bearing a seven-membered ring with a tertiary amine (PC7A)-activates innate-immunity pathways through the polymer-induced formation of STING-PC7A condensates. In contrast to the natural STING ligand 2',3'-cyclic-GMP-AMP (cGAMP), PC7A stimulates the prolonged production of pro-inflammatory cytokines by binding to a non-competitive STING surface site that is distinct from the cGAMP binding pocket. PC7A induces antitumour responses that are dependent on STING expression and CD8+ T-cell activity, and the combination of PC7A and cGAMP led to synergistic therapeutic outcomes (including the activation of cGAMP-resistant STING variants) in mice bearing subcutaneous tumours and in resected human tumours and lymph nodes. The activation of the STING pathway through polymer-induced STING condensation may offer new therapeutic opportunities.


Assuntos
Imunidade Inata , Proteínas de Membrana/agonistas , Neoplasias/terapia , Nucleotídeos Cíclicos/administração & dosagem , Polímeros/administração & dosagem , Animais , Linfócitos T CD8-Positivos/metabolismo , Feminino , Células HEK293 , Células HeLa , Humanos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos , Neoplasias/imunologia , Nucleotídeos Cíclicos/farmacologia , Polímeros/farmacologia , Células THP-1
7.
Adv Healthc Mater ; 10(4): e2000549, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32431046

RESUMO

Export of lactic acid from glycolytic cancer cells to the extracellular tumor milieu has been reported to enhance tumor growth and suppress antitumor immunity. In this study, a pH-activatable nanodrug is reported for tumor-targeted inhibition of monocarboxylate transporter-1 (MCT1) that reverses lactic acid-induced tumor immunosuppression. The nanodrug is composed of an MCT1 inhibitor (AZD3965) loaded inside the ultra-pH-sensitive nanoparticles (AZD-UPS NPs). AZD-UPS NP is produced by a microfluidics method with improved drug loading efficiency and optimal nanoparticle size over sonication methods. The nanodrug remains as intact micelles at pH 7.4 but rapidly disassembles and releases payload upon exposure to acidic pH. When combined with anti-PD-1 therapy, AZD-UPS NP leads to potent tumor growth inhibition and increases survival in two tumor models over oral administration of AZD3965 at dramatically reduced dose (>200-fold). Safety evaluations demonstrate reduced drug distribution in heart and liver tissues with decrease in toxic biomarkers such as cardiac troponin by the nanodrug. Increased T-cell infiltration and reduced exhaustive PD1+ Tim3+ T cells are found in tumors. These data illustrate that tumor-targeted inhibition of MCT1 can reverse the immune suppressive microenvironment of solid tumors for increased safety and antitumor efficacy of cancer immunotherapy.


Assuntos
Nanopartículas , Linfócitos T , Linhagem Celular Tumoral , Imunoterapia , Ácido Láctico
8.
J Control Release ; 329: 353-360, 2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33301836

RESUMO

Cancer vaccines hold great promise to produce antigen-specific T cell immunity for personalized therapy of cancer. Previously, we reported an ultra-pH-sensitive nanoparticle, PC7A, capable of priming an efficacious immune response without significant systemic toxicity. Despite the early success, the relationship between antigen properties and encapsulation efficiency for downstream immune activation remains poorly understood. In this study, we investigated a small library of melanoma antigens and the effects of several formulation methods on the efficiency of peptide loading inside PC7A nanoparticles. Results show loading efficiency is not highly dependent on the formulation methods, but instead mainly driven by the peptide antigen properties. In particular, we identified a phase transition event, namely the folding of antigenic peptides from random coils to α-helical structure, is important for antigen loading inside PC7A nanoparticles. Mutation of a peptide that abrogates the formation of helical structure resulted in poor loading efficiency. Antitumor efficacy studies in melanoma-bearing mice demonstrate the importance of peptide loading in vaccine-induced antitumor immunity. This study highlights the contribution of phase transition of peptide antigens on vaccine formulation in order to make widespread use of personalized nanoparticle vaccines feasible.


Assuntos
Vacinas Anticâncer , Nanopartículas , Animais , Antígenos , Células Dendríticas , Camundongos , Camundongos Endogâmicos C57BL
9.
Nat Commun ; 11(1): 5828, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33203928

RESUMO

Stimuli-sensitive nanomaterials with cooperative response are capable of converting subtle and gradual biological variations into robust outputs to improve the precision of diagnostic or therapeutic outcomes. In this study, we report the design, synthesis and characterization of a series of degradable ultra-pH sensitive (dUPS) polymers that amplify small acidic pH changes to efficacious therapeutic outputs. A hydrolytically active polycarbonate backbone is used to construct the polymer with pH-dependent degradation kinetics. One dUPS polymer, PSC7A, can achieve activation of the stimulator of interferon genes and antigen delivery upon endosomal pH activation, leading to T cell-mediated antitumor immunity. While a non-degradable UPS polymer induces granulomatous inflammation that persists over months at the injection site, degradable PSC7A primes a transient acute inflammatory response followed by polymer degradation and complete tissue healing. The improved therapeutic window of the dUPS polymers opens up opportunities in pH-targeted drug and protein therapy.


Assuntos
Vacinas Anticâncer/farmacocinética , Nanopartículas/química , Cimento de Policarboxilato/química , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacocinética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Concentração de Íons de Hidrogênio , Imunoterapia/métodos , Espectroscopia de Ressonância Magnética , Melanoma/terapia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/uso terapêutico , Cimento de Policarboxilato/metabolismo , Polietilenoglicóis/química , Polímeros/síntese química , Polímeros/química , Polímeros/metabolismo , Linfócitos T/imunologia
10.
Adv Drug Deliv Rev ; 158: 63-72, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32882321

RESUMO

Precise spatiotemporal control of molecular transport is vital to functional physiological systems. Nature evolved to apply macromolecular cooperativity to achieve precision over systemic delivery of important molecules. In drug delivery, conventional nanocarriers employ inert materials and rely on passive accumulation for tissue targeting and diffusion for drug release. Early clinical studies show these nanodrugs have not delivered the anticipated impact on therapy. Inspired by nature, we propose a design principle that incorporates nanoscale cooperativity and phase transition to sense and amplify physiological signals to improve the therapeutic outcome. Using ultra-pH-sensitive (UPS) nanoparticles as an example, we demonstrate how all-or-nothing protonation cooperativity during micelle assembly/disassembly can be exploited to increase dose accumulation and achieve rapid drug release in acidic microenvironments. In a separate study, we show the effectiveness of a single polymer composition to accomplish cytosolic delivery of tumor antigens with activation of stimulator of interferon genes (STING) in lymph node-resident dendritic cells for cancer immunotherapy. Molecular cooperativity is a hallmark of nanobiology that offers a valuable strategy to functionalize nanomedicine systems to achieve precision medicine.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Micelas , Nanopartículas/química , Neoplasias/tratamento farmacológico , Medicina de Precisão/métodos , Antineoplásicos Imunológicos/uso terapêutico , Relação Dose-Resposta a Droga , Liberação Controlada de Fármacos , Humanos , Concentração de Íons de Hidrogênio
11.
Acc Chem Res ; 52(6): 1485-1495, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31067025

RESUMO

Electronic transistors have revolutionized the fields of microelectronics, computers, and mobile devices. Their ability to digitize electronic signals allows high fidelity data transfer as well as formation of logic gates. Inspired by electronic transistors, transistor-like organic materials have been under intensive investigation to amplify biological signals in a broad range of applications such as biosensing, diagnostic imaging, and therapeutic delivery. This Account highlights the inception and implementation of a "proton transistor" nanoparticle that can digitize acidotic pH signals in biological systems. Similar to electronic transistors, the ultra-pH-sensitive (UPS) nanoparticles derive their binary threshold response from phase separation phenomena. Hydrophobic micellization drives nanophase separation from unimers to aggregated polymeric micelles, which is responsible for the all-or-nothing proton distribution between the micelle and unimer states. Depending on the assembly status, conjugated fluorophores are quenched (micelle state) or freely fluoresce (solution unimer state) allowing robust detection of the phase transition behavior across a narrow pH range. Based on this mechanistic insight, we created a UPS nanoparticle library encompassing a broad physiological pH range from 4.0 to 7.4. For biological applications, we engineered a barcode-like nanosensor capable of digitizing multiple pH signals at a single organelle resolution in live cells. The barcode system allowed easy identification of mutant Kirsten rat sarcoma viral oncogene (KRAS), a common mutation involved in tumorigenesis, which leads to rapid cellular proliferation, as the protein driver for accelerated organelle acidification and lysosome catabolism in a broad set of isogenic as well as heterogeneous cancer cell lines. Adoption of the technology to an ON-OFF/Always-ON design allowed the quantification of proton flux across the membranes of endocytic organelles. For medical applications, we demonstrate the ability to achieve binary detection of solid cancers with clear tumor margin delineation by near-infrared fluorescence imaging. Image-guided resection of head/neck and breast tumors resulted in significantly improved long-term survival over white light or tumor debulking surgeries in tumor-bearing mice, catapulting the clinical evaluation of the UPS nanosensor in cancer patients. This Account serves as the first comprehensive summary of the molecular mechanism and biological applications of the digital pH threshold sensors. Building on the concept of cooperative phase transition behavior, we hope this Account will promote the rational design and development of additional transistor-like chemical sensors to digitize analog biological signals.


Assuntos
Corantes Fluorescentes/química , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Polímeros/química , Animais , Fluorescência , Fluorometria/métodos , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Micelas , Organelas/química , Transição de Fase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA