Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(3): e0299541, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38551930

RESUMEN

The activities of the phospholipase C gamma (PLCγ) 1 and 2 enzymes are essential for numerous cellular processes. Unsurprisingly, dysregulation of PLCγ1 or PLCγ2 activity is associated with multiple maladies including immune disorders, cancers, and neurodegenerative diseases. Therefore, the modulation of either of these two enzymes has been suggested as a therapeutic strategy to combat these diseases. To aid in the discovery of PLCγ family enzyme modulators that could be developed into therapeutic agents, we have synthesized a high-throughput screening-amenable micellular fluorogenic substrate called C16CF3-coumarin. Herein, the ability of PLCγ1 and PLCγ2 to enzymatically process C16CF3-coumarin was confirmed, the micellular assay conditions were optimized, and the kinetics of the reaction were determined. A proof-of-principle pilot screen of the Library of Pharmacologically Active Compounds 1280 (LOPAC1280) was performed. This new substrate allows for an additional screening methodology to identify modulators of the PLCγ family of enzymes.


Asunto(s)
Colorantes Fluorescentes , Fosfatidilinositoles , Fosfolipasa C gamma , Hidrolasas Diéster Fosfóricas , Cumarinas/farmacología , Fosfolipasas de Tipo C
2.
SLAS Discov ; 28(4): 170-179, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36933698

RESUMEN

A rare coding variant in PLCγ2 (P522R) expressed in microglia induces a mild activation of enzymatic activity when compared to wild-type. This mutation is reported to be protective against the cognitive decline associated with late-onset Alzheimer's disease (LOAD) and therefore, activation of wild-type PLCγ2 has been suggested as a potential therapeutic target for the prevention and treatment of LOAD. Additionally, PLCγ2 has been associated with other diseases such as cancer and some autoimmune disorders where mutations with much greater increases in PLCγ2 activity have been identified. Here, pharmacological inhibition may provide a therapeutic effect. In order to facilitate our investigation of the activity of PLCγ2, we developed an optimized fluorogenic substrate to monitor enzymatic activity in aqueous solution. This was accomplished by first exploring the spectral properties of various "turn-on" fluorophores. The most promising turn-on fluorophore was incorporated into a water-soluble PLCγ2 reporter substrate, which we named C8CF3-coumarin. The ability of PLCγ2 to enzymatically process C8CF3-coumarin was confirmed, and the kinetics of the reaction were determined. Reaction conditions were optimized to identify small molecule activators, and a pilot screen of the Library of Pharmacologically Active Compounds 1280 (LOPAC1280) was performed with the goal of identifying small molecule activators of PLCγ2. The optimized screening conditions allowed identification of potential PLCγ2 activators and inhibitors, thus demonstrating the feasibility of this approach for high-throughput screening.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Colorantes Fluorescentes , Fosfolipasa C gamma/genética , Ensayos Analíticos de Alto Rendimiento , Cumarinas
3.
Life Sci Space Res (Amst) ; 35: 88-104, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36336374

RESUMEN

As humanity begins to venture further into space, approaches to better protect astronauts from the hazards found in space need to be developed. One particular hazard of concern is the complex radiation that is ever present in deep space. Currently, it is unlikely enough spacecraft shielding could be launched that would provide adequate protection to astronauts during long-duration missions such as a journey to Mars and back. In an effort to identify other means of protection, prophylactic radioprotective drugs have been proposed as a potential means to reduce the biological damage caused by this radiation. Unfortunately, few radioprotectors have been approved by the FDA for usage and for those that have been developed, they protect normal cells/tissues from acute, high levels of radiation exposure such as that from oncology radiation treatments. To date, essentially no radioprotectors have been developed that specifically counteract the effects of chronic low-dose rate space radiation. This review highlights how high-throughput screening (HTS) methodologies could be implemented to identify such a radioprotective agent. Several potential target, pathway, and phenotypic assays are discussed along with potential challenges towards screening for radioprotectors. Utilizing HTS strategies such as the ones proposed here have the potential to identify new chemical scaffolds that can be developed into efficacious radioprotectors that are specifically designed to protect astronauts during deep space journeys. The overarching goal of this review is to elicit broader interest in applying drug discovery techniques, specifically HTS towards the identification of radiation countermeasures designed to be efficacious towards the biological insults likely to be encountered by astronauts on long duration voyages.


Asunto(s)
Radiación Cósmica , Protectores contra Radiación , Vuelo Espacial , Humanos , Radiación Cósmica/efectos adversos , Ensayos Analíticos de Alto Rendimiento , Astronautas , Nave Espacial , Protectores contra Radiación/farmacología , Dosis de Radiación
4.
Front Immunol ; 13: 816761, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250995

RESUMEN

Although CAR T cell therapies have proven to be effective in treating hematopoietic cancers, their abilities to regress solid tumors have been less encouraging. Mechanisms to explain these disparities have focused primarily on differences in cancer cell heterogeneity, barriers to CAR T cell penetration of solid tumors, and immunosuppressive microenvironments. To evaluate the contributions of immunosuppressive tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) on CAR T cell efficacies, we have exploited the ability of a folate-targeted Toll-like receptor 7 agonist (FA-TLR7-1A) to specifically reactivate TAMs and MDSCs from an immunosuppressive to pro-inflammatory phenotype without altering the properties of other immune cells. We report here that FA-TLR7-1A significantly augments standard CAR T cell therapies of 4T1 solid tumors in immune competent mice. We further show that co-administration of the FA-TLR7-1A with the CAR T cell therapy not only repolarizes TAMs and MDSCs from an M2-like anti-inflammatory to M1-like pro-inflammatory phenotype, but also enhances both CAR T cell and endogenous T cell accumulation in solid tumors while concurrently increasing their states of activation. Because analogous myeloid cells in healthy tissues ar not altered by administration of FA-TLR7-1A, no systemic activation of the immune system nor accompanying weight loss is observed. These data argue that immunosuppressive myeloid cells contribute prominently to the failure of CAR T cells to eradicate solid tumors and suggest that methods to reprogram tumor associated myeloid cells to a more inflammatory phenotype could significantly augment the potencies of CAR T cell therapies.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias , Animales , Inmunoterapia Adoptiva , Ratones , Células Mieloides , Receptor Toll-Like 7 , Microambiente Tumoral
5.
J Exp Med ; 218(10)2021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34436509

RESUMEN

To egress from its erythrocyte host, the malaria parasite, Plasmodium falciparum, must destabilize the erythrocyte membrane by activating an erythrocyte tyrosine kinase. Because imatinib inhibits erythrocyte tyrosine kinases and because imatinib has a good safety profile, we elected to determine whether coadministration of imatinib with standard of care (SOC) might be both well tolerated and therapeutically efficacious in malaria patients. Patients with uncomplicated P. falciparum malaria from a region in Vietnam where one third of patients experience delayed parasite clearance (DPC; continued parasitemia after 3 d of therapy) were treated for 3 d with either the region's SOC (40 mg dihydroartemisinin + 320 mg piperaquine/d) or imatinib (400 mg/d) + SOC. Imatinib + SOC-treated participants exhibited no increase in number or severity of adverse events, a significantly accelerated decline in parasite density and pyrexia, and no DPC. Surprisingly, these improvements were most pronounced in patients with the highest parasite density, where serious complications and death are most frequent. Imatinib therefore appears to improve SOC therapy, with no obvious drug-related toxicities.


Asunto(s)
Antimaláricos/efectos adversos , Antimaláricos/uso terapéutico , Mesilato de Imatinib/uso terapéutico , Malaria Falciparum/tratamiento farmacológico , Adolescente , Adulto , Artemisininas/uso terapéutico , Quimioterapia Combinada , Fiebre/tratamiento farmacológico , Fiebre/microbiología , Humanos , Mesilato de Imatinib/efectos adversos , Malaria Falciparum/parasitología , Persona de Mediana Edad , Quinolinas/uso terapéutico , Resultado del Tratamiento , Vietnam , Adulto Joven
6.
Molecules ; 26(12)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205289

RESUMEN

The inadvertent severing of a ureter during surgery occurs in as many as 4.5% of colorectal surgeries. To help prevent this issue, several near-infrared (NIR) dyes have been developed to assist surgeons with identifying ureter location. However, the majority of these dyes exhibit at least some issue that precludes their widespread usage such as high levels of uptake in other tissues, overlapping emission wavelengths with other NIR dyes used for other fluorescence-guided surgeries, and/or rapid excretion times through the ureters. To overcome these limitations, we have synthesized and characterized the spectral properties and biodistribution of a new series of PEGylated UreterGlow derivatives. The most promising dye, UreterGlow-11 was shown to almost exclusively excrete through the kidneys/ureters with detectable fluorescence observed for at least 12 h. Additionally, while the excitation wavelength is similar to that of other NIR dyes used for cancer resections, the emission is shifted by ~30 nm allowing for discrimination between the different fluorescence-guided surgery probes. In conclusion, these new UreterGlow dyes show promising optical and biodistribution characteristics and are good candidates for translation into the clinic.


Asunto(s)
Abdomen/cirugía , Imagen Óptica/métodos , Espectroscopía Infrarroja Corta/métodos , Uréter/cirugía , Animales , Fluorescencia , Colorantes Fluorescentes/metabolismo , Humanos , Riñón/cirugía , Ratones , Distribución Tisular/fisiología , Uréter/metabolismo
7.
Bioorg Med Chem Lett ; 42: 128091, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33964441

RESUMEN

Activated macrophages contribute prominently to the progression and maintenance of almost all inflammatory and autoimmune diseases. Although non-specific elimination of these phagocytes has been shown to treat animal models of inflammatory disease, the same therapies have been compromised by unacceptable toxicities, because they also kill quiescent macrophages in healthy tissues. In the studies below, we exploit upregulation of folate receptor beta (FRß) on inflammatory (but not resting) macrophages to target a cytotoxic drug selectively to the inflammatory subset of macrophages. Because many of these activated macrophages are nondividing, we also employ verrucarin A as the cytotoxic payload, since it kills both mitotic and nonmitotic cells by blocking protein synthesis. By inserting a redox-sensitive self-immolative linker between the folate and verrucarin A, we further assure that release of unmodified verrucarin A is triggered primarily after internalization by an FRß-positive cell. The resulting folate-verrucarin A conjugate is shown to kill FR-expressing cells in vitro in a manner that can be inhibited by competition with 100-fold excess folic acid. The folate-verrucarin A conjugate is also shown to successfully treat a murine model of inflammatory peritonitis by eliminating inflammatory macrophages without killing other cells in the same peritonitis fluid. Based on this high specificity for inflammatory macrophages, we conclude that folate-verrucarin A warrants continued exploration as a potential therapy for inflammatory and autoimmune diseases in humans.


Asunto(s)
Modelos Animales de Enfermedad , Ácido Fólico/farmacología , Macrófagos/efectos de los fármacos , Peritonitis/tratamiento farmacológico , Tricotecenos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Receptor 2 de Folato/metabolismo , Ácido Fólico/química , Macrófagos/metabolismo , Ratones , Estructura Molecular , Peritonitis/metabolismo , Relación Estructura-Actividad , Tricotecenos/química
8.
Cancer Drug Resist ; 4(1): 223-232, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35582012

RESUMEN

Aim: Ligand-targeted therapeutics are experiencing increasing use for treatment of human diseases due to their ability to concentrate a desired drug at a pathologic site while reducing accumulation in healthy tissues. For many ligand-targeted drug conjugates, a critical aspect of conjugate design lies in engineering release of the therapeutic payload to occur only after its internalization by targeted cells. Because disulfide bond reduction is frequently exploited to ensure intracellular drug release, an understanding of the redox properties of endocytic compartments can be critical to ligand-targeted drug design. While the redox properties of folate receptor trafficking endosomes have been previously reported, little is known about the trafficking of prostate-specific membrane antigen (PSMA), a receptor that is experiencing increasing use for drug targeting in humans. Methods: To obtain this information, we have constructed a PSMA-targeted fluorescence resonance energy transfer pair that reports on disulfide bond reduction by changing fluorescence from red to green. Results: We show here that this reporter exhibits rapid and selective uptake by PSMA-positive cells, and that reduction of its disulfide bond proceeds steadily but incompletely following internalization. The fact that maximal disulfide reduction reaches only ~50%, even after 24 h incubation, suggests that roughly half of the conjugates must traffic through endosomes that display no reducing capacity. Conclusion: As the level of disulfide reduction differs between PSMA trafficked and previously published folate trafficked conjugates, it also follows that not all internalizing receptors are translocated through similar intracellular compartments. Taken together, these data suggest that the efficiency of disulfide bond reduction must be independently analyzed for each receptor trafficking pathway when disulfide bond reduction is exploited for intracellular drug release.

9.
Biophys J ; 119(9): 1749-1759, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33069269

RESUMEN

Single-particle tracking offers a method to interrogate the organization of transmembrane proteins by measuring their mobilities within a cell's plasma membrane. Using this technique, the diffusion characteristics of the Duffy antigen (DARC), glycophorin A, band 3, and GLUT1 were compared under analogous conditions on intact human erythrocyte membranes. Microscopic diffusion coefficients revealed that the vast majority of all four transmembrane proteins exhibit very restricted movement but are not completely immobile. In fact, only 12% of GLUT1 resolved into a highly mobile subpopulation. Macroscopic diffusion coefficients and compartment sizes were also similar for all four proteins, with movements confined to the approximate dimensions of the "corrals" of the cortical spectrin cytoskeleton. Taken together, these data suggest that almost the entire populations of all four transmembrane proteins are immobilized by either the incorporation within large multiprotein complexes or entrapment within the protein network of the cortical spectrin cytoskeleton.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito , Glicoforinas , Difusión , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Transportador de Glucosa de Tipo 1 , Humanos , Espectrina/metabolismo
10.
Arthritis Res Ther ; 21(1): 143, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31174578

RESUMEN

OBJECTIVES: Most therapies for autoimmune and inflammatory diseases either neutralize or suppress production of inflammatory cytokines produced by activated macrophages (e.g., TNFα, IL-1, IL-6, IL-17, GM-CSF). However, no approved therapies directly target this activated subset of macrophages. METHODS: First, we undertook to examine whether the folate receptor beta (FR-ß) positive subpopulation of macrophages, which marks the inflammatory subset in animal models of rheumatoid arthritis, might constitute the prominent population of macrophages in inflamed lesions in humans. Next, we utilized anti-FR-ß monoclonal antibodies capable of mediating antibody-dependent cell cytotoxicity (ADCC) to treat animal models of rheumatoid arthritis and peritonitis. RESULTS: Human tissue samples of rheumatoid arthritis, Crohn's disease, ulcerative colitis, idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, chronic obstructive pulmonary disease, systemic lupus erythematosus, psoriasis, and scleroderma are all characterized by dramatic accumulation of macrophages that express FR-ß, a protein not expressed on resting macrophages or any other healthy tissues. A monoclonal antibody to FR-ß accumulates specifically in inflamed lesions of murine inflammatory disease models and successfully treats such models of rheumatoid arthritis and peritonitis. More importantly, elimination of FR-ß-positive macrophages upon treatment with an anti-FR-ß monoclonal antibody promotes the departure of other immune cells, including T cells, B cells, neutrophils, and dendritic cells from the inflamed lesions. CONCLUSIONS: These data suggest that specific elimination of FR-ß-expressing macrophages may constitute a highly specific therapy for multiple autoimmune and inflammatory diseases and that a recently developed human anti-human FR-ß monoclonal antibody (m909) might contribute to suppression of this subpopulation of macrophages.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Artritis Reumatoide/inmunología , Receptor 2 de Folato/inmunología , Inmunidad Celular , Activación de Macrófagos , Macrófagos/metabolismo , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Macrófagos/inmunología , Macrófagos/patología , Ratones
11.
Nat Commun ; 10(1): 2681, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31213606

RESUMEN

Although chimeric antigen receptor (CAR) T cell therapies have demonstrated considerable success in treating hematologic malignancies, they have simultaneously been plagued by a cytokine release syndrome (CRS) that can harm or even kill the cancer patient. We describe a CAR T cell strategy in which CAR T cell activation and cancer cell killing can be sensitively regulated by adjusting the dose of a low molecular weight adapter that must bridge between the CAR T cell and cancer cell to initiate tumor eradication. By controlling the concentration and dosing schedule of adapter administration, we document two methods that can rapidly terminate (<3 h) a pre-existing CRS-like toxicity and two unrelated methods that can pre-emptively prevent a CRS-like toxicity that would have otherwise occurred. Because all four methods concurrently enhance CAR T cell potency, we conclude that proper use of bispecific adapters could potentially avoid a life-threatening CRS while enhancing CAR T cell tumoricidal activity.


Asunto(s)
Enfermedades del Sistema Inmune/prevención & control , Inmunoterapia Adoptiva/efectos adversos , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Ingeniería Celular/métodos , Línea Celular Tumoral , Citocinas/inmunología , Fluoresceína/metabolismo , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/metabolismo , Humanos , Enfermedades del Sistema Inmune/etiología , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Ratones , Neoplasias/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Síndrome , Linfocitos T/metabolismo , Linfocitos T/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood Cells Mol Dis ; 77: 61-66, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30974390

RESUMEN

Glucose transporter 1 (GLUT1) is one of 13 members of the human equilibrative glucose transport protein family and the only glucose transporter thought to be expressed in human erythrocyte membranes. Although GLUT1 has been shown to be anchored to adducin at the junctional spectrin-actin complex of the membrane through interactions with multiple proteins, whether other populations of GLUT1 also exist in the human erythrocyte membrane has not been examined. Because GLUT1 plays such a critical role in erythrocyte biology and since it comprises 10% of the total membrane protein, we undertook to evaluate the subpopulations of erythrocyte GLUT1 using single particle tracking. By monitoring the diffusion of individual AlexaFluor 488-labeled GLUT1 molecules on the surfaces of intact erythrocytes, we are able to identify three distinct subpopulations of GLUT1. While the mobility of the major subpopulation is similar to that of the anion transporter, band 3, both a more mobile and more anchored subpopulation also exist. From these studies, we conclude that ~65% of GLUT1 resides in similar or perhaps the same protein complex as band 3, while the remaining 1/3rd are either freely diffusing or interacting with other cytoskeletally anchored membrane protein complexes.


Asunto(s)
Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Biomarcadores , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Microscopía Fluorescente , Imagen Molecular , Transporte de Proteínas , Coloración y Etiquetado
13.
Leuk Lymphoma ; 59(12): 2982-2989, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29616859

RESUMEN

Receptor-targeted delivery of imaging and therapeutic agents has emerged as an attractive strategy to diagnosis and treat many diseases including cancer. One of the most well-studied receptors for targeted therapies is the folate receptor (FR) family. FR-α and FR-ß are present on many cancers with little expression in normal tissues; leading to the testing of at least six folate-targeted drugs in human clinical trials for various cancers. However, the expression of FR in blood cancers has not been fully explored with no reports of FR expression in myelomas. Herein, we report the expression of both FR-α and FR-ß on CD138 + plasma cells isolated from patients with multiple myeloma. In addition, all-trans retinoic acid was shown to increase the levels of FR-α and FR-ß in two myeloma cell lines. Altogether, this data suggests that folate-targeted therapies for the treatment of multiple myeloma warrants further investigation.


Asunto(s)
Antineoplásicos/farmacología , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/efectos de los fármacos , Tretinoina/farmacología , Antineoplásicos/uso terapéutico , Médula Ósea/patología , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mieloma Múltiple/tratamiento farmacológico , Células Plasmáticas/metabolismo , Sindecano-1/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Nanomedicine ; 14(3): 1033-1043, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29410110

RESUMEN

Activated macrophages play a key role in the development and maintenance of inflammatory diseases such as atherosclerosis, lupus, psoriasis, rheumatoid arthritis, ulcerative colitis, and many others. These activated macrophages, but not resting or quiescent macrophages highly up-regulate folate receptor beta (FR-ß). This differential expression of FR-ß provides a mechanism to selectively deliver imaging and therapeutic agents utilizing folate as a targeting molecule. In an effort to determine whether inflammatory diseases can be targeted utilizing a folate-linked nanosize carrier, a PEG-coated liposome was prepared that incorporated a folate conjugated PEG that also could transport imaging or therapeutic cargo. We demonstrate that these folate-liposomes specifically bind to folate receptor positive cells and accumulate at sites of inflammation in mouse models of colitis and atherosclerosis. These two animal models show that folate-targeted liposomes could be successfully utilized to deliver fluorescent molecules and an anti-inflammatory drug (betamethasone) for diagnostic and therapeutic applications.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Betametasona/farmacología , Colitis/tratamiento farmacológico , Modelos Animales de Enfermedad , Receptor 2 de Folato/metabolismo , Liposomas/administración & dosificación , Macrófagos Peritoneales/inmunología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Antiinflamatorios/farmacología , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Betametasona/administración & dosificación , Betametasona/química , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Femenino , Ácido Fólico/metabolismo , Regulación de la Expresión Génica , Liposomas/química , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Nanotecnología
15.
Oncotarget ; 9(4): 4485-4495, 2018 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-29435118

RESUMEN

A number of folate receptor (FR) targeted small molecular drugs and monoclonal antibodies have been introduced into clinical trials to treat FR positive cancers. Because the therapeutic efficacy of these drugs depends prominently on the level of FR-α expression on the cancer cells, patients have been commonly selected for FR-targeted therapies based on the intensity of a folate-targeted radioimaging agent. Unfortunately, uptake of such imaging agents can be mediated by both major isoforms of the folate receptor, FR-α and FR-ß. Logically, if the FR positive cell population in a tumor mass is dominated by FR-ß positive macrophages, patients could be selected for therapy that have few FR-expressing cancer cells. Although several IHC studies have examined expression of either FR-α or FR-ß, no study to date has investigated expression of both FR-α and FR-ß in the same tumor mass. Herein, we utilize monoclonal antibodies specific for FR-α (mAb343) and FR-ß (m909) to query each isoform's expression in a range of cancers. We show that lung and pancreatic adenocarcinomas express the full spectrum of FR-α and FR-ß combinations with ~76% of lung adenocarcinomas expressing both FR-α and FR-ß while pancreatic cancers express primarily FR-ß. Thus, while folate-targeted imaging of lung cancer patients might accurately reflect the expression of FR-α on lung cancer cells, imaging of pancreatic cancer patients could mislead a physician into treating a nonresponding patient. Overall, these data suggest that an independent analysis of both FR-α and FR-ß should be obtained to predict the potential efficacy of a folate-targeted drug.

16.
Nanomedicine (Lond) ; 12(20): 2441-2451, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28972462

RESUMEN

AIM: We endeavored to create a folate-targeted liposome (Fol-liposome) that could selectively target areas of inflammation. MATERIALS & METHODS: Fol-liposomes were prepared with encapsulated DiD fluorophore or betamethasone (BM) to image and treat an adjuvant-induced rat model of rheumatoid arthritis. RESULTS: Fol-liposomes selectively accumulated in arthritic rat paws to a greater extent than nontargeted liposomes. When these Fol-liposomes were used to encapsulate BM and administered to arthritic rats, animals exhibited less paw swelling, lower arthritis scores, a reduction in bone erosion, less splenomegaly and better maintenance of body weight when compared with nontreated or nontargeted BM-containing liposome groups. CONCLUSION: Fol-liposomes can selectively deliver imaging and therapeutic agents to sites of inflammation in a rat model of rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Ácido Fólico/química , Ácido Fólico/metabolismo , Liposomas/química , Terapia Molecular Dirigida/métodos , Adyuvantes Inmunológicos/metabolismo , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Liberación de Fármacos , Femenino , Colorantes Fluorescentes/química , Transportadores de Ácido Fólico/metabolismo , Inflamación/tratamiento farmacológico , Macrófagos , Imagen Óptica/métodos , Tamaño de la Partícula , Ratas , Ratas Endogámicas Lew , Propiedades de Superficie
17.
Biomacromolecules ; 18(10): 3082-3088, 2017 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-28863264

RESUMEN

Folate-receptor-positive activated macrophages are critical for the development and maintenance of many chronic inflammatory and autoimmune diseases. Previously, small-molecule folate-targeted conjugates were found to specifically bind to these activated macrophages in vitro and selectively accumulate at sites of inflammation in vivo. While these small-molecule conjugates have shown promise, the use of a folate-targeted, higher cargo capacity nanovehicle may prove superior in delivering imaging or therapeutic agents in vivo. This nanoparticle strategy has been demonstrated in oncology, where targeted dendrimers have shown superior delivery capabilities; however, little research has been pursued in the area of folate-targeted dendrimers for inflammation and autoimmune diseases. Therefore, we endeavored to create a folate-decorated dendrimer to explore its uptake in mouse models of ulcerative colitis and atherosclerosis. We demonstrate that our final poly(ethylene glycol)-coated, acetic-anhydride-capped, folate-targeted poly(amidoamine) dendrimer exhibits no discernible cytotoxicity in vitro, specifically binds to a folate-receptor-expressing macrophage cell line in vitro, and selectively accumulates in areas of inflammation in vivo.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Colitis Ulcerosa/tratamiento farmacológico , Dendrímeros/farmacocinética , Ácido Fólico/análogos & derivados , Anhídridos Acéticos/química , Animales , Línea Celular , Dendrímeros/química , Femenino , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/farmacocinética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Poliaminas/química , Polietilenglicoles/química , Distribución Tisular
18.
Mol Cancer Ther ; 16(3): 453-460, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27980101

RESUMEN

The majority of tumors contain regions of hypoxia, which cause marked phenotypic changes to resident cells. This altered gene expression often leads to increased resistance to anticancer treatments. Therefore, elimination of these resistant hypoxic cells is crucial to prevent disease recurrence. Herein, we describe the selective delivery of imaging and chemotherapeutic agents to cells expressing carbonic anhydrase IX (CA IX), a highly upregulated hypoxia receptor. These agents were conjugated to a potent divalent CA IX ligand through a hydrophilic PEG linker. These conjugates are shown to bind CA IX-expressing cells in a receptor-dependent manner in vitro with mid-nanomolar affinities and in vivo with good tumor selectivity. In a mouse xenograft tumor model using HT-29 cells, a cytotoxic tubulysin B conjugate completely inhibited tumor growth. Overall, the targeting of a hypoxia marker, such as CA IX, to selectively deliver imaging or chemotherapeutic agents may lead to better treatment options for solid, hypoxic tumors. In addition, the combination of standard chemotherapeutics that are most potent in normoxic dividing cells and drugs specifically designed to eliminate hypoxic nondividing cells may elicit a superior clinical outcome. Mol Cancer Ther; 16(3); 453-60. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Anhidrasa Carbónica IX/genética , Hipoxia/genética , Hipoxia/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Antineoplásicos/química , Anhidrasa Carbónica IX/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Hipoxia/tratamiento farmacológico , Ligandos , Masculino , Ratones , Estructura Molecular , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Polietilenglicoles/química , Radiofármacos/química , Rodaminas/química , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Bioconjug Chem ; 27(7): 1762-9, 2016 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-27362480

RESUMEN

As tumors grow, vasculature is often deficient or malformed, resulting in many localized areas of hypoxia. Cells located in these hypoxic regions exhibit an altered gene expression pattern that can significantly alter resistance to conventional anticancer treatments such as ionizing radiation and chemotherapeutic drugs. A priori knowledge of the level of hypoxia within a tumor may better guide clinical care. In an effort to create a hypoxia specific imaging agent, a ligand for the tissue hypoxia marker, carbonic anhydrase IX (CA IX), was synthesized and used as a targeting ligand to deliver an attached (99m)Tc-chelating agent. Binding of the resulting conjugates to hypoxic cancer cells was first characterized in vitro. Whole animal imaging and biodistribution studies then were performed to determine tumor specificity in vivo. Several conjugates were found to bind selectively to CA IX expressing tumors in a receptor-dependent manner. We suggest that such conjugates could prove useful in identifying hypoxic cancers and/or quantitating the level of hypoxia within a tumor.


Asunto(s)
Anhidrasa Carbónica IX/metabolismo , Inhibidores de Anhidrasa Carbónica/química , Regulación Neoplásica de la Expresión Génica , Tomografía Computarizada de Emisión de Fotón Único/métodos , Hipoxia Tumoral , Animales , Anhidrasa Carbónica IX/antagonistas & inhibidores , Inhibidores de Anhidrasa Carbónica/metabolismo , Inhibidores de Anhidrasa Carbónica/farmacocinética , Transformación Celular Neoplásica , Células HT29 , Humanos , Ligandos , Masculino , Ratones , Polietilenglicoles/química , Rodaminas/química , Tecnecio/química , Distribución Tisular
20.
Mol Pharm ; 13(5): 1618-25, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27043317

RESUMEN

Proof-of-principle studies in ovarian, lung, and brain cancer patients have shown that fluorescence-guided surgery can enable removal of otherwise undetectable malignant lesions, decrease the number of cancer-positive margins, and permit identification of disease-containing lymph nodes that would have normally evaded resection. Unfortunately, the current arsenal of tumor-targeted fluorescent dyes does not permit identification of all cancers, raising the need to design new tumor-specific fluorescent dyes to illuminate the currently undetectable cancers. In an effort to design a more universal fluorescent cancer imaging agent, we have undertaken to synthesize a fluorophore that could label all hypoxic regions of tumors. We report here the synthesis, in vitro binding, and in vivo imaging of a near-infrared (NIR) fluorescent dye that is targeted to carbonic anhydrase IX (CA IX), i.e., a widely accepted marker of hypoxic tissues. The low molecular weight NIR probe, named Hypoxyfluor, is shown to bind CA IX with high affinity and accumulate rapidly and selectively in CA IX positive tumors. Because nearly all human cancers contain hypoxic regions that express CA IX abundantly, this NIR probe should facilitate surgical resection of a wide variety of solid tumors.


Asunto(s)
Anhidrasa Carbónica IX/metabolismo , Hipoxia de la Célula/fisiología , Colorantes Fluorescentes/metabolismo , Neoplasias/metabolismo , Neoplasias/cirugía , Animales , Línea Celular Tumoral , Femenino , Fluorescencia , Células HT29 , Humanos , Ganglios Linfáticos/metabolismo , Ratones , Ratones Desnudos , Espectroscopía Infrarroja Corta/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA